1
|
Zhang J, Zeng F, Li Y, Mu C, Liu C, Wang L, Peng X, He L, Su Y, Li H, Wang A, Feng L, Gao D, Zhang Z, Xu G, Wang Y, Yue R, Si J, Zheng L, Zhang X, He F, Yi H, Tang Z, Li G, Ma K, Li Q. The characterization of technical design of a virus-like structure (VLS) nanodelivery system as vaccine candidate against SARS-CoV-2 variants. Hum Vaccin Immunother 2025; 21:2473183. [PMID: 40045463 PMCID: PMC11901403 DOI: 10.1080/21645515.2025.2473183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
The constant mutation of SARS-CoV-2 has led to the continuous appearance of viral variants and their pandemics and has improved the development of vaccines with a broad spectrum of antigens to curb the spread of the virus. The work described here suggested a novel vaccine with a virus-like structure (VLS) composed of combined mRNA and protein that is capable of stimulating the immune system in a manner similar to that of viral infection. This VLS vaccine is characterized by its ability to specifically target dendritic cells and/or macrophages through S1 protein recognition of the DC-SIGN receptor in cells, which leads to direct mRNA delivery to these innate immune cells for activation of robust immunity with a broad spectrum of neutralizing antibodies and immune protective capacity against variants. Research on its composition characteristics and structural features has suggested its druggability. Compared with the current mRNA vaccine, the VLS vaccine was identified as having no cytotoxicity at its effective application dosage, while the results of safety observations in animals revealed fewer adverse reactions during immunization.
Collapse
MESH Headings
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Animals
- SARS-CoV-2/immunology
- SARS-CoV-2/genetics
- COVID-19/prevention & control
- COVID-19/immunology
- Antibodies, Neutralizing/immunology
- Humans
- Antibodies, Viral/immunology
- Mice
- Dendritic Cells/immunology
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/genetics
- Lectins, C-Type/immunology
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/genetics
- mRNA Vaccines
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Macrophages/immunology
- Mice, Inbred BALB C
- Female
- Cell Adhesion Molecules
Collapse
Affiliation(s)
- Jingjing Zhang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
- Shandong Weigao Litong Biological Products Co, Ltd, Weihai, China
| | - Fengyuan Zeng
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Yanmei Li
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Changyong Mu
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Change Liu
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Lichun Wang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Xiaowu Peng
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Liping He
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Yanrui Su
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Hongbing Li
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - An Wang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Lin Feng
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Dongxiu Gao
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Zhixiao Zhang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Gang Xu
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Yixuan Wang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Rong Yue
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Junbo Si
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Lichun Zheng
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Xiong Zhang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Fuyun He
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Hongkun Yi
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Zhongshu Tang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Gaocan Li
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Kaili Ma
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
- Shandong Weigao Litong Biological Products Co, Ltd, Weihai, China
| | - Qihan Li
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| |
Collapse
|
2
|
Tica J, Rezelj VV, Baron B, van Paassen V, Zaidman J, Fairlie L, Scheper G, Le Gars M, Struyf F, Douoguih M, Ruiz-Guiñazú J. Safety and immunogenicity of Ad26.COV2.S in adolescents: Phase 2 randomized clinical trial. Hum Vaccin Immunother 2025; 21:2450120. [PMID: 39868766 PMCID: PMC11776467 DOI: 10.1080/21645515.2025.2450120] [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: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 01/28/2025] Open
Abstract
We conducted a randomized, Phase 2 trial to assess the safety and humoral immunogenicity of reduced doses/dose volume of the standard dose of Ad26.COV2.S COVID-19 vaccine (5 × 1010 viral particles [vp]) in healthy adolescents aged 12-17 years. Participants were randomly assigned to receive Ad26.COV2.S at reduced dose levels of 0.625 × 1010 (0.5 mL), 1.25 × 1010 (0.5 mL) or 2.5 × 1010 (0.5 mL or low volume 0.25 mL) vp in a 1- or 2-dose (56-day interval) primary schedule. Adolescents who received a 1-dose primary schedule received a 2.5 × 1010 vp booster dose 6 months later. Safety and humoral immunogenicity were assessed up to 6 months post-last vaccination. All regimens were well tolerated, with no safety concerns identified. Local and systemic solicited AEs in adolescents were consistent with the known safety profile in adults. All 1- and 2-dose Ad26.COV2.S primary schedules elicited robust peak Spike-binding antibody responses and virus neutralizing titers against the reference strain, in participants with and without preexisting SARS-CoV-2 immunity. Immune responses were durable for at least 6 months. Spike-binding antibody responses were comparable to those elicited in young adults aged 18-25 years who received a standard dose of Ad26.COV2.S in Phase 3 efficacy studies Reduced doses/dose volume of Ad26.COV2.S had an acceptable safety profile and elicited robust humoral immune responses in adolescents aged 12-17 years. All 1- and 2-dose schedules elicited Spike-binding antibody responses that were comparable to an adult population in whom efficacy has been demonstrated using a higher vaccine dose. (clinicaltrials.gov NCT05007080).
Collapse
Affiliation(s)
- Jelena Tica
- Scientific Affairs and Late Development, Janssen-Cilag GmbH, Neuss, Germany
| | - Veronica V. Rezelj
- Biomarkers, Viral Vaccines, Janssen Vaccines and Prevention, Leiden, The Netherlands
| | - Benoit Baron
- Biostatistics – Vaccines, Janssen Vaccines and Prevention, Leiden, The Netherlands
| | - Vitalija van Paassen
- Biostatistics – Vaccines, Janssen Vaccines and Prevention, Leiden, The Netherlands
| | | | - Lee Fairlie
- Maternal and Child Health, Wits RHI Shandukani, Johannesburg, South Africa
| | - Gert Scheper
- Vaccine Research, Janssen Vaccines and Prevention, Leiden, The Netherlands
| | - Mathieu Le Gars
- Biomarkers, Viral Vaccines, Janssen Vaccines and Prevention, Leiden, The Netherlands
| | - Frank Struyf
- Scientific Affairs and Late Development, Janssen Research and Development, Beerse, Belgium
| | - Macaya Douoguih
- Clinical Development and Medical Affairs, Janssen Vaccines and Prevention, Leiden, The Netherlands
| | | | | |
Collapse
|
3
|
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
|
4
|
de Morais Gomes V, Santos DM, Macedo-da-Silva J, Lazari LC, Machado RRG, Dos Santos AF, Araujo DB, Coutinho JVP, Arini GS, Angeli CB, de Souza EE, Marques RF, Boscardin SB, Wrenger C, Marinho CRF, Oliveira DBL, Durigon EL, Labriola L, Rosa-Fernandes L, Palmisano G. P.1 and P.2 SARS-CoV-2 Brazilian variants activate the unfolded protein response with a time and pathway specificity. J Proteomics 2025; 315:105397. [PMID: 39909104 DOI: 10.1016/j.jprot.2025.105397] [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: 08/15/2024] [Revised: 01/07/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
COVID-19 is a human respiratory syndrome caused by the infection of the SARS-CoV-2 virus that has a high rate of infection and mortality. Viruses modulate the host machinery by altering cellular mechanisms that favor their replication. One of the mechanisms that viruses exploit is the protein folding and processing of post-translational modifications that occur in the endoplasmic reticulum (ER). When ER function is impaired, there is an accumulation of misfolded proteins leading to endoplasmic reticulum stress (ER stress). To maintain homeostasis, cells trigger an adaptive signaling mechanism called the Unfolded Protein Response (UPR) which helps cells deal with stress, but under severe conditions, can activate the apoptotic cell death mechanism. This study elucidated an activation of a diversity of molecular mechanisms by Brazilian variants of SARS-CoV-2 by a time-resolved and large-scale characterization of SARS-CoV-2-infected cells proteomics and immunoblotting. Furthermore, it was shown that pharmacological UPR modulation could reduce viral release by counteracting the different viral activations of its cellular response. Analysis of human clinical specimens and disease outcomes focusing on ER stress reinforces the importance of UPR modulation as a host regulatory mechanism during viral infection and could point to novel therapeutic targets. SIGNIFICANCE: Since the emergence of SARS-CoV-2 and the consequent COVID-19 pandemic, the rapid emergence of variants of this new coronavirus has been a cause for concern since many of them have significantly higher rates of transmissibility and virulence, being called Variants of Concern (VOC). In this work, we studied the VOCs Gamma (P.1) and Zeta (P.2), also known as Brazilian variants. Constant evidence has reported that there are particularities related to each variant of SARS-CoV-2, with different rates of transmissibility, replication and modulation of host biological processes being observed, in addition to the mutations present in the variants. For this reason, this work focused on infections caused by the Brazilian variants of SARS-CoV-2 in different cell lines, in which we were able to observe that the infections caused by the variants induced endoplasmic reticulum stress in the infected cells and activated the UPR pathways, presenting specific modulations of each variant in this pathway. Furthermore, transcriptome analysis of patients revealed a correlation between ER-related genes and COVID-19 progression. Finally, we observed that the use of UPR modulators in host cells decreased viral release of all variants without affecting cell viability. The data presented in this work complement the observations of other studies that aim to understand the pathogenicity of SARS-CoV-2 VOCs and possible new therapeutic strategies, mainly targeting biological processes related to the endoplasmic reticulum.
Collapse
Affiliation(s)
| | - Deivid Martins Santos
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Janaina Macedo-da-Silva
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Lucas C Lazari
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | | | | | - Danielle Bastos Araujo
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil
| | | | - Gabriel Santos Arini
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Brazil
| | - Claudia B Angeli
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Edmarcia E de Souza
- Unit for Drug Discovery, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Rodolfo F Marques
- Laboratory of Antigen Targeting for Dendritic Cells, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Laboratory of Antigen Targeting for Dendritic Cells, Department of Parasitology, ICB, University of São Paulo, Brazil
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, ICB, University of São Paulo, Brazil
| | | | - Danielle B L Oliveira
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil
| | - Edison L Durigon
- Laboratory of Clinical and Molecular Virology, Department of Microbiology, ICB, University of São Paulo, Brazil; Scientific Platform Pasteur USP, Sao Paulo, Brazil
| | - Leticia Labriola
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Brazil
| | - Livia Rosa-Fernandes
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil; Laboratory of Experimental Immunoparasitology, Department of Parasitology, ICB, University of São Paulo, Brazil; Centre for Motor Neuron Disease Research, Faculty of Medicine, Health & Human Sciences, Macquarie Medical School, Sydney, Australia
| | - Giuseppe Palmisano
- GlycoProteomics Laboratory, Department of Parasitology, ICB, University of São Paulo, Brazil; School of Natural Sciences, Macquarie University, Sydney, Australia.
| |
Collapse
|
5
|
Hachimi A, El-Mansoury B, Merzouki M. Incidence, pathophysiology, risk factors, histopathology, and outcomes of COVID-19-induced acute kidney injury: A narrative review. Microb Pathog 2025; 202:107360. [PMID: 39894232 DOI: 10.1016/j.micpath.2025.107360] [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: 09/03/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has led to a significant burden on global healthcare systems. COVID-19-induced acute kidney injury (AKI) is among one of the complications, that has emerged as a critical and frequent condition in COVID-19 patients. This AKI among COVID-19 patients is associated with poor outcomes, and high mortality rates, especially in those with severe AKI or requiring renal replacement therapy. COVID-19-induced AKI represents a significant complication with complex pathophysiology and multifactorial risk factors. Indeed, several pathophysiological mechanisms, including direct viral invasion of renal cells, systemic inflammation, endothelial and thrombotic abnormalities as well as nephrotoxic drugs and rhabdomyolysis are believed to underlie this condition. Moreover, histopathological and immunohistopathological findings commonly observed in postmortem studies include acute tubular necrosis, glomerular injury, and the presence of viral particles within renal tissue and urine. Identified risk factors for developing AKI vary among studies, depending on regions, underlying conditions, and the severity of the disease. Moreover, histopathological and immunohistopathological findings commonly observed in postmortem studies include show acute tubular necrosis, glomerular injury, and viral particles within renal tissue and urine. While, identified risk factors for developing AKI vary among studies, according to regions, underlying conditions, and the gravity of the disease. This narrative review aims to synthesize current knowledge on the incidence, pathophysiology, risk factors, histopathology, and outcomes of AKI induced by COVID-19.
Collapse
Affiliation(s)
- Abdelhamid Hachimi
- Medical ICU, Mohammed VI(th) University Hospital of Marrakech, Marrakech, Morocco; Morpho-Science Research Laboratory, Faculty of Medicine and Pharmacy, Cadi Ayyad University, Marrakech, Morocco; Life Sciences Department, Bioengineering Laboratory, Faculty of Sciences and Technics, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Bilal El-Mansoury
- Nutritional Physiopathologies, Neuroscience and Toxicology Team, Laboratory of Anthropogenic, Biotechnology and Health, Faculty of Sciences, Chouaib Doukkali University, El Jadida, Morocco
| | - Mohamed Merzouki
- Life Sciences Department, Bioengineering Laboratory, Faculty of Sciences and Technics, Sultan Moulay Slimane University, Beni Mellal, Morocco.
| |
Collapse
|
6
|
Jiao JB, Kang Q, Cui SX, Cao JL, Lin T, Ma CJ, Xiao ZH, Du T, Wang N, Du XJ, Wang S. Target-driven functionalized DNA hydrogel capillary sensor for SARS-CoV-2 dual-mode detection. Talanta 2025; 285:127342. [PMID: 39644672 DOI: 10.1016/j.talanta.2024.127342] [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: 10/22/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Coronavirus disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused secondary pandemic, which still poses a serious threat to physical health and economic development. Herein, the target-driven functionalized DNA hydrogel capillary sensor based on cascade signal amplification and carbon coated cobalt manganese modified by prussian blue and platinum nanoparticles (MnCo@C-Pt-PB NPs) has been successfully developed for dual-mode detection of SARS-CoV-2. The cascade signal amplification triggered by target RNA causes the permeability of the DNA hydrogel loaded in the capillary to be destroyed, thereby releasing the embedded MnCo@C-Pt-PB NPs as signal molecules into 3,3',5,5'-tetramethylbenzidine/hydrogen peroxide (TMB/H2O2) solution under the driving of capillarity. The colorless TMB is then catalyzed to blue oxidation products (oxTMB) due to peroxidase-like activity of MnCo@C-Pt-PB NPs, and MnCo@C-Pt-PB NPs and oxTMB with photothermal properties synergistically increase the system temperature under near-infrared irradiation, which are recorded by portable devices to achieve dual-mode detection. Signals intensity are proportional to the logarithm of T-RNA concentration in a wide detection range (100 aM-100 pM), with a detection limit of 100 aM. Moreover, the reliability of the developed method in oropharyngeal swabs samples has also been validated. The signal conversion and amplification function of functionalized DNA hydrogel enhances the convenience, sensitivity and versatility of the developed method, which is promising to be applied in environmental safety, molecular diagnostic assays and disease prevention.
Collapse
Affiliation(s)
- Jing-Bo Jiao
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China; College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Qing Kang
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Shu-Xin Cui
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Jiang-Li Cao
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Tong Lin
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Chen-Jing Ma
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ze-Hui Xiao
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ting Du
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Nan Wang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, China.
| | - Xin-Jun Du
- State Key Laboratory of Food Nutrition and Safety, Engineering Research Center of Food Biotechnology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, 300457, China.
| | - Shuo Wang
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
7
|
Bhatt KJ, Schulder T, Rudenstine S, McNeal K, Ettman CK, Galea S. Understanding the Mental Health Impact of the COVID-19 Pandemic Among Individuals With Chronic Illness. Psychol Rep 2025; 128:596-616. [PMID: 36932930 PMCID: PMC10028451 DOI: 10.1177/00332941231164338] [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: 03/19/2023]
Abstract
People with chronic illnesses are vulnerable to stress and psychopathology during population-level disasters, as a subset of individuals with disabilities. We aimed to examine the relationships between chronic illness, cumulative and specific stressors, and probable depression, probable anxiety, and post-traumatic stress in an under-resourced urban population in New York City during the COVID-19 pandemic. Using cross-sectional survey data collected in April 2020, we utilized bivariate chi-square analyses and multivariable logistic regression models to estimate differences in and adjusted odds of stressor endorsement and diagnostic prevalence between people with and without chronic illness. We also assessed effect modification of the relationship between stressor exposure and psychopathology by chronic illness status. Compared to people without chronic illness, those who reported having a chronic illness experienced increased odds of probable depression, probable anxiety, and post-traumatic stress. They were also more likely to report high cumulative COVID-19-related stress exposure, death of someone close to them due to coronavirus or COVID-19, family problems, feeling alone, supply shortages, and financial problems. Chronic illness was found to be an effect modifier in the relationship between the death of someone close due to coronavirus or COVID-19 and probable depression and between household job loss and probable anxiety.
Collapse
Affiliation(s)
- Krish J Bhatt
- Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Talia Schulder
- City University of New York, The City College of New York, New York, NY, USA
| | - Sasha Rudenstine
- City University of New York, The City College of New York, New York, NY, USA
| | - Kat McNeal
- City University of New York, The City College of New York, New York, NY, USA
| | | | - Sandro Galea
- School of Public Health, Boston University, Boston, MA, USA
| |
Collapse
|
8
|
Hassan O, Elbhairy AA, Siam AM, Abdelwahab T, Hamad AA, Mahmoud OE, Nabeh OA. Evaluating the safety and efficacy of nirmatrelvir-ritonavir therapy in pregnant women with COVID-19: a systematic review and meta-analysis. Eur J Clin Pharmacol 2025; 81:495-506. [PMID: 39948217 DOI: 10.1007/s00228-025-03808-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/25/2025] [Indexed: 03/20/2025]
Abstract
PURPOSE Pregnant women are at heightened risk for severe COVID-19 outcomes. However, treatment options during pregnancy remain limited due to concerns over their safety and efficacy. METHODS This systematic review and meta-analysis assessed the safety and efficacy of nirmatrelvir-ritonavir in pregnant women diagnosed with mild-to-moderate COVID-19. The analysis focused on cases where the treatment was initiated within five days of symptom onset. A single-arm meta-analysis was performed to comprehensively evaluate outcomes across maternal, delivery, and neonatal domains. RESULTS In line with PRISMA guidelines, six studies involving a total of 427 pregnant patients were included in the analysis. Hospitalization was reported in 2% of patients (95% CI: 1%-5%), with low heterogeneity across studies (I2 = 21.9%). Drug discontinuation and new-onset gestational diabetes (NOGDM) had a pooled estimate of 0.7% (95% CI: 3% to 15%) and 4.0% (95% CI: 1% to 16%), respectively, with substantial heterogeneity (I2 = 64.7% and 66.5%), respectively. New-onset gestational hypertension (NOGHTN) had a pooled estimate of 4% (95% CI: 1% to 26%), with considerable heterogeneity (I2 = 78.81%). For neonatal outcomes, the pooled estimate for birth weight was 3186 g (95% CI: 3123-3248 g; I2 = 0%), and no maternal or neonatal deaths were reported across the included studies. CONCLUSION Nirmatrelvir-ritonavir appears safe and effective for mild-to-moderate COVID-19 in pregnant women, with low rates of hospitalization and adverse maternal outcomes. Larger, randomized studies are crucial to confirm these findings and ensure safety in diverse populations.
Collapse
Affiliation(s)
- Omar Hassan
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Aya Magdy Siam
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | | | | - Omnia Azmy Nabeh
- Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
9
|
Barrachina-Esteve O, Anguita A, Reverter A, Espinosa J, Lafuente C, Rubio-Roy M, Crosas M, Vila-Sala C, Acero C, Navarro M, Cánovas D, Ribera G, Jodar M, Estela J. Neurologic features in hospitalized patients with COVID-19: a prospective cohort in a catalan hospital. Neurol Sci 2025; 46:1477-1488. [PMID: 39951175 PMCID: PMC11920300 DOI: 10.1007/s10072-025-08031-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVES To study the prevalence and timing of neurological manifestations, including cognitive involvement, in patients hospitalized for Coronavirus disease 2019 (COVID-19). To analyze the pathogenic mechanisms and any association they have with disease severity. METHODS Longitudinal cohort study with prospective follow-up of patients who required hospitalization. Patients under 65 who had no pre-existing cognitive impairment and did not require an ICU stay were evaluated 3 and 12 months after discharge using a battery of neuropsychological tests. RESULTS Of 205 patients hospitalized for COVID-19, 153 (74.6%) presented with neurological manifestations. The most frequent were myalgia (32.7%), headache (31.7%), dysgeusia (29.2%), and anosmia (24.9%). Patients with more severe illness at the time of hospitalization presented fewer neurological manifestations. Of the 62 patients who underwent neuropsychological examination 3 months after discharge, 22.6% had impaired attention, 19.4% impaired working memory, 16.1% impaired learning and retrieval, 9.7% impaired executive functions, and 8.2% impaired processing speed. Patients with anosmia also presented with more headache (OR 5.45; p < 0.001) and greater risk of working memory impairment (OR 5.87; p 0.03). At follow-up 12 months after hospital discharge, 14.3% of patients still showed impaired attention, 2.4% impaired working memory, 2.5% impaired executive functions, and 2.5% impaired processing speed. DISCUSSION Neurological manifestations are common in patients hospitalized for COVID-19 regardless of severity. The high prevalence of anosmia and its association with headache and working memory impairment at 3 months, suggest potential direct or indirect damage to the prefrontal cortex via invasion of the olfactory bulb by COVID-19.
Collapse
Affiliation(s)
- Oriol Barrachina-Esteve
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain.
- Department of Neurology, Manacor Hospital, Manacor, Mallorca, Spain.
| | - A Anguita
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
- Department of Neurology, Sant Joan de Déu Hospital, Althaia Foundation, Manresa, Spain
| | - A Reverter
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - J Espinosa
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - C Lafuente
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - M Rubio-Roy
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - M Crosas
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - C Vila-Sala
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - C Acero
- Department of Ophthalmology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - M Navarro
- Department of Infectious Diseases, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - D Cánovas
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - G Ribera
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| | - M Jodar
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
- Department of Clinical and Health Psychology, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centre for Biomedical Research in the Mental Health Network (CIBERSAM), National Institute of Health Carlos III, Madrid, Spain
| | - J Estela
- Department of Neurology, Parc Taulí University Hospital, Parc Taulí Research and Innovation Institute Foundation (I3PT), Universitat Autònoma de Barcelona, Sabadell, Spain
| |
Collapse
|
10
|
Benlarbi M, Kenfack DD, Dionne K, Côté-Chenette M, Beaudoin-Bussières G, Bélanger É, Ding S, Goni OH, Ngoume YF, Tauzin A, Medjahed H, Ghedin E, Duerr R, Finzi A, Tongo M. Longitudinal humoral immunity against SARS-CoV-2 Spike following infection in individuals from Cameroon. Virology 2025; 605:110467. [PMID: 40037139 PMCID: PMC11937844 DOI: 10.1016/j.virol.2025.110467] [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: 10/30/2024] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
In May 2023 the World Health Organization (WHO) declared the end of COVID-19 as a public health emergency. Seroprevalence studies performed in African countries, such as Cameroon, depicted a much higher COVID-19 burden than reported by the WHO. To better understand humoral responses kinetics following infection, we enrolled 333 participants from Yaoundé, Cameroon between March 2020 and January 2022. We measured the levels of antibodies targeting the SARS-CoV-2 receptor-binding-domain (RBD) and the Spike glycoproteins of Delta, Omicron BA.1 and BA.4/5 and the common cold coronavirus HCoV-OC43. We also evaluated plasma capacity to neutralize authentic SARS-CoV-2 virus and to mediate Antibody-Dependent Cellular Cytotoxicity (ADCC). Most individuals mounted a strong antibody response against SARS-CoV-2 Spike. Plasma neutralization waned faster than anti-Spike binding and ADCC. We observed differences in humoral responses by age and circulating variants. Altogether, we show a global overview of antibody dynamics and functionality against SARS-CoV-2 in Cameroon.
Collapse
Affiliation(s)
- Mehdi Benlarbi
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Dell-Dylan Kenfack
- Center of Research for Emerging and Re-Emerging Diseases (CREMER), Institute of Medical Research and Study of Medicinal Plants (IMPM), Yaoundé, Cameroon
| | - Katrina Dionne
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Maxime Côté-Chenette
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Guillaume Beaudoin-Bussières
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Étienne Bélanger
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Shilei Ding
- Centre de Recherche du CHUM, Montréal, Québec, Canada
| | - Oumarou H Goni
- Center of Research for Emerging and Re-Emerging Diseases (CREMER), Institute of Medical Research and Study of Medicinal Plants (IMPM), Yaoundé, Cameroon
| | - Yannick F Ngoume
- Center of Research for Emerging and Re-Emerging Diseases (CREMER), Institute of Medical Research and Study of Medicinal Plants (IMPM), Yaoundé, Cameroon
| | - Alexandra Tauzin
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Halima Medjahed
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Elodie Ghedin
- Systems Genomics Section, Laboratory of Parasitic Diseases, NIAID, National Institutes of Health, Bethesda, MD, USA
| | - Ralf Duerr
- Vaccine Center, NYU Grossman School of Medicine, New York, USA; Department of Medicine, NYU Grossman School of Medicine, New York, USA; Department of Microbiology, NYU Grossman School of Medicine, New York, USA
| | - Andrés Finzi
- Centre de Recherche du CHUM, Montréal, Québec, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada.
| | - Marcel Tongo
- Center of Research for Emerging and Re-Emerging Diseases (CREMER), Institute of Medical Research and Study of Medicinal Plants (IMPM), Yaoundé, Cameroon; HIV Pathogenesis Program, The Doris Duke Medical Research Institute, University of KwaZulu Natal, Durban, South Africa.
| |
Collapse
|
11
|
Gasnier M, Pinson P, Beeker N, Truong-Allié C, Becquemont L, Falissard B, Corruble E, Colle R. Acute COVID-19 severity markers predict post-COVID new-onset psychiatric disorders: A 2-year cohort study of 34,489 patients. Mol Psychiatry 2025; 30:1329-1337. [PMID: 39284906 DOI: 10.1038/s41380-024-02739-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 03/20/2025]
Abstract
New-onset psychiatric disorders are frequent after COVID-19. We aim to determine whether acute COVID-19 severity markers can predict post-COVID new-onset psychiatric disorders. We conducted an electronic health records (EHR) cohort study of patients hospitalized for COVID-19 and without any known history of psychiatric disorders. Patients were included between January 2020 and September 2022 in one of the 36 university hospitals of the Assistance Publique - Hôpitaux de Paris. Acute COVID-19 clinical and biological severity markers were recorded during hospitalization for COVID-19. Psychiatric ICD-10 diagnoses were recorded up to 2 years and 9 months after hospitalization for COVID-19. Predictors of post-COVID new-onset psychiatric disorders were identified based on Cox regression models and sensitivity analyses. Predictive scores were built and tested in age- and sex-stratified populations. A total 34,489 patients hospitalized for COVID-19 were included; 3717 patients (10.8%) had at least one post-COVID new-onset psychiatric disorder. Hospital stay >7 days (HR = 1.72, 95%CI [1.59-1.86], p < 0.001), acute delirium (HR = 1.49, 95%CI [1.28-1.74], p < 0.001), elevated monocyte count (HR = 1.14, 95%CI [1.06-1.23], p < 0.001) and elevated plasma CRP (HR = 0.92, 95%CI [0.86-0.99], p = 0.04) independently predicted post-COVID new-onset psychiatric disorders. Sensitivity analyses confirmed hospital stay >7 days, acute delirium, and elevated monocyte count as predictors. Predictive scores based on these variables had good 12-month positive predictive values, up to 7.5 times more accurate than random in women < 65 years. In conclusion, hospital stay >7 days, acute delirium, and elevated monocyte count during acute COVID-19 predict post-COVID new-onset psychiatric disorders.
Collapse
Affiliation(s)
- Matthieu Gasnier
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Mood Center Paris Saclay, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Paris, Le Kremlin Bicêtre, France
- MOODS Team, INSERM 1018, CESP (Centre de Recherche en Epidémiologie et Santé des Populations), Université Paris-Saclay, Faculté de Médecine Paris-Saclay, Paris, Le Kremlin Bicêtre, France
| | - Pierre Pinson
- Unité de Recherche clinique, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Nathanael Beeker
- Unité de Recherche clinique, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Camille Truong-Allié
- Unité de Recherche clinique, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Laurent Becquemont
- MOODS Team, INSERM 1018, CESP (Centre de Recherche en Epidémiologie et Santé des Populations), Université Paris-Saclay, Faculté de Médecine Paris-Saclay, Paris, Le Kremlin Bicêtre, France
- Université Paris-Saclay, AP-HP, Centre de Recherche Clinique, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Paris, Le Kremlin Bicêtre, France
| | - Bruno Falissard
- CESP (Centre de Recherche en Epidémiologie et Santé des Populations), Paris, France
| | - Emmanuelle Corruble
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Mood Center Paris Saclay, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Paris, Le Kremlin Bicêtre, France.
- MOODS Team, INSERM 1018, CESP (Centre de Recherche en Epidémiologie et Santé des Populations), Université Paris-Saclay, Faculté de Médecine Paris-Saclay, Paris, Le Kremlin Bicêtre, France.
| | - Romain Colle
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Mood Center Paris Saclay, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Paris, Le Kremlin Bicêtre, France
- MOODS Team, INSERM 1018, CESP (Centre de Recherche en Epidémiologie et Santé des Populations), Université Paris-Saclay, Faculté de Médecine Paris-Saclay, Paris, Le Kremlin Bicêtre, France
| |
Collapse
|
12
|
Konitsioti AM, Laurent S, Ellenberger D, Stahmann A, Rommer P, Haas J, Warnke C. MS treatment trends before, during, and after the COVID-19 pandemic: insights from the German MS Register. J Neurol 2025; 272:294. [PMID: 40137994 DOI: 10.1007/s00415-025-13010-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/16/2025] [Accepted: 03/01/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND The COVID-19 pandemic affected healthcare management for people with multiple sclerosis (PwMS), leading to alterations in disease-modifying therapies (DMTs) due to concerns about COVID-19 outcomes and vaccine efficacy. OBJECTIVES To compare DMT prescription patterns in PwMS before, during, and after the COVID-19 pandemic. METHODS PwMS from the German MS Register, between 2019 and 2024, either newly diagnosed (Cohort A) or who discontinued or switched DMT (Cohort B), were analyzed over a follow-up period of 3 months. Data from the pre-pandemic period were compared to early-, late-, and post-pandemic periods. DMTs were categorized as medium efficacy (meDMT) or high efficacy (heDMT). RESULTS In Cohort A (n = 1810), pre-pandemic 46% had no DMT within 3 months of diagnosis, 39% received meDMT, and 15% heDMT (7.5% B cell-depleting therapies (BCD)). heDMT use increased during later periods ("early" 19%, "late" 29%, "post" 41%), with a shift toward BCD. In cohort B (n = 4246), pre-pandemic 47% paused DMT, 19% switched to meDMT, and 34% to heDMT (17% BCD). heDMT use also rose during the pandemic ("early" 37%, "late" 47%, "post" 48%), with increased BCD use. CONCLUSIONS There were no delays in DMT initiation or resumption during the pandemic with a notable increase in heDMT and BCD use, reflecting growing confidence in these treatment options.
Collapse
Affiliation(s)
- Agni-Maria Konitsioti
- Department of Neurology, Medical Faculty, University Hospital of Cologne, Kerpener Str. 62, Cologne, Germany.
| | - Sarah Laurent
- Neurology, CHU Saint-Pierre, Université Libre de Bruxelles, Brussels, Belgium
| | - David Ellenberger
- German MS-Register, MS Forschungs-und Projektentwicklungs gGmbH (MS Research and Project Development gGmbH [MSFP]), Hannover, Germany
| | - Alexander Stahmann
- German MS-Register, MS Forschungs-und Projektentwicklungs gGmbH (MS Research and Project Development gGmbH [MSFP]), Hannover, Germany
| | - Paulus Rommer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
- Neuroimmunological Section, Department of Neurology, University of Rostock, Rostock, Germany
| | - Judith Haas
- Deutsche Multiple Sklerose Gesellschaft, Bundesverband e.V. (German Multiple Sclerosis Society [DMSG]), Hannover, Germany
| | - Clemens Warnke
- Department of Neurology, University Hospital Marburg, Marburg, Germany
| |
Collapse
|
13
|
Ham SY, Lee S, Kim MK, Jeon J, Lee E, Kim S, Choi JP, Jang HC, Park SW. Incidence and Temporal Dynamics of Combined Infections in SARS-CoV-2-Infected Patients With Risk Factors for Severe Complications. J Korean Med Sci 2025; 40:e38. [PMID: 40132535 DOI: 10.3346/jkms.2025.40.e38] [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: 06/25/2024] [Accepted: 10/17/2024] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is a newly emerged infectious disease that needs further clinical investigation. Characterizing the temporal pattern of combined infections in patients with COVID-19 may help clinicians understand the clinical nature of this disease and provide valuable diagnostic and therapeutic guidelines. METHODS We retrospectively analyzed COVID-19 patients isolated in four study hospitals in Korea for one year period from May 2021 to April 2022 when the delta and omicron variants were dominant. The temporal characteristics of combined infections based on specific diagnostic tests were analyzed. RESULTS A total of 16,967 COVID-19 patients were screened, 2,432 (14.3%) of whom underwent diagnostic microbiologic tests according to the clinical decision-making, 195 of whom had positive test results, and 0.55% (94/16,967) of whom were ultimately considered to have clinically meaningful combined infections. The median duration for the diagnosis of combined infections was 15 (interquartile range [IQR], 5-25) days after admission. The proportion of community-acquired coinfections (≤ 2 days after admission) was 11.7% (11/94), which included bacteremia (10/94, 10.63%) and tuberculosis (1/94, 1.06%). Combined infections after 2 days of admission were diagnosed at median 16 (IQR, 9-26) days, and included bacteremia (72.3%), fungemia (19.3%), cytomegalovirus (CMV) diseases (8.4%), Pneumocystis jerovecii pneumonia (PJP, 8.4%) and invasive pulmonary aspergillosis (IPA, 4.8%). CONCLUSION Among COVID-19 patients with risk factors for severe complications, 0.55% had laboratory-confirmed combined infections, which included community and nosocomial pathogens in addition to unusual pathogens such as CMV disease, PJP and IPA.
Collapse
Affiliation(s)
- Sin Young Ham
- Seoul Veterans Hospital Medical Center, Seoul, Korea
| | - Seungjae Lee
- Seoul Veterans Hospital Medical Center, Seoul, Korea
| | - Min-Kyung Kim
- Division of Infectious Diseases, Department of Internal Medicine, National Medical Center, Seoul, Korea
| | - Jaehyun Jeon
- Seoul Veterans Hospital Medical Center, Seoul, Korea
- Division of Infectious Diseases, Department of Internal Medicine, National Medical Center, Seoul, Korea
| | - Eunyoung Lee
- Department of Internal Medicine, Seoul National University College of Medicine and Boramae Medical Center, Seoul, Korea
| | - Subin Kim
- Division of Infectious Diseases, Seoul Medical Center, Seoul, Korea
| | - Jae-Phil Choi
- Division of Infectious Diseases, Seoul Medical Center, Seoul, Korea
| | - Hee-Chang Jang
- National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Sang-Won Park
- Department of Internal Medicine, Seoul National University College of Medicine and Boramae Medical Center, Seoul, Korea.
| |
Collapse
|
14
|
Li H, Xing J, Song Z, Fan Z, Wen H, Liang S, Yan Q, Feng H, Han S, Yang N, Wang P, Zhang K. Effect of mild-to-moderate COVID‑19 on the incidence and risk factors for deep vein thrombosis in patients with hip fracture: a retrospective study. BMC Surg 2025; 25:113. [PMID: 40128691 PMCID: PMC11931773 DOI: 10.1186/s12893-025-02831-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/06/2025] [Indexed: 03/26/2025] Open
Abstract
PURPOSE This retrospective study aimed to investigate the effect of mild-to-moderate COVID-19 on the risk of deep vein thrombosis (DVT) in patients with hip fractures. Hip fractures are common in the elderly, and previous research has shown that they accounted for 58.3% of traumatic fractures in older inpatients during the COVID-19 pandemic in China. Meanwhile, the relationship between COVID-19 and DVT is complex. Some studies have reported that the incidence of DVT in critically ill COVID-19 patients can be as high as 46%, and 20% in those with moderate-to-severe cases. However, the impact of mild-to-moderate COVID-19 on DVT risk in hip fracture patients remains unclear. METHODS Adult patients who underwent surgery for hip fractures between December 8, 2022, and January 9, 2023, were included in the study. All patients were tested for SARS-CoV-2 nucleic acid and were assessed for DVT preoperatively using doppler ultrasonography (DUS). Logistic regression was used to identify risk factors for DVT. RESULTS The records of 98 patients with hip fractures, were included in the analysis, of whom 63 were SARS-CoV-2 positive and 35 were SARS-CoV-2 negative. Pre-operative DUS showed that 36/98 patients (37%) had DVT, including 25/63 (40%) patients with COVID-19, and 11/35 (31%) patients without COVID-19. Multivariable logistic regression analysis showed that pre-operative leukocyte count and platelet-to-lymphocyte ratio (PLR) were independent risk factors for DVT, whereas mild-to-moderate COVID-19 was not an independent risk factor for DVT. In patients with hip fractures, COVID-19 did not significantly increase the risk of DVT. CONCLUSIONS Therefore, in patients with hip fractures, DVT prevention measures should be implemented routinely, regardless of COVID-19 status.
Collapse
Affiliation(s)
- Haoran Li
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jian Xing
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zhe Song
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Zhiqiang Fan
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Hongquan Wen
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Shaobo Liang
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qiang Yan
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Haoxuan Feng
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Shuang Han
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Na Yang
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Pengfei Wang
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.
| | - Kun Zhang
- Orthopedic Trauma Department, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
15
|
He Y, Zheng Q, Zhifang Z, Xiaofeng N, Shenggen W, Xue M, Zheng C, Liu Z. When COVID-19 meets diabetes: A bibliometric analysis. Diabetes Res Clin Pract 2025; 223:112118. [PMID: 40132732 DOI: 10.1016/j.diabres.2025.112118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Coronavirus disease 2019 (COVID-19) survivors are concerned about the likelihood of developing further diseases. This study examines the global trends in scientific research on diabetes associated with COVID-19 from several perspectives. Bibliometric analyses are used to undertake a scientific review of the literature. The Web of Science Core Collection (WoSCC) database was used to acquire bibliographic information on diabetes related to COVID-19 from Jan 2020 to Dec. 2023. The visual map was built via advanced CiteSpace 6.2.R6. 7,348 papers were found. Khunti Kamlesh and Rizzo-Manfredi are the most well-known high-yield authors in this area, and the top ten authors collaborate extensively. Most of these papers came from universities. Harvard Medical School has the most publications, followed by Wuhan University and Huazhong University of Science and Technology. China and the United States are the countries with the most publications. Angiotensin-converting enzymes, chronic disease, intensive care unit, viral infection, and gestational diabetes mellitus were scored 0-11, 2, 3, and 4, respectively. Zhou et al.'s work on this topic, which appeared in the prominent medical journal The Lancet, was cited 1,366 times, highlighting its importance. "clinical characteristics," "diabetes mellitus," "metabolic syndrome," and "angiotensin-converting enzyme" were used as keywords for reference co-citation and clustering data identify. Over the last four years, related investigations have focused primarily on observing clinical aspects. This report is important for developing treatment strategies, directing future research, and guiding clinical practice.
Collapse
Affiliation(s)
- Yingli He
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Qingcong Zheng
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhang Zhifang
- Fujian Center for Disease Control and Prevention, Fuzhou 350012, China
| | | | - Wu Shenggen
- Fujian Center for Disease Control and Prevention, Fuzhou 350012, China
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada.
| | - Zhijun Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Xi'an Jiaotong University, China.
| |
Collapse
|
16
|
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] [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
|
17
|
Yildiz Gulhan P, Eroz R, Ozturk CE, Yekenkurul D, Altinsoy HB, Gulec Balbay E, Ercelik M, Davran F, Yildiz S. Determination of both the expression and serum levels of epidermal growth factor and transforming growth factor β1 genes in COVID-19. Sci Rep 2025; 15:9771. [PMID: 40118922 PMCID: PMC11928509 DOI: 10.1038/s41598-025-92304-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/26/2025] [Indexed: 03/24/2025] Open
Abstract
We aimed to evaluate the effects of both the expression and serum levels of Epidermal growth factor (EGF) and Transforming growth factor-β1 (TGF-β1) genes in patients with different degrees of cellular damage as mild, moderate, severe, and critical illness that can lead to fibrosis caused by SARS-CoV-2. Totally 45 individuals (male: 21(46.67%); female: 24(53.33%)) with COVID-19 infection were included in this study. Four groups were constituted as mild (n = 16)], moderate (n = 10), severe (n = 10), and critical (n = 9) according to the severity of the disease. Blood samples were drawn from the patients, and all of the hemograms, EGF and TGFβ1 gene expression, and serum levels were evaluated. The mean age of individuals was 57.311 ± 18.383 (min: 28, max: 94). Significant differences were found among the groups for PLT (χ2 = 9.955; p = 0.019), CRP (χ2 = 7.693; p = 0.053), Ferritin (χ2 = 22.196; p < 0.001), D-dimer (χ2 = 21.982; p = 0.000), LDH (χ2 = 21.807; p < 0.001) and all these parameters (exclude PLT in severe groups) was increased depending on the severity of the disease. Additionally, significant differences were detected for EGF (χ2 = 29.528; p < 0.001), TGFB1 (χ2 = 28.981; p < 0.001) expression (that increased depending on the disease severity), and EGF (χ2 = 7.84; p = 0.049), TGFB1 (χ2 = 17.451; p = 0.001) serum concentration levels (that decreased depending on the disease severity). This study found statistically significant differences for both EGF 2-ΔΔCt. TGFβ1 2-ΔΔCt and EGF, TGFβ1 serum concentration values among all patient groups. As disease severity increased, EGF 2-ΔΔCt. TGFβ1 2-ΔΔCt levels increased, while EGF and TGFβ1 serum concentration levels decreased. Perhaps this study will be useful in managing COVID-19 infection severity and pulmonary fibrosis cases secondary to COVID-19.
Collapse
Affiliation(s)
- Pinar Yildiz Gulhan
- Department of Chest Diseases, Faculty of Medicine, Duzce University, Konuralp Campus, 81010, Duzce, Turkey.
| | - Recep Eroz
- Department of Medical Genetics, Aksaray University Medical Faculty, Aksaray, Turkey
| | | | - Dilek Yekenkurul
- Department of Infection Diseases, Duzce University Medical Faculty, Duzce, Turkey
| | | | - Ege Gulec Balbay
- Department of Chest Diseases, Faculty of Medicine, Duzce University, Konuralp Campus, 81010, Duzce, Turkey
| | - Merve Ercelik
- Department of Chest Diseases, Faculty of Medicine, Duzce University, Konuralp Campus, 81010, Duzce, Turkey
| | - Fatih Davran
- Department of Biochemistry, Faculty of Medicine, Duzce University, Duzce, Turkey
| | - Seyma Yildiz
- Deparment of Hematology, Gazi University, Ankara, Turkey
| |
Collapse
|
18
|
Azcarate D, Olasagasti Arsuaga F, Granizo Rodriguez E, Arana-Arri E, España PP, Intxausti M, Sancho C, García de Vicuña Meléndez A, Ibarrondo O, M de Pancorbo M. Human-genetic variants associated with susceptibility to SARS-CoV-2 infection. Gene 2025; 953:149423. [PMID: 40120867 DOI: 10.1016/j.gene.2025.149423] [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: 11/19/2024] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
SARS-CoV-2, the third major coronavirus of the 21st century, causing COVID-19 disease, profoundly impacts public health and workforces worldwide. Identifying individuals at heightened risk of SARS-CoV-2 infection is crucial for targeted interventions and preparedness. This study investigated 35 SNVs within viral infection-associated genes in SARS-CoV-2 patients and uninfected controls from the Basque Country (March 2020-July 2021). Its primary aim was to uncover genetic markers indicative of SARS-CoV-2 susceptibility and explore genetic predispositions to infection. Association analyses revealed previously unreported associations between SNVs and susceptibility. Haplotype analyses uncovered novel links between haplotypes and susceptibility, surpassing individual SNV associations. Descriptive modelling identified key susceptibility factors, with rs11246068-CC (IFITM3), rs5742933-GG (ORMDL1), rs35337543-CG (IFIH1), and GGGCT (rs2070788, rs2298659, rs17854725, rs12329760, rs3787950) variation in TMPRSS2 emerging as main infection-susceptibility indicators for a COVID-19 pandemic situation. These findings underscore the importance of integrated SNV and haplotype analyses in delineating susceptibility to SARS-CoV-2 and informing proactive prevention strategies. The genetic markers profiled in this study offer valuable insights for future pandemic preparedness.
Collapse
Affiliation(s)
- Daniel Azcarate
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Zoology and animal cellular biology department, Faculty of Science and Technology (UPV/EHU), 48940 Leioa, Biscay (Basque Country), Spain
| | - Felix Olasagasti Arsuaga
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Biochemistry and molecular biology department, Faculty of Pharmacy (UPV/EHU), 01006 Vitoria-Gasteiz, Alava (Basque Country), Spain.
| | - Eva Granizo Rodriguez
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Zoology and animal cellular biology department, Faculty of Science and Technology (UPV/EHU), 48940 Leioa, Biscay (Basque Country), Spain
| | - Eunate Arana-Arri
- Clinical Epidemiology Unit, Cruces University Hospital, 48903 Barakaldo, Biscay (Basque Country), Spain
| | - Pedro Pablo España
- Pulmonology Service, Galdakao-Usansolo University Hospital, 48960 Galdakao, Biscay (Basque Country), Spain
| | - Maider Intxausti
- Pulmonology Service, Alava University Hospital - Txagorritxu, 01009 Vitoria-Gasteiz, Álava (Basque Country), Spain
| | - Cristina Sancho
- Department of Pneumology, Basurto University Hospital, 48013 Bilbao, Biscay (Basque Country), Spain
| | | | - Oliver Ibarrondo
- Consultant in Statistics and Health Economics Research, Debagoiena AP-OSI Research Unit, 20500 Arrasate, Gipuzkoa (Basque Country), Spain
| | - Marian M de Pancorbo
- BIOMICs Research Group (BIOMICS and Microfluidics cluster), Zoology and animal cellular biology department, Faculty of Science and Technology (UPV/EHU), 48940 Leioa, Biscay (Basque Country), Spain.
| |
Collapse
|
19
|
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] [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
|
20
|
Kang MA, Lee SK. Exploring Coronavirus Disease 2019 Risk Factors: A Text Network Analysis Approach. J Clin Med 2025; 14:2084. [PMID: 40142892 DOI: 10.3390/jcm14062084] [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: 02/15/2025] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: The coronavirus disease 2019 (COVID-19) pandemic has significantly affected global health, economies, and societies, necessitating a deeper understanding of the factors influencing its spread and severity. Methods: This study employed text network analysis to examine relationships among various risk factors associated with severe COVID-19. Analyzing a dataset of published studies from January 2020 to December 2021, this study identifies key determinants, including age, hypertension, and pre-existing health conditions, while uncovering their interconnections. Results: The analysis reveals five thematic clusters: biomedical, occupational, demographic, behavioral, and complication-related factors. Temporal trend analysis reveals distinct shifts in research focus over time. In early 2020, studies primarily addressed immediate clinical characteristics and acute complications of COVID-19. By mid-2021, research increasingly emphasized long COVID, highlighting its prolonged symptoms and impact on quality of life. Concurrently, vaccine efficacy became a dominant topic, with studies assessing protection rates against emerging viral variants, such as Alpha, Delta, and Omicron. This evolving landscape underscores the dynamic nature of COVID-19 research and the adaptation of public health strategies accordingly. Conclusions: These findings offer valuable insights for targeted public health interventions, emphasizing the need for tailored strategies to mitigate severe outcomes in high-risk groups. This study demonstrates the potential of text network analysis as a robust tool for synthesizing complex datasets and informing evidence-based decision-making in pandemic preparedness and response.
Collapse
Affiliation(s)
- Min-Ah Kang
- Department of Nursing, Keimyung College University, Daegu 42601, Republic of Korea
| | - Soo-Kyoung Lee
- Department of Medical Informatics, College of Nursing & Health, Kongju National University, Kongju 32588, Republic of Korea
| |
Collapse
|
21
|
Liu Z, Yuan R, Wang S, Liao W, Yan L, Hu R, Chen J, Yu L. Skin-Inspired Self-Aligned Silicon Nanowire Thermoreceptors for Rapid and Continuous Temperature Monitoring. NANO LETTERS 2025; 25:4196-4203. [PMID: 40062983 DOI: 10.1021/acs.nanolett.4c05235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Real-time and precise evaluation of human body temperature offers crucial insights for health monitoring and disease diagnosis, while integration of high-performance and miniaturized sensors remains a challenge. Inspired by the thermal sensory pathway of skin, here we developed a new route for scalable fabrication of rapid-response and miniaturized thermoreceptor sensors using self-aligned in-plane silicon nanowire (SiNW) arrays as sensitive channels. These SiNW arrays, with a diameter of 100 ± 14 nm, were integrated into temperature sensors with a density of 445 devices/cm2 without using any high-precision lithography. The sensors exhibited an excellent temperature coefficient of resistance of -1.8%/°C, enabling the precise spatial identification of heat sources. They achieved real-time monitoring of temperature changes during breathing and blowing activities, with a rapid response time of ∼0.2 s and recovery time of ∼1 s. This study provides a robust foundation for the integration of advanced miniaturized temperature sensors for biological monitoring applications.
Collapse
Affiliation(s)
- Zongguang Liu
- Microelectronics Industry Research Institute, College of Physics Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Rongrong Yuan
- School of Electronic Science and Engineering/National Laboratory of Solid-State Microstructures, Nanjing University, Nanjing, 210023, P. R. China
| | - Shuyi Wang
- School of Electronic Science and Engineering/National Laboratory of Solid-State Microstructures, Nanjing University, Nanjing, 210023, P. R. China
| | - Wei Liao
- School of Electronic Science and Engineering/National Laboratory of Solid-State Microstructures, Nanjing University, Nanjing, 210023, P. R. China
| | - Lei Yan
- School of Electronic Science and Engineering/National Laboratory of Solid-State Microstructures, Nanjing University, Nanjing, 210023, P. R. China
| | - Ruijin Hu
- Microelectronics Industry Research Institute, College of Physics Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Jianmei Chen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Linwei Yu
- School of Electronic Science and Engineering/National Laboratory of Solid-State Microstructures, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
22
|
Monteiro AHA, Freitas KM, Montuori-Andrade ACM, de Lima EBS, Carvalho AFS, Cardoso C, Lara ES, Oliveira LC, Zaidan I, da Santos FRS, Resende F, Souza-Costa LP, Queiroz-Junior CM, Chaves IDM, Nóbrega NRC, Rabelo MBO, Rocha MP, Campana PRV, Pádua RM, Ferreira RS, Barreto LV, Kronenberger T, Maltarollo VG, de Godoy MO, Oliva G, Guido RVC, Teixeira MM, Costa VV, Sousa LP, Braga FC. Ouratein D, a Biflavanone From Ouratea spectabilis, Alleviates Betacoronavirus Infection by Mitigating Inflammation, Lung Damage and Viral Replication. Phytother Res 2025. [PMID: 40099709 DOI: 10.1002/ptr.8462] [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: 08/19/2024] [Revised: 02/03/2025] [Accepted: 02/08/2025] [Indexed: 03/20/2025]
Abstract
Severe coronavirus outbreaks, including SARS, MERS, and COVID-19, have underscored the urgent need for effective antiviral therapies. This study evaluated the antiviral activity of biflavanones isolated from Ouratea spectabilis-specifically ouratein (Our-) A, B, C, and D-against murine hepatitis virus (MHV-3) and human SARS-CoV-2. Cells infected with MHV-3 or SARS-CoV-2 were treated with ourateins, and viral replication was assessed using plaque assays. Mice infected with MHV-3 were treated with Our-D either orally or intraperitoneally. Key assessments included leukocyte counts, cytokine and chemokine levels, histological analysis, and survival rates. The mechanism of action was explored through in silico and in vitro studies focused on the binding and inhibition of the main protease (Mpro). Our-D significantly inhibited the replication of both viruses, with a selective index of 2.5 for MHV-3 and 14.9 for SARS-CoV-2. In vivo, Our-D reduced leukocyte infiltration in the lungs, decreased CCL2 levels, increased IL-10, and lowered plasma IL-6 and CXCL1 levels. Additionally, Our-D mitigated lung damage, partially restored betacoronavirus-induced lymphopenia, and reduced viral loads in the lungs, heart, and spleen, ultimately improving survival in mice. In silico studies revealed that Our-A and Our-C had strong binding affinity for Mpro, and both significantly inhibited Mpro activity in vitro, unlike Our-D. Our-D protected mice from coronavirus infection by modulating the inflammatory response and reducing viral loads, with minimal effect on Mpro inhibition, suggesting alternative mechanisms for its antiviral activity.
Collapse
Affiliation(s)
- Adelson Héric A Monteiro
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Kátia M Freitas
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Clara M Montuori-Andrade
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Erick Bryan Sousa de Lima
- Department of Clinical Analysis and Toxicology, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Antônio Felipe S Carvalho
- Department of Clinical Analysis and Toxicology, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Camila Cardoso
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Edvaldo S Lara
- Department of Clinical Analysis and Toxicology, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo Camilo Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabella Zaidan
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Felipe Rocha Silva da Santos
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Filipe Resende
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiz Pedro Souza-Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso M Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ian de Meira Chaves
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Natália R C Nóbrega
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria Beatriz O Rabelo
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marina P Rocha
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Priscilla R V Campana
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rodrigo M Pádua
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rafaela S Ferreira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiza V Barreto
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thales Kronenberger
- Partner-Site Tübingen, German Center for Infection Research (DZIF), Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Vinícius G Maltarollo
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Glaucius Oliva
- Institute of Physics, Universidade de São Paulo, São Carlos, Brazil
| | - Rafael V C Guido
- Institute of Physics, Universidade de São Paulo, São Carlos, Brazil
| | - Mauro M Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian V Costa
- Department of Morphology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Department of Clinical Analysis and Toxicology, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernão C Braga
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
23
|
Liao R, Luo D, Yang D, Liu J. Opportunities and Challenges of DNA Materials toward Sustainable Development Goals. ACS NANO 2025. [PMID: 40099911 DOI: 10.1021/acsnano.4c17718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Sustainable development represents a significant and pressing challenge confronting the global community at present. A wide variety of macroscopic engineering systems has been developed to promote sustainable development. Recent advancements in DNA materials have showcased their substantial contributions toward achieving sustainable development goals (SDGs). Compared to nonbiological materials, DNA materials possess exceptional properties such as genetic functionality, molecular programmability, recognition capabilities, and biocompatibility. These unique characteristics enable DNA materials to serve as general and versatile substrates beyond their genetic role. Consequently, they can be used to develop DNA-based engineering systems that offer versatile solutions to support sustainable development. In this Perspective, we critically examine the opportunities that DNA-based engineering systems present in contributing to the achievement of the SDGs within various real-world scenarios. We establish direct relationships between DNA-based engineering systems and the SDGs, highlighting their inherent merits in accelerating sustainable development. Furthermore, in order to successfully achieve SDGs, we address the challenges associated with these systems and emphasize the urgent need for developing multifunctional, reliable, biosafe, and intelligent DNA-based engineering systems to overcome these challenges.
Collapse
Affiliation(s)
- Renkuan Liao
- College of Land Science and Technology, Key Laboratory of Arable Land Conservation in North China, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing 100193, People's Republic of China
- State Key Laboratory of Efficient Utilization of Agricultural Water Resources, China Agricultural University, Beijing 100083, People's Republic of China
| | - Dan Luo
- Department of Biological & Environmental Engineering, Cornell University, Ithaca, New York 14853, United States
| | - Dayong Yang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, College of Chemistry and Materials, Fudan University, Shanghai 200438, People's Republic of China
| | - Jianguo Liu
- Center for Systems Integration and Sustainability, Department of Fisheries and Wildlife, Michigan State University, East Lansing, Michigan 48823, United States
| |
Collapse
|
24
|
Nandula SR, Brichacek B, Sen S. Podocyte-Specific Protein Expression in Urine Exosome Acts as a Marker for Renal Injury in Post-COVID State. Metab Syndr Relat Disord 2025. [PMID: 40100769 DOI: 10.1089/met.2024.0199] [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: 03/20/2025] Open
Abstract
Introduction: Severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2) has been associated with the development of COVID-19. COVID-19 may cause endothelial cell dysfunction (ECD), which can lead to cardiometabolic diseases and podocytopathy. In this study, we explored whether presence of hyperglycemia predisposes to SARS-CoV-2 infection, in vitro, and whether COVID-19 can put an individual at a higher risk of persistent renal damage in the long-term following acute COVID infection. To estimate renal damage, we evaluated albuminuria and podocytopathy. Podocytopathy was estimated by measuring podocyte-specific protein levels in urine-derived exosomes from patients who were admitted with acute COVID-19 at 10 days, 6 months, and 12 months post-acute SARS-CoV-2 infection. Methods: Blood and urine samples from patients with SARS-CoV-2 post-infection were procured from the George Washington University COVID repository. Peripheral blood mononuclear cells and urine exosomes were isolated. Podocyte-specific proteins Podocalyxin (PODXL) and Nephrin (NEPH) were identified from urine exosomes. Results: Urine exosomal podocalyxin levels were significantly high at 10 week (n = 18; P = 0.001), 6 month (n = 25; P = 0.003) and 12 month (n = 14; P = 0.0001) time points. Nephrin levels were also noted to be high at 10 week (n = 18; P = 0.001) and 12 month (n = 14; P = 0.007) time points, compared with urine samples obtained from type 2 diabetes subjects who never had COVID-19. Though urinary podocyte-specific proteins were high, compared to control, there were no significant differences noted on urine albumin:creatinine ratios (UACR) between the groups. Conclusion: Persistent high levels of podocyte-specific proteins noted in urinary exosomes even at 12 months post-Covid may lead to the development of chronic kidney disease.
Collapse
Affiliation(s)
- Seshagiri Rao Nandula
- Department of Medicine and Biochemistry, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Beda Brichacek
- Department of Medicine and Biochemistry, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Sabyasachi Sen
- Department of Medicine and Biochemistry, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| |
Collapse
|
25
|
Uzoigwe CE. Long-lived Plasma Cells Can't Forget the Original Antigenic Sin. J Infect Dis 2025; 231:824-825. [PMID: 39680507 DOI: 10.1093/infdis/jiae621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024] Open
Affiliation(s)
- Chika Edward Uzoigwe
- Department of Medicine, Surgery & Science, Harcourt House, Sheffield, United Kingdom
| |
Collapse
|
26
|
Nègre P, Tayac D, Jamme T, Combis MS, Maupas-Schwalm F. Early suPAR levels as a predictor of COVID-19 severity: A new tool for efficient patient triage. Infect Dis Now 2025; 55:105058. [PMID: 40101896 DOI: 10.1016/j.idnow.2025.105058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/18/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Following several waves of the COVID-19 pandemic, we are now facing a lower but persistent rate of SARS-CoV-2 infections, with seasonal resurgences often coinciding with other respiratory tract infections. OBJECTIVE We aimed to identify early clinico-biological variables predictive of an unfavorable outcome in patients with primary SARS-CoV-2 infection. We also evaluated the role of suPAR, an innovative biomarker, in predicting disease severity. METHODS We included 255 patients with PCR-confirmed primary SARS-CoV-2 infection and with a 30-day follow-up minimum. Blood samples were collected within the first 24 h of hospitalization to measure suPAR levels. Comprehensive data from medical records were analyzed to assess their predictive value in stratifying patients into seven severity groups, with groups 1 to 3 representing severe COVID-19 (death, intubation, ECMO, or non-invasive ventilation). RESULTS Early plasma suPAR levels were significantly associated with severe disease progression, as evidenced by ANOVA and logistic regression models, highlighting suPAR as a persistent predictive factor for unfavorable outcomes. CONCLUSION Our findings suggest that a single suPAR measurement, performed early after a positive PCR test for SARS-CoV-2, holds strong predictive value for patient outcomes. This biomarker, alongside pulse oximetry and CT scan results, could be instrumental in early patient triage during seasonal COVID-19 resurgences.
Collapse
Affiliation(s)
- Pauline Nègre
- Faculty of Pharmacy, Toulouse III University, France; Medical Biochemistry Laboratory, CHU Toulouse, France
| | - Didier Tayac
- Medical Biochemistry Laboratory, CHU Toulouse, France
| | - Thibaut Jamme
- Medical Biochemistry Laboratory, CHU Toulouse, France
| | | | - Françoise Maupas-Schwalm
- Medical Biochemistry Laboratory, CHU Toulouse, France; Faculty of Medicine, Toulouse III University, France.
| |
Collapse
|
27
|
Wang J, Ma Y, Li Z, Yuan H, Liu B, Li Z, Su M, Habib G, Liu Y, Fu L, Wang P, Li M, He J, Chen J, Zhou P, Shi Z, Chen X, Xiong X. SARS-related coronavirus S-protein structures reveal synergistic RBM interactions underpinning high-affinity human ACE2 binding. SCIENCE ADVANCES 2025; 11:eadr8772. [PMID: 40085715 PMCID: PMC11908486 DOI: 10.1126/sciadv.adr8772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 02/07/2025] [Indexed: 03/16/2025]
Abstract
High-affinity and specific binding toward the human angiotensin-converting enzyme 2 (hACE2) receptor by severe acute respiratory syndrome coronavirus (SARS)-related coronaviruses (SARSr-CoVs) remains incompletely understood. We report cryo-electron microscopy structures of eight different S-proteins from SARSr-CoVs found across Asia, Europe, and Africa. These S-proteins all adopt tightly packed, locked, prefusion conformations. These structures enable the classification of SARSr-CoV S-proteins into three types, based on their receptor-binding motif (RBM) structures and ACE2 binding characteristics. Type-2 S-proteins often preferentially bind bat ACE2 (bACE2) over hACE2. We report a structure of a type-2 BtKY72-RBD in complex with bACE2 to understand ACE2 specificity. Structure-guided mutagenesis of BtKY72-RBD reveals that multiple synergistic mutations in four different regions of RBM are required to achieve high-affinity hACE2 binding. Similar RBM changes can also confer hACE2 binding to another type-2 BM48-31 S-protein, which is primarily non-ACE2 binding. These results provide an understanding of how high-affinity hACE2 binding may be acquired by SARSr-CoV S-proteins.
Collapse
Affiliation(s)
- Jingjing Wang
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yong Ma
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zimu Li
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
- Graduate School of Guangzhou Medical University, Guangzhou, China
| | - Hang Yuan
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Banghui Liu
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Zexuan Li
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengzhen Su
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Science and Technology of China, Hefei, China
| | - Gul Habib
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yutong Liu
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Lutang Fu
- Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, China
| | - Peiyi Wang
- Cryo-electron Microscopy Center, Southern University of Science and Technology, Shenzhen, China
| | - Mei Li
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Jun He
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jing Chen
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Peng Zhou
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Zhengli Shi
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Xiaoli Xiong
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Research Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
28
|
Shahrebabak AG, Rezaei M, Shahpar A, Nezhad NZ, Sarasyabi MS, Nakhaie M, Shahrebabak MG, Bahri RA. The efficacy of COVID-19 vaccination in cystic fibrosis patients: a systematic review. BMC Infect Dis 2025; 25:358. [PMID: 40082759 PMCID: PMC11907974 DOI: 10.1186/s12879-025-10736-6] [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: 11/19/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
This systematic review evaluates the efficacy and safety of COVID-19 vaccines in individuals with cystic fibrosis (CF). A systematic search of major databases conducted between December 2019 and January 2024 identified eight cohort studies comprising 1,361 CF patients. Studies without subgroup analyses specific to CF patients were excluded, which may have limited the generalizability of findings, particularly for CF lung transplant recipients. COVID-19 vaccines generally induced robust serological responses following the second and third doses, although reduced antibody levels were observed in lung transplant recipients. Factors influencing humoral response included prior SARS-CoV-2 infection, age, inhaled corticosteroid use, and immunosuppressive therapy. Vaccination-related adverse events were predominantly mild. Although breakthrough infections were reported, severe COVID-19 outcomes were infrequent among vaccinated CF patients. The evidence supports the immunogenicity and safety of COVID-19 vaccines in the CF patients. However, individualized vaccination strategies may be necessary for CF lung transplant recipients and those on immunosuppressive therapies. Further research is essential to optimize vaccination strategies and to identify risk factors associated with breakthrough infections in this high-risk population.
Collapse
Affiliation(s)
- Azam Gholami Shahrebabak
- Department of Pediatric ،Afzalipour Hospital, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Masoud Rezaei
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
| | - Amirhossein Shahpar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman, Iran
| | - Nazanin Zeinali Nezhad
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Sharifi Sarasyabi
- Medical Informatics Research Center, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohsen Nakhaie
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman, Iran
- Clinical Research Development Unit, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Gholami Shahrebabak
- Department of Pediatrics, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | | |
Collapse
|
29
|
Wang Z, Zhao L, Xie K. Development and validation of a nomogram to assess the occurrence of liver dysfunction in patients with COVID-19 pneumonia in the ICU. BMC Infect Dis 2025; 25:332. [PMID: 40065225 PMCID: PMC11892215 DOI: 10.1186/s12879-025-10684-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
The global pandemic of novel coronavirus pneumonia (COVID-19) has resulted in millions of deaths over the past three years. As one of the most commonly affected extra-pulmonary organs, numerous studies have reported varying degrees of liver injury in a significant proportion of patients with COVID-19, particularly in severe and critically ill patients. Early prediction of liver dysfunction in hospitalized patients would facilitate the clinical management of COVID-19 and improve clinical prognosis, but reliable and valid predictive models are still lacking.MethodsWe collected data from 286 patients with RT-PCR confirmed COVID-19 admitted to various ICUs from the case system. These patients were randomly divided into a training cohort (50%) and a validation cohort (50%). In the training cohort, we first used ROC curves to measure the predictive efficiency of each of the variables for the development of liver damage during hospitalization in patients with COVID-19, followed by LASSO regression analysis to screen the variables for predictive models and logistic regression analysis to identify relevant risk factors. A nomogram based on these variables was created following the above model. Finally, the efficiency of the prediction models in the training and validation cohorts was assessed using AUC, consistency index (C index), calibration curves and Decision Curve Analysis.ResultsOut of a total of 80 parameters for COVID-19 patients admitted to the ICUs, 10 were determined to be significantly associated with the occurrence of liver dysfunction during hospitalization. Based on these predictors, further prediction models were used to construct and develop a nomogram that was offered for practical clinical application. The C-index of the column line graphs for the training and validation cohorts was 0.956 and 0.844 respectively. in addition, the calibration curves for the model showed a high degree of agreement between the predicted and actual incidence of liver dysfunction in patients with COVID-19.ConclusionBy developing a predictive model and associated nomogram, we predicted the incidence of liver dysfunction during hospitalization in patients with COVID-19 in the ICU. The model's predictive performance was determined in both the training and validation cohorts, contributing to the clinical management of COVID-19.
Collapse
Affiliation(s)
- Zhiwei Wang
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Lina Zhao
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China.
- Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, Weifangaq, Weifang, Shandong, 261053, China.
| |
Collapse
|
30
|
Xue Y, Hou X, Zhong Y, Zhang Y, Du S, Kang DD, Wang L, Wang C, Li H, Wang S, Liu Z, Tian M, Guo K, Cao D, Deng B, McComb DW, Purisic E, Dai J, Hamon P, Brown BD, Tsankova NM, Merad M, Irvine DJ, Weiss R, Dong Y. LNP-RNA-mediated antigen presentation leverages SARS-CoV-2-specific immunity for cancer treatment. Nat Commun 2025; 16:2198. [PMID: 40038251 DOI: 10.1038/s41467-025-57149-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 02/13/2025] [Indexed: 03/06/2025] Open
Abstract
Lipid nanoparticle (LNP)-mRNA vaccines have demonstrated protective capability in combating SARS-CoV-2. Their extensive deployment across the global population leads to the broad presence of T-cell immunity against the SARS-CoV-2 spike protein, presenting an opportunity to harness this immunological response as a universal antigen target for cancer treatment. Herein, we design and synthesize a series of amino alcohol- or amino acid-derived ionizable lipids (AA lipids) and develop an LNP-RNA-based antigen presentation platform to redirect spike-specific T-cell immunity against cancer in mouse models. First, in a prime-boost regimen, AA2 LNP encapsulating spike mRNA elicit stronger T-cell immunity against the spike epitopes compared to FDA-approved LNPs (ALC-0315 and SM-102), highlighting the superior delivery efficiency of AA2 LNP. Next, AA15V LNP efficiently delivers self-amplifying RNAs (saRNAs) encoding spike epitope-loaded single-chain trimer (sSE-SCT) MHC I molecules into tumor tissues, thereby inducing the presentation of spike epitopes. Our results show that a single intratumoral (i.t.) treatment of AA15V LNP-sSE-SCTs suppresses tumor growth and extends the survival of B16F10 melanoma and A20 lymphoma tumor-bearing mice vaccinated with AA2 LNP-spike mRNA. Additionally, AA15V LNP-sSE-SCTs enable SE-SCT expression in ex vivo human glioblastoma and lung cancer samples, suggesting its potential in clinical translation.
Collapse
Affiliation(s)
- Yonger Xue
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xucheng Hou
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Yichen Zhong
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yuebao Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Shi Du
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diana D Kang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leiming Wang
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chang Wang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Haoyuan Li
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Siyu Wang
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhengwei Liu
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meng Tian
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kaiyuan Guo
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dinglingge Cao
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Binbin Deng
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, USA
| | - David W McComb
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH, USA
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, USA
| | - Eric Purisic
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jinye Dai
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pauline Hamon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian D Brown
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nadejda M Tsankova
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Darrell J Irvine
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Ron Weiss
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
31
|
Barberán J, Ramos M, Villanueva J, Villares P, Villareal M, Vivas M, Orche S, Tejera-Gonzalez M, Menéndez JM, Hinojosa LT, Almirall C, Antolin L, Martinez L, Mendoza S, Pelaez A, Segarra-Cañamares M, Guerrero JE, Pelaez J, Cardinal-Fernández P. Epidemiology of the COVID-19 pneumonia in a group of hospitals from Madrid-Spain during the full period of the State of Alarm HM cohort. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2025; 38:97-107. [PMID: 39950446 PMCID: PMC11894567 DOI: 10.37201/req/110.2024] [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: 11/13/2024] [Accepted: 01/16/2025] [Indexed: 03/08/2025]
Abstract
INTRODUCTION To describe the epidemiology pattern of the COVID-19 pandemic during all Spanish State of Alarm. METHODS Retrospective, observational, cohort and multicenter study. Inclusion criteria: age ≥18 years old, admitted for COVID-19 pneumonia in any of the centers of the HM Hospitals Group. Exclusion criteria: voluntary discharge, death in the emergency department, transfer to centers outside the HM group or incomplete data. State of Alarm period: 31/01/2020 to 05/07/2023. Predominant COVID-19 variant was defined when it exceeded 50% of the total isolates. RESULTS During the study period, 2,992 patients were admitted due to a COVID-19 pneumonia, 295 patients (9.86%) non-survive. Survivors and non-survivors were different in age and comorbidities. However, both cohorts presented a similar net of interaction between comorbidities. Hospital admissions per week showed an evolution in "peaks" with "troughs". A total of 197 (6.48%) patients were admitted to the ICU, of whom 52 (26.39%) non-survive; this subgroup stood out for having a higher proportion of septic shock, orotracheal intubation and acute renal failure, as well as a lower proportion of pulmonary thromboembolism and delirium. Concerning the viral variants, the incidence for the original variant was 4.05 cases/day, for the alpha variant 3.82 cases/day, for the delta variant 1.16 cases/day and for the omicron variant 1.35 cases/day. CONCLUSION Almost 1 of 10 patients with COVID-19 pneumonia death, a proportion that increased to 1 of 4 in case of being admitted to the ICU. Unexpectedly, interaction between comorbidities did not differ between survivors and non-survivors patients. Predominant variants were associated with different hospital admission rates but not influence the presence of peak-troughs evolution of the pandemic.
Collapse
Affiliation(s)
- José Barberán
- Hospital Universitario HM Monteprincipe, Madrid, Spain; Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | - María Ramos
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Julio Villanueva
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Paula Villares
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Mercedes Villareal
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital Universitario HM Torrelodones, Madrid, Spain
| | - María Vivas
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital Universitario HM Torrelodones, Madrid, Spain
| | - Susana Orche
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital Universitario HM Madrid, Madrid, Spain
| | - María Tejera-Gonzalez
- Hospital Universitario HM Monteprincipe, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital Universitario HM Torrelodones, Madrid, Spain
| | - Justo M Menéndez
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Lenin Tolentino Hinojosa
- Hospital Universitario HM Torrelodones, Madrid, Spain; Hospital Nacional Ramiro Prialé Huancayo, Perú
| | - Cristina Almirall
- Hospital Universitario HM Sanchinarro, Madrid, Spain; Laboratorio de análisis clínicos ABACID, Madrid, Spain
| | - Leonor Antolin
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Lady Martinez
- Hospital Universitario HM Monteprincipe, Madrid, Spain; Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | - Silvia Mendoza
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; Hospital Universitario HM Torrelodones, Madrid, Spain
| | - Adrián Pelaez
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | | | - José E Guerrero
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital Universitario HM Torrelodones, Madrid, Spain; Unidad de Cuidados Intensivos del Hospital Universitario "Gregorio Marañón", Madrid, Spain
| | - Jesús Pelaez
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital Universitario HM Torrelodones, Madrid, Spain
| | - Pablo Cardinal-Fernández
- Facultad HM de Ciencias de la Salud, Universidad Camilo José Cela, Madrid, Spain; Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain; Hospital Universitario HM Torrelodones, Madrid, Spain.
| |
Collapse
|
32
|
Chae DH, Park HS, Kim KM, Yu A, Park JH, Oh MK, Choi SW, Ryu J, Dunbar CE, Yoo HM, Yu KR. SARS-CoV-2 pseudovirus dysregulates hematopoiesis and induces inflammaging of hematopoietic stem and progenitor cells. Exp Mol Med 2025:10.1038/s12276-025-01416-1. [PMID: 40025168 DOI: 10.1038/s12276-025-01416-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 10/15/2024] [Accepted: 12/16/2024] [Indexed: 03/04/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection primarily affects the respiratory system but may induce hematological alterations such as anemia, lymphopenia and thrombocytopenia. Previous studies have reported that SARS-CoV-2 efficiently infects hematopoietic stem and progenitor cells (HSPCs); however, the subsequent effects on hematopoiesis and immune reconstitution have not yet been described. Here we evaluated the pathological effects of infection of umbilical-cord-blood-derived HSPCs with the SARS-CoV-2 Omicron variant pseudovirus (PsV). Transcriptomic analysis of Omicron PsV-infected HSPCs revealed the upregulation of genes involved in inflammation, aging and the NLRP3 inflammasome, suggesting a potential trigger of inflammaging. Omicron PsV-infected HSPCs presented decreased numbers of multipotential progenitors (granulocyte‒erythrocyte‒macrophage‒megakaryocyte colony-forming units) ex vivo and repopulated primitive hematopoietic stem cells (Ki-67-hCD34+ cells) in an HSPC transplantation NOD-scid IL2rγnull mouse model (Omicron mouse). Furthermore, Omicron PsV infection induced myeloid-biased differentiation of HSPCs. Treatment with nanographene oxide, an antiviral agent, partially mitigated the myeloid bias and inflammaging phenotype both in vitro and in vivo. These findings provide insights into the abnormal hematopoietic and immune effects of SARS-CoV-2 infection and highlight potential therapeutic interventions.
Collapse
Affiliation(s)
- Dong-Hoon Chae
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyun Sung Park
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kyoung-Myeon Kim
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
- Biometrology Group, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea
| | - Aaron Yu
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jae Han Park
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Mi-Kyung Oh
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | - Soon Won Choi
- Institutes of Convergence Technology, INBCT, Seoul, Republic of Korea
| | - Jaechul Ryu
- Institutes of Convergence Technology, INBCT, Seoul, Republic of Korea
| | - Cynthia E Dunbar
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Hee Min Yoo
- Biometrology Group, Korea Research Institute of Standards and Science, Daejeon, Republic of Korea.
- Department of Precision Measurement, University of Science and Technology, Daejeon, Republic of Korea.
| | - Kyung-Rok Yu
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
33
|
George J, Gautam D, Dominic MR, Malhotra R. Osteonecrosis following Steroid Therapy in COVID-19 Patients: An Outlook on the Emerging Problem. Hip Pelvis 2025; 37:26-37. [PMID: 40012145 PMCID: PMC11885787 DOI: 10.5371/hp.2025.37.1.26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/14/2024] [Accepted: 04/28/2024] [Indexed: 02/28/2025] Open
Abstract
Steroids are used in management of coronavirus disease 2019 (COVID-19) patients with severe illness and their use has been demonstrated to decrease mortality. Although life-saving, steroids are well documented as risk factors for osteonecrosis. Osteonecrosis of the hip can be debilitating and surgery may be required to improve the quality of life. With the increasing number of COVID-19 cases, osteonecrosis of the hip and other joints resulting from steroid use is expected to show a sharp rise in the coming years. In this review we discuss the association between steroids and osteonecrosis, indications for steroid therapy in COVID-19 patients, and incidence, diagnosis, and treatment of osteonecrosis secondary to steroids in COVID-19.
Collapse
Affiliation(s)
- Jaiben George
- Department of Orthopedic Surgery, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Deepak Gautam
- Department of Orthopedic Surgery, Medicover, Navi Mumbai, India
| | | | - Rajesh Malhotra
- Department of Orthopedic Surgery, Apollo Hospitals, New Delhi, India
| |
Collapse
|
34
|
Park S, Lee YW, Choi S, Jo H, Kim N, Cho S, Lee E, Choi EB, Park I, Jeon Y, Noh H, Seok SH, Oh SH, Choi YK, Kwon HK, Seo JY, Nam KT, Park JW, Choi KS, Lee HY, Yun JW, Seong JK. Post-COVID metabolic enzyme alterations in K18-hACE2 mice exacerbate alcohol-induced liver injury through transcriptional regulation. Free Radic Biol Med 2025; 229:1-12. [PMID: 39798903 DOI: 10.1016/j.freeradbiomed.2025.01.015] [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: 11/02/2024] [Revised: 12/22/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), poses a significant threat to global public health. Despite reports of liver injury during viral disease, the occurrence and detailed mechanisms underlying the development of secondary exogenous liver injury, particularly in relation to changes in metabolic enzymes, remain to be fully elucidated. Therefore, this study was aimed to investigate the mechanisms underlying SARS-CoV-2-induced molecular alterations in hepatic metabolism and the consequent secondary liver injury resulting from alcohol exposure. We investigated the potential effects of SARS-CoV-2 infection on alcohol-induced liver injury in Keratin 18 promoter-human angiotensin converting enzyme 2 (K18-hACE2) transgenic mice. Mice were intranasally infected with 1 × 102 PFU of SARS-CoV-2. Following a 14 d recovery period from infection, the recovered mice were orally administered alcohol at 6 g/kg. Prior SARS-CoV-2 infection aggravated alcohol-induced liver injury based on increased alanine aminotransferase levels and cytoplasmic vacuolation. Interestingly, infected mice exhibited lower blood alcohol levels and higher levels of acetaldehyde, a toxic alcohol metabolite, compared to uninfected mice after the same period of alcohol consumption. Along with alterations of several metabolic process-related terms identified through RNA sequencing, notably, upregulation of cytochrome P450 2E1 (CYP2E1) and CYP1A2 was observed in infected mice compared to control value prior to alcohol exposure, with no significant impact of SARS-CoV-2 on intestinal damage. Tumor necrosis factor-alpha persistently showed upregulated expression in the infected mice; it also enhanced aryl hydrocarbon receptor and Sp1 expressions and their binding activity to Cyp1a2 and Cyp2e1 promoters, respectively, in hepatocytes, promoting the upregulation of their transcription. Our findings suggest that SARS-CoV-2 infection exacerbates alcohol-induced liver injury through the transcriptional activation of Cyp1a2 and Cyp2e1, providing valuable insights for the development of clinical recommendations on long COVID.
Collapse
Affiliation(s)
- SiYeong Park
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Youn Woo Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, 23488, Republic of Korea
| | - Seunghoon Choi
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea; Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 PLUS, Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Harin Jo
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - NaHyun Kim
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sumin Cho
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eunji Lee
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Eun-Bin Choi
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Inyoung Park
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young Jeon
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyuna Noh
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea; Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 PLUS, Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sang-Hyuk Seok
- Laboratory of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Hyun Oh
- Laboratory of Histology, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, 05029, Republic of Korea
| | - Ho-Keun Kwon
- Department of Microbiology and Immunology and Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jun-Young Seo
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Ki Taek Nam
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea; Graduate School of Medical Science, BK21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jun Won Park
- Laboratory of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kang-Seuk Choi
- Laboratory of Avian Diseases, BK21 PLUS Program for Veterinary Science and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam, 23488, Republic of Korea.
| | - Jun-Won Yun
- Laboratory of Veterinary Toxicology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Je Kyung Seong
- Korea Mouse Phenotyping Center, Seoul National University, Seoul, 08826, Republic of Korea; Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, and BK21 PLUS, Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea; Interdisciplinary Program for Bioinformatics, Program for Cancer Biology and BIO-MAX/N-Bio Institute, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
35
|
Ahmad S, Alafnan A, Alobaida A, Shahab U, Rehman S, Khan S, Khan MY, Puri P, Pandey RP, Ahmad I, Rafi Z. Decoding the SARS-CoV-2 infection process: Insights into origin, spread, and therapeutic approaches. Microb Pathog 2025; 200:107328. [PMID: 39863091 DOI: 10.1016/j.micpath.2025.107328] [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: 08/09/2024] [Revised: 10/29/2024] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Globally, over 768 million confirmed cases and 6.9 million deaths had been documented as of July 17, 2023. Coronaviruses have a relatively large RNA genome. As with other viruses, SARS-CoV-2 does have an envelope film produced from host cells that are assisted by virally encoded glycoproteins that are required for infectivity, immunological assault, and viral particle production. Although the intermediate source of origin and transmission to humans is unexplained, rapid transmission from human to human has been established. This review focuses on the mechanistic framework for understanding the SARS-CoV-2 viral infection. Additionally, it discusses the origins and implications of COVID-19 using direct quotations from the published scientific literature to avoid misinterpretation of this catastrophic event that resulted in a massive loss of human life and impact on the global economy. The current available information unfolds large number of topics related with COVID-19 and/or the coronavirus (SARS-CoV-2) responsible of the disease. This review article also delves into the multifaceted aspects of COVID-19 and SARS-CoV-2, with a specific focus on a controversial yet essential issue: the possible association between SARS-CoV-2's origin and aldose reductase, an enzyme known for its role in diabetic retinopathy. Exploring this connection holds utmost significance, offering valuable insights into COVID-19's pathogenesis and unlocking new avenues for therapeutic interventions. It is important to trace back the evolution of coronaviruses and reveal the possible origin of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes the coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, 2440, Saudi Arabia.
| | - Ahmed Alafnan
- Department of Pharmacology, College of Pharmacy, University of Hail, Hail City, 2440, Saudi Arabia.
| | - Ahmed Alobaida
- Department of Pharmacology, College of Pharmacy, University of Hail, Hail City, 2440, Saudi Arabia.
| | - Uzma Shahab
- Department of Pharmacology, College of Pharmacy, University of Hail, Hail City, 2440, Saudi Arabia.
| | - Shahnawaz Rehman
- IIRC-1, Laboratory of Glycation Biology and Metabolic Disorders, Integral University, U.P., India.
| | - Saif Khan
- Department of Basic Dental and Medical Sciences, College of Dentistry, University of Hail, 2440, Hail, 2440, Saudi Arabia.
| | - Mohd Yasir Khan
- Department of Biotechnology, School of Applied & Life Science, Uttaranchal University Dehradun, India.
| | - Paridhi Puri
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India.
| | - Ramendra Pati Pandey
- Department of Biotechnology, SRM University, Delhi-NCR, Sonepat, Haryana, 131029, India.
| | - Irfan Ahmad
- Central Labs, King Khalid University, AlQura'a, P.O. Box 960, Abha, Saudi Arabia; Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Zeeshan Rafi
- Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, 226026, India.
| |
Collapse
|
36
|
Zhou B, Zhang Y, Han S, Zhang J, Song L, Wang H. Myocardial Dysfunction and Risk of Long COVID in Patients Recovered From Mild and Moderate COVID-19. Echocardiography 2025; 42:e70120. [PMID: 40029146 PMCID: PMC11875044 DOI: 10.1111/echo.70120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/16/2025] [Accepted: 02/22/2025] [Indexed: 03/05/2025] Open
Abstract
PURPOSE Numerous recovered COVID-19 patients exhibit persistent cardiovascular symptoms. However, the degree of myocardial dysfunction and its associated risk factors remain unclear. This study aims to evaluate myocardial dysfunction in recovered patients and pinpoint predictors of persistent cardiovascular symptoms. METHODS We reviewed the echocardiograms of patients who recovered from mild or moderate COVID-19 and presented with cardiovascular symptoms during the Omicron surge. Myocardial strain was analyzed in 546 patients before and after infection, and in 351 prepandemic healthy controls. Clinical follow-up at 12 months post-infection was used to evaluated symptom persistence, and multivariable logistic regression was used to identify independent predictors. RESULTS Baseline characteristics showed no significant differences between patients and controls (all p > 0.05). Although the left ventricle global longitudinal strain (LVGLS) remained stable post-infection, significant reductions emerged in regional left ventricle longitudinal strains (LVLS) and all left atrial strains (LAS) (all p < 0.05). Persistent cardiovascular symptoms affected 16.5% (90/546) of patients at 1-year follow-up. Multivariate analysis showed that only LA conduit strain (OR = 0.919, 95% CI: 0.857, 0.985, p = 0.017) and basal inferoseptal LVLS (OR = 0.883, 95% CI: 0.792, 0.986, p = 0.026) correlated with persisting cardiovascular symptoms. CONCLUSION Our findings demonstrate that subclinical but persistent COVID-19-associated myocardial dysfunction is characterized by regional LVLS impairment and LAS reduction. The identified strain parameters (LAScd and basal inferoseptal LVLS) serve as novel imaging markers for stratifying patients at risk of persistent cardiovascular symptoms. These results advocate for targeted echocardiographic surveillance and early intervention strategies in post-COVID care pathways. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT06170307.
Collapse
Affiliation(s)
- Binyu Zhou
- Department of Medical UltrasoundThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Yiran Zhang
- Department of Medical UltrasoundThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Shuang Han
- Department of Medical UltrasoundThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Jiqing Zhang
- Department of Medical UltrasoundThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Lin Song
- Department of Medical UltrasoundThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| | - Haiyan Wang
- Department of Medical UltrasoundThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanChina
| |
Collapse
|
37
|
Apiwattanakul M, Ounmuang C, Aungsumart S. Impact of COVID-19 and vaccination on multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD). Mult Scler Relat Disord 2025; 95:106316. [PMID: 39954490 DOI: 10.1016/j.msard.2025.106316] [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: 04/16/2024] [Revised: 08/10/2024] [Accepted: 02/01/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Concerns have arisen regarding the potential impact of the coronavirus disease 2019 (COVID-19) pandemic on increasing the incidence and relapse rates of multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD) due to vaccination against severe acute respiratory syndrome coronavirus-2. However, confirmation of this relationship is still lacking. OBJECTIVE The primary objective of this study was to compare relapse rates among patients with MS and NMOSD between the pre-pandemic and pandemic eras. The secondary objective was to determine the incidence of newly diagnosed cases of MS and NMOSD and analyze the relationship between COVID-19/vaccination and relapse. METHODS We divided the study period into two phases: the pre-pandemic era (January 1, 2017, to December 31, 2019) and the pandemic era (January 1, 2020, to December 31, 2022). Relapse events were compared between the two periods. Multivariate time-to-event techniques were used to assess relapse rates between both periods using the Anderson-Gill (AG), Prentice, Williams, and Peterson (PWP), total time (TT) and gap time (GT), and Cox frailty models. RESULTS A total of 697 data contributions from 405 patients with MS and NMOSD were evaluated and categorized into two groups: the pre-pandemic group (n = 328) and the pandemic group (n = 369). A total of 1720.21 person-years were recorded, with 794.38 and 925.83 person-years before and during the pandemic, respectively. The results showed that the relapse rates of MS and NMOSD did not differ significantly between the pre-pandemic and pandemic periods (p = 0.96). The relapse rate did not change significantly between the two periods after adjusting for rituximab use and disease duration, as demonstrated by the different statistical models for multiple events (AG model), event-specific inter-individual analyses (PWP-TT and PWP-GT), and intra-individual analyses (Cox frailty model). CONCLUSION These results suggest that there is insufficient evidence to establish a link between COVID-19 or COVID-19 vaccination and a higher frequency of relapse in patients with MS and NMOSD.
Collapse
Affiliation(s)
- Metha Apiwattanakul
- Department of Neurology, Neurological Institute of Thailand, Bangkok, Thailand
| | - Chawarat Ounmuang
- Department of Neurology, Neurological Institute of Thailand, Thailand
| | - Saharat Aungsumart
- Department of Neurology, Neurological Institute of Thailand, Bangkok, Thailand.
| |
Collapse
|
38
|
Cekin N, Akin S, Pinarbasi E, Doğan OH. Impact of IL-6 rs1800795 and rs1800796 polymorphisms on clinical outcomes of COVID-19: a study on severity of disease in Turkish population. Mamm Genome 2025; 36:213-229. [PMID: 39567384 DOI: 10.1007/s00335-024-10085-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/09/2024] [Indexed: 11/22/2024]
Abstract
Coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is exacerbated by cytokine storms, leading to severe inflammation. Interleukin-6 (IL-6) plays a critical role in this process, and variations in its promoter may influence disease severity. This study aims to investigate the relationship between IL6 promoter polymorphisms rs1800795 (G > C) and rs1800796 (G > C) and the severity of COVID-19 in the Turkish population. A total of 332 participants were included: 84 control, 80 with mild COVID-19, and 168 with severe COVID-19. IL6 polymorphisms were genotyped using the restriction fragment length polymorphism (RFLP) method. The genotypes rs1800795 GC (OR = 3.00, 95% CI: 1.669-5.398, p < 0.000), CC (OR = 7.44, 95% CI: 2.899-19.131, p < 0.000), and rs1800796 GC (OR = 2.76, 95% CI: 1.603-4.761, p < 0.000), as well as the alleles rs1800795 C (OR = 3.01, p < 0.000) and rs1800796 C (OR = 1.97, p = 0.002), may be associated with the severity of COVID-19. According to the Jonckheere-Terpstra (J-T) test, the most significant trends that vary linearly with disease severity were observed for D-dimer [J-T = 15.896, Effect size = 0.68 (0.61 to 0.76), p < 0.000] and CRP [J-T = 15.389, Effect size = 0.66 (0.59 to 0.73), p < 0.000]. The distribution of clinical parameters across genotype combinations (rs1800796/rs1800795*) showed that GC/GC* and GC/CC* were linked to a higher risk of severe inflammation, clotting, and organ damage. Additionally, it has been determined that the G-C and C-C haplotypes may be associated with increased severity of COVID-19. The rs1800795 and rs1800796 polymorphisms are linked to COVID-19 severity and could help guide future treatment strategies.
Collapse
Affiliation(s)
- Nilgun Cekin
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey.
- Faculty of Medicine, Department of Medical Biochemistry, Sivas Cumhuriyet University, Sivas, Turkey.
| | - Seyda Akin
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Ergun Pinarbasi
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Okan Halef Doğan
- Department of Medical Biology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
39
|
Li Y, Wang J, Huang B, Wang R, Xu Y, Zhang P, Wang R, Wang W, Cao J, Hou M, Hou Y. Nucleoside reverse transcriptase inhibition enhances platelet production and megakaryocyte maturation in patients with COVID-19. Br J Haematol 2025; 206:846-857. [PMID: 39930897 DOI: 10.1111/bjh.20006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/28/2025] [Indexed: 03/08/2025]
Abstract
Coronavirus disease 2019 (COVID-19) is a systemic infection frequently involving the haematopoietic system. Thrombocytopenia is associated with increased risks of severe disease progression and mortality. Antiviral agents have shown much promise in decreasing viral load and shortening the time to the resolution of symptoms. However, their effect on platelet counts in patients with COVID-19 remains unexplored. Therefore, we first performed a retrospective study to evaluate the variation in platelet mass of hospitalized patients with high-risk COVID-19 who were given either SARS-CoV-2 main protease inhibitor, nucleoside reverse transcriptase inhibitor or no antiviral agents. A total of 177 patients were included, among which 64 received azvudine, 12 received nirmatrelvir-ritonavir and 101 patients received none. Compared to those without antiviral treatment, significantly higher platelet counts' increments were observed in patients receiving azvudine or nirmatrelvir-ritonavir (p = 0.001). Of note, this elevation was significantly more profound in the azvudine group than that in the control group (p < 0.001) or the nirmatrelvir-ritonavir group (p = 0.042). Subsequently, in vitro experiments were conducted to investigate the mechanism of platelet elevation underlying the activity of azvudine. Results showed that azvudine promoted the polyploidization and platelet production of the MEG-01 cell line. Although azvudine had minimal effect on megakaryopoiesis, it could significantly trigger platelet release of megakaryocytes in the presence of SARS-CoV-2 spike-membrane recombinant fusion protein or not. Finally, RNA-sequencing demonstrated that azvudine-treated MEG-01 exhibited a marked increase in VWF, TUBB1 and GP1BA, and upregulated genes associated with the PI3K/AKT and JAK/STAT signalling pathways. In conclusion, our findings indicated that nucleoside reverse transcriptase inhibition potentially enhances platelet production and megakaryocyte maturation in patients with COVID-19, suggesting a new therapeutic option for thrombocytopenia.
Collapse
Affiliation(s)
- Yubin Li
- Department of Hematology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Juan Wang
- Department of Hematology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Bingqian Huang
- Department of Hematology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Ruixue Wang
- Department of Hematology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Yitong Xu
- Department of Hematology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Ping Zhang
- Department of Hematology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Ruting Wang
- Department of Hematology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Wanru Wang
- Department of Hematology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Junying Cao
- Department of Hematology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| | - Yu Hou
- Department of Hematology, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, China
| |
Collapse
|
40
|
Unterberger S, Terrazzini N, Sacre S. Convalescent COVID-19 monocytes exhibit altered steady-state gene expression and reduced TLR2, TLR4 and RIG-I induced cytokine expression. Hum Immunol 2025; 86:111249. [PMID: 39922089 DOI: 10.1016/j.humimm.2025.111249] [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: 08/23/2024] [Revised: 01/12/2025] [Accepted: 01/21/2025] [Indexed: 02/10/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes COVID-19, can induce trained immunity in monocytes. Trained immunity is the result of metabolic and epigenetic reprogramming of progenitor cells leading to an altered inflammatory response to subsequent activation. To investigate the monocyte response 3-6 months post SARS-CoV-2 infection, steady-state gene expression and innate immune receptor stimulation were investigated in monocytes from unvaccinated SARS-CoV-2 naïve individuals and convalescent COVID-19 participants. The differentially expressed genes (DEGs) identified were involved in the regulation of innate immune signalling pathways associated with anti-viral defence. COVID-19 participants who had experienced severe symptoms exhibited a larger number of DEGs than participants that had mild symptoms. Interestingly, genes encoding receptors that recognise SARS-CoV-2 RNA were downregulated. DDX58, encoding retinoic-acid inducible gene I (RIG-I), was downregulated which corresponded with a reduced response to RIG-I activation. Furthermore, toll-like receptor (TLR)1/2 and TLR4 activation also exhibited reduced cytokine secretion from convalescent COVID-19 monocytes. These data suggest that following SARS-CoV-2 infection, monocytes exhibit altered steady-state gene expression and reduced responsiveness to innate immune receptor activation. As both RIG-I and TLRs recognise components of SARS-CoV-2, this may lead to a moderated inflammatory response to SARS-CoV-2 reinfection in the months following the initial infection.
Collapse
Affiliation(s)
- Sarah Unterberger
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Nadia Terrazzini
- Centre for Regenerative Medicine and Devices, School of Applied Sciences, University of Brighton, Brighton, UK
| | - Sandra Sacre
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK.
| |
Collapse
|
41
|
Straub J, Estrada Lobato E, Paez D, Langs G, Prosch H. Artificial intelligence in respiratory pandemics-ready for disease X? A scoping review. Eur Radiol 2025; 35:1583-1593. [PMID: 39570367 PMCID: PMC11835992 DOI: 10.1007/s00330-024-11183-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/02/2024] [Accepted: 09/26/2024] [Indexed: 11/22/2024]
Abstract
OBJECTIVES This study aims to identify repeated previous shortcomings in medical imaging data collection, curation, and AI-based analysis during the early phase of respiratory pandemics. Based on the results, it seeks to highlight essential steps for improving future pandemic preparedness. MATERIALS AND METHODS We searched PubMed/MEDLINE, Scopus, and Cochrane Reviews for articles published from January 1, 2000, to December 31, 2021, using the terms "imaging" or "radiology" or "radiography" or "CT" or "x-ray" combined with "SARS," "MERS," "H1N1," or "COVID-19." WHO and CDC Databases were searched for case definitions. RESULTS Over the last 20 years, the world faced several international health emergencies caused by respiratory diseases such as SARS, MERS, H1N1, and COVID-19. During the same period, major technological advances enabled the analysis of vast amounts of imaging data and the continual development of artificial intelligence algorithms to support radiological diagnosis and prognosis. Timely availability of data proved critical, but so far, data collection attempts were initialized only as individual responses to each outbreak, leading to long delays and hampering unified guidelines and data-driven technology to support the management of pandemic outbreaks. Our findings highlight the multifaceted role of imaging in the early stages of SARS, MERS, H1N1, and COVID-19, and outline possible actions for advancing future pandemic preparedness. CONCLUSIONS Advancing international cooperation and action on these topics is essential to create a functional, effective, and rapid counteraction system to future respiratory pandemics exploiting state of the art imaging and artificial intelligence. KEY POINTS Question What has been the role of radiological data for diagnosis and prognosis in early respiratory pandemics and what challenges were present? Findings International cooperation is essential to developing an effective rapid response system for future respiratory pandemics using advanced imaging and artificial intelligence. Clinical relevance Strengthening global collaboration and leveraging cutting-edge imaging and artificial intelligence are crucial for developing rapid and effective response systems. This approach is essential for improving patient outcomes and managing future respiratory pandemics more effectively.
Collapse
Affiliation(s)
- Jennifer Straub
- Computational Imaging Research Lab, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090, Vienna, Austria
| | - Enrique Estrada Lobato
- Nuclear Medicine and Diagnostic Imaging Section, Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency (IAEA), 1220, Vienna, Austria
| | - Diana Paez
- Nuclear Medicine and Diagnostic Imaging Section, Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency (IAEA), 1220, Vienna, Austria
| | - Georg Langs
- Computational Imaging Research Lab, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090, Vienna, Austria.
- Christian Doppler Laboratory for Machine Learning Driven Precision Imaging, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090, Vienna, Austria.
| | - Helmut Prosch
- Christian Doppler Laboratory for Machine Learning Driven Precision Imaging, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090, Vienna, Austria
- Division of General and Paediatric Radiology, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, 1090, Vienna, Austria
| |
Collapse
|
42
|
Nakakubo S. Evolving COVID-19 symptoms and the ongoing course of research. THE LANCET. INFECTIOUS DISEASES 2025; 25:245-246. [PMID: 39419048 DOI: 10.1016/s1473-3099(24)00668-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Affiliation(s)
- Sho Nakakubo
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan.
| |
Collapse
|
43
|
Yi JP, Yoon CI, Lim SH, Choi H, Oh SJ, Kim H, Park DS, Baek JM, Kim YS, Jeon YW, Rhu J, Kang YJ. Prognosis of Patients with Breast Cancer Following Delayed Diagnosis During the COVID-19 Pandemic: A Real-World Cohort Study. Ann Surg Oncol 2025; 32:2029-2037. [PMID: 39633173 DOI: 10.1245/s10434-024-16645-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic has globally impacted healthcare systems, delaying cancer screening, diagnosis, and treatment. South Korea experienced a 3-month lockdown during the peak of the pandemic, leading to postponements in breast cancer diagnosis and treatment. Although the pandemic's severity has decreased, addressing the treatment gap remains a concern. METHODS This retrospective cohort study included patients diagnosed with breast cancer at five academic hospitals in South Korea during the pre-COVID (May-July 2019) and COVID (May-July 2020) periods. The primary endpoint was recurrence-free survival (RFS), and secondary endpoints included overall survival (OS), stage distribution, and age subgroup analyses. RESULTS Among 716 patients (374 pre-COVID, 342 COVID), the 3-year RFS was significantly lower in the COVID group (88.3 vs. 89.7%, p = 0.030), while the 3-year OS was similar between groups (96.2 vs. 95.5%, p = 0.439). Age subgroup analysis revealed a significant RFS difference in patients under 65 years of age (p = 0.005) but not in those aged 65 years and older (p = 0.682). Stage distribution did not differ significantly between groups (p = 0.531). CONCLUSIONS The COVID-19 pandemic adversely affected 3-year RFS, especially among younger patients, despite no significant increase in advanced-stage distribution. Timely cancer screening remains crucial for young patients with fewer infectious disease complications.
Collapse
Affiliation(s)
- Jae Pak Yi
- Department of Surgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
- Department of Medicine, Graduate School, Kyung Hee University, Seoul, Korea
| | - Chang Ik Yoon
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Su Hyun Lim
- Department of Surgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
| | - Hoon Choi
- Department of Surgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
| | - Se Jeong Oh
- Department of Surgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
| | - Hyobin Kim
- Department of Surgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
| | - Dae Sun Park
- Department of Surgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
| | - Jong Min Baek
- Department of Surgery, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yong-Seok Kim
- Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Korea
| | - Ye Won Jeon
- Department of Surgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Korea
| | - Jiyoung Rhu
- Department of Surgery, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Bucheon, Korea
| | - Young-Joon Kang
- Department of Surgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea.
| |
Collapse
|
44
|
Sahli W, Vitte J, Desnues B. Eosinophils and COVID-19: Insights into immune complexity and vaccine safety. Clin Transl Allergy 2025; 15:e70050. [PMID: 40120088 PMCID: PMC11929522 DOI: 10.1002/clt2.70050] [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: 08/27/2024] [Revised: 02/23/2025] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND COVID-19 exhibits a variety of symptoms and may lead to multi-organ failure and death. This clinical complexity is exacerbated by significant immune dysregulation affecting nearly all cells of the innate and adaptive immune system. Granulocytes, including eosinophils, are affected by SARS-CoV-2. OBJECTIVES Eosinophil responses remain poorly understood despite early recognition of eosinopenia as a hallmark feature of COVID-19 severity. RESULTS The heterogeneous nature of eosinophil responses categorizes them as dual-function cells with contradictory effects. Eosinophil activation can suppress virus-induced inflammation by releasing type 2 cytokines like IL-13 and granular proteins with antiviral action such as eosinophil-derived neurotoxins and eosinophil cationic protein, and also by acting as antigen-presenting cells. In contrast, eosinophil accumulation in the lungs can induce tissue damage triggered by cytokines or hormones like IFN-γ and leptin. Additionally, they can affect adaptive immune functions by interacting with T cells through direct formation of membrane complexes or soluble mediator action. Individuals with allergic disorders who have elevated levels of eosinophils in tissues and blood, such as asthma, do not appear to be at an increased risk of developing severe COVID-19 following SARS-CoV-2 infection. However, the SARS-CoV-2 vaccine appears to be associated with complications and eosinophilic infiltrate-induced immunopathogenicity, which can be mitigated by corticosteroid, anti-histamines and anti-IL-5 therapy and avoided by modifying adjuvants or excipients. CONCLUSION This review highlights the importance of eosinophils in COVID-19 and contributes to a better understanding of their role during natural infection and vaccination.
Collapse
Affiliation(s)
- Wided Sahli
- Aix Marseille UniversityMEPHIMarseilleFrance
- IHU‐Méditerranée InfectionMarseilleFrance
| | - Joana Vitte
- Laboratory of ImmunologyUniversity Hospital of ReimsReimsFrance
- INSERM UMR‐S 1250 P3CELLUniversity of ReimsReimsFrance
| | - Benoit Desnues
- Aix Marseille UniversityMEPHIMarseilleFrance
- IHU‐Méditerranée InfectionMarseilleFrance
| |
Collapse
|
45
|
Fratta Pasini AM, Stranieri C, Di Leo EG, Bertolone L, Aparo A, Busti F, Castagna A, Vianello A, Chesini F, Friso S, Girelli D, Cominacini L. Identification of Early Biomarkers of Mortality in COVID-19 Hospitalized Patients: A LASSO-Based Cox and Logistic Approach. Viruses 2025; 17:359. [PMID: 40143288 DOI: 10.3390/v17030359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/06/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
This study aimed to identify possible early biomarkers of mortality among clinical and biochemical parameters, iron metabolism parameters, and cytokines detected within 24 h from admission in hospitalized COVID-19 patients. We enrolled 80 hospitalized patients (40 survivors and 40 non-survivors) with COVID-19 pneumonia and acute respiratory failure. The median time from the onset of COVID-19 symptoms to hospital admission was lower in non-survivors than survivors (p < 0.05). Respiratory failure, expressed as the ratio of arterial oxygen partial pressure to the fraction of inspired oxygen (P/F), was more severe in non-survivors than survivors (p < 0.0001). Comorbidities were similar in both groups. Among biochemical parameters and cytokines, eGFR and interleukin (IL)-1β were found to be significantly lower (p < 0.05), while LDH, IL-10, and IL-8 were significantly higher in non-survivors than in survivors (p < 0.0005, p < 0.05 and p < 0.005, respectively). Among other parameters, LDH values distribution showed the most significant difference between study groups (p < 0.0001). LASSO feature selection combined with Cox proportional hazards and logistic regression models was applied to identify features distinguishing between survivors and non-survivors. Both approaches highlighted LDH as the strongest predictor, with IL-22 and creatinine emerging in the Cox model, while IL-10, eGFR, and creatinine were influential in the logistic model (AUC = 0.744 for Cox, 0.723 for logistic regression). In a similar manner, we applied linear regression for predicting LDH levels, identifying the P/F ratio as the top predictor, followed by IL-10 and eGFR (NRMSE = 0.128). Collectively, these findings underscore LDH's critical role in mortality prediction, with P/F and IL-10 as key determinants of LDH increases in this Italian COVID-19 cohort.
Collapse
Affiliation(s)
- Anna Maria Fratta Pasini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Chiara Stranieri
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Edoardo Giuseppe Di Leo
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Lorenzo Bertolone
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Antonino Aparo
- Interdepartmental Laboratory of Medical Research, Research Center LURM, University of Verona, 37134 Verona, Italy
| | - Fabiana Busti
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Annalisa Castagna
- Department of Medicine, Section of Internal Medicine B, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Alice Vianello
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Fabio Chesini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Simonetta Friso
- Department of Medicine, Section of Internal Medicine B, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Domenico Girelli
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| | - Luciano Cominacini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy
| |
Collapse
|
46
|
Guttmann A, Heidinger A, Woltsche N, Brodmann M, Kurzmann-Gütl K, Nemecz V, Kaindl M, Wurzer H, Schwantzer G, Horwath-Winter J. Ocular symptoms in COVID-19 patients with a history of hospitalization in the first pandemic wave in Styria, Austria. FRONTIERS IN OPHTHALMOLOGY 2025; 5:1540904. [PMID: 40084349 PMCID: PMC11903703 DOI: 10.3389/fopht.2025.1540904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
Purpose Our study aimed to investigate the prevalence and timing of ocular surface manifestations in hospitalized COVID-19 patients, providing insights into the occurrence of eye involvement before, during, or after the illness. This study contributes to understanding the extent of ocular involvement in COVID-19, which has been suggested to occur due to potential viral entry through the eyes. Methods 451 confirmed COVID-19 patients had a history of hospitalization in Styria, Austria. The study included 176 patients aged 18-95 years who tested positive for SARS-CoV-2 in nasopharyngeal swabs by RT-PCR and received treatment at two hospitals. Telephone interviews were conducted after recovery, focusing on ocular symptoms and medical history (openMEDOCS). Results Seventeen percent (n=30) reported new-onset ocular symptoms in the context of COVID-19. Patients with ocular symptoms were younger (p<0.001). Sore throat (p=0.013) and high fever (p=0.038) were significantly more prevalent in patients with new-onset ocular symptoms. Persistent ocular symptoms beyond the duration of hospitalization affected more than half (56.7%) of the participants with new-onset ocular symptoms. However, there were no differences in blood parameters, lung imaging, or comorbidities between groups with and without ocular symptoms. Conclusions In hospitalized COVID-19 patients, ocular symptoms occur with a significant prevalence of 17%. Younger age (p<0.001) and the presence of sore throat (p=0.013) are associated with an increased risk of developing new-onset ocular symptoms in the context of COVID-19.
Collapse
Affiliation(s)
- Andreas Guttmann
- Department of Ophthalmology, Medical University Graz, Graz, Austria
| | - Astrid Heidinger
- Department of Ophthalmology, Medical University Graz, Graz, Austria
| | - Nora Woltsche
- Department of Ophthalmology, Medical University Graz, Graz, Austria
| | | | | | - Viktoria Nemecz
- Division of Angiology, Medical University Graz, Graz, Austria
| | - Matthias Kaindl
- Division of Angiology, Medical University Graz, Graz, Austria
| | - Herbert Wurzer
- Department of Internal Medicine, LKH Graz II, Graz, Austria
| | - Gerold Schwantzer
- Institute for Medical Informatics, Statistics and Documentation, Medical University Graz, Graz, Austria
| | | |
Collapse
|
47
|
Bertron E, Doutrelon C, Etchegoyen G, Gislot C, Clausse O, Verret C, Coste S, Josnard J. Characterizing Symptom Profiles in a Military Outpatient Cohort With COVID-19 in the Île-de-France Region. Mil Med 2025; 190:e838-e844. [PMID: 39235768 DOI: 10.1093/milmed/usae412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/10/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024] Open
Abstract
INTRODUCTION The emergence of coronavirus disease 2019 (COVID-19) pandemic, driven by severe acute respiratory syndrome coronavirus 2, precipitated an unprecedented public health crisis, necessitating comprehensive response worldwide. The Ile-De-France region has been particularly affected, leading to rapid health care system strain and hospital saturation. Within this context, the 1st Armed Forces Medical Centre swiftly adapted its primary care services to cater to the unique needs of military personnel, leveraging telemedicine technologies for efficient health care delivery. MATERIALS AND METHODS This study undertook an observational retrospective analysis to characterize the epidemiological profile of soldiers seeking outpatient care for non-severe COVID-19 symptoms at the 1st Armed Forces Medical Centre's sub-centers between March and December 2020. Electronic medical records of patients were scrutinized to discern patterns in symptom presentation and evolution over time. RESULTS The cross-section sample consisted of 519 patients, predominantly males (71%), with an average age of 38 years. Predominant symptoms reported were asthenia (43%), cephalalgia (41%), myalgia (40%), pyrexia (38%), and tussis (33%). A significant proportion of patients (33%) were asymptomatic at the follow-up consultations, marking a notable increase from initial assessments (14%). Although symptoms indicative of flu-like syndrome and sore throat exhibited regression between consultations, certain manifestations, such as anosmia, ageusia, and dyspnea, persisted without significant variance. Our analysis delineated five distinct symptom profiles within the cohort. CONCLUSIONS This study provides valuable insights into the clinical course of non-severe COVID-19 cases among military personnel receiving outpatient care, delineating five distinct symptom profiles. This complexity underscores the challenges in predicting and managing symptomatology effectively. Our findings align with existing literature but offer a unique perspective on military cohorts. Notably, in confined military settings with predominantly young and healthy individuals, symptomatic presentations tend to be milder. Robust public health interventions, including quarantine and contact tracing, are crucial to maintaining operational readiness amidst the pandemic's challenges.
Collapse
Affiliation(s)
- Emilie Bertron
- Ecole du Val-De-Grâce, Val-De Grâce, Paris 75005, France
| | - Caroline Doutrelon
- Service de Médecine Interne et Rhumatologie, Hopital d'Instruction des Armées Percy, Clamart 92140, France
| | - Gabriel Etchegoyen
- 170ème Antenne Médicale de Castelnaudary, 11ème Centre Médical des Armées, Castelnaudary 11400, France
| | - Charlotte Gislot
- 6ème antenne médicale de Vincennes, 1er Centre Médical des Armées, Vincennes 94304, France
| | - Odile Clausse
- 1ère antenne médicale de Balard, 1er Centre Médical des Armées, Paris 75115, France
| | - Catherine Verret
- Direction de la Recherche et de la Formation, Val-De-Grâce, Paris 75005, France
| | | | - Justine Josnard
- 145ème Antenne Médicale de Draguignan, 9ème Centre Médical des Armées, Draguignan 83007, France
| |
Collapse
|
48
|
Popazu C, Romila A, Petrea M, Grosu RM, Lescai AM, Vlad AL, Oprea VD, Baltă AAȘ. Overview of Inflammatory and Coagulation Markers in Elderly Patients with COVID-19: Retrospective Analysis of Laboratory Results. Life (Basel) 2025; 15:370. [PMID: 40141715 DOI: 10.3390/life15030370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Elderly patients with COVID-19 often exhibit a complex interplay between hypercoagulability and coagulopathy, key factors in determining the risk of severe complications and mortality. This study aimed to analyze coagulation and inflammatory markers to identify critical predictors of adverse outcomes in this vulnerable population. Material and Methods: The retrospective study was conducted on a sample of 1429 elderly patients (≥60 years) diagnosed with COVID-19, hospitalized in "Sf. Ap. Andrei" St. Apostle Andrew's County Emergency Hospital in various wards between March 2020 and August 2022. Data were collected from medical records and included inflammatory markers (C-reactive protein, procalcitonin, ESR) and coagulation markers (prothrombin time, INR, fibrinogen, D-dimer). The SPSS 2.0 statistical software was used to conduct the study. Results:Coagulation markers: Prothrombin activity averaged 74.22%, below normal levels, indicating a heightened bleeding risk, while fibrinogen levels were significantly elevated (mean: 531.69 mg/dL), reflecting hypercoagulability. Prolonged prothrombin time (mean: 17.28 s) and elevated INR (International normalized ratio) (mean: 1.51) were associated with increased mortality, emphasizing their role in risk stratification. Elevated D-dimer levels (mean: 2.75 mg/L) further highlighted thromboembolic risks. Inflammatory markers: C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) showed marked elevations (mean CRP: 92.09 mg/L, mean ESR: 58.47 mm/h), correlating with heightened systemic inflammation and poor outcomes. Bacterial infections: Elevated procalcitonin (mean: 1.98 ng/mL) suggested secondary bacterial infections, particularly in mechanically ventilated patients, significantly worsening prognosis. Conclusions: The duality of hypercoagulability and coagulopathy in elderly COVID-19 patients underscores the importance of consistently monitoring coagulation markers such as prothrombin time, INR, D-dimer, and fibrinogen. Simultaneously, elevated inflammatory markers and secondary bacterial infections require prompt therapeutic interventions. This study highlights the critical need for personalized management strategies to mitigate complications and reduce mortality in this high-risk population.
Collapse
Affiliation(s)
- Corina Popazu
- Clinical-Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Str. Domnească 35, 800201 Galaţi, Romania
| | - Aurelia Romila
- Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Str. Domnească 35, 800201 Galaţi, Romania
| | - Marius Petrea
- Pre-Clinical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Str. Domnească 35, 800201 Galaţi, Romania
| | - Robert Marius Grosu
- Pre-Clinical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Str. Domnească 35, 800201 Galaţi, Romania
| | - Alina-Maria Lescai
- Clinical-Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Str. Domnească 35, 800201 Galaţi, Romania
| | - Adriana Liliana Vlad
- Clinical-Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Str. Domnească 35, 800201 Galaţi, Romania
| | - Violeta Diana Oprea
- Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Str. Domnească 35, 800201 Galaţi, Romania
| | - Alexia Anastasia Ștefania Baltă
- Clinical-Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Str. Domnească 35, 800201 Galaţi, Romania
| |
Collapse
|
49
|
Watase M, Shiraishi Y, Chubachi S, Tanabe N, Maetani T, Asakura T, Namkoong H, Tanaka H, Shimada T, Azekawa S, Otake S, Fukushima T, Nakagawara K, Masaki K, Terai H, Mochimaru T, Sasaki M, Ueda S, Kato Y, Harada N, Suzuki S, Yoshida S, Tateno H, Yamada Y, Jinzaki M, Okada Y, Koike R, Ishii M, Kimura A, Imoto S, Miyano S, Ogawa S, Kanai T, Fukunaga K. Coronary Artery Calcification on Chest Computed Tomography as a Predictor of Cardiovascular Adverse Events in Patients With COVID-19 - A Multicenter Retrospective Study in Japan. Circ J 2025; 89:373-381. [PMID: 39828330 DOI: 10.1253/circj.cj-24-0661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
BACKGROUND Coronary artery calcification (CAC) detected through chest computed tomography (CT) strongly predicts cardiovascular events in asymptomatic individuals undergoing primary prevention. Few studies with limited sample sizes have investigated the predictive value of CAC for cardiovascular complications in COVID-19. This study examined the impact of CAC on cardiovascular complications using a large-scale COVID-19 database. METHODS AND RESULTS This multicenter retrospective cohort study used data from the Japan COVID-19 Task Force database. After exclusion based on missing information, 1,109 patients with COVID-19 were included. The Agatston score was used to evaluate CAC, dividing the population into 3 groups based on calcification degree (no, moderate, and severe CAC). The primary outcome was cardiovascular complications; the secondary outcome was critical outcomes. The severe CAC group had a higher rate of cardiovascular complications than the other groups. Multivariable analysis, considering COVID-19 severity factors, identified severe CAC as independently associated with cardiovascular complications but not with critical outcomes. Subgroup analysis revealed that, in patients without hypertension, diabetes, cardiovascular disease, or chronic kidney disease, severe CAC was significantly correlated with cardiovascular complications, whereas this association was not observed in patients with these underlying conditions. CONCLUSIONS Patients with COVID-19 and severe CAC had increased cardiovascular complications, and identifying cardiovascular and pulmonary findings on chest CT is essential. Measuring CAC via non-electrocardiogram-gated CT helps predict patient risk.
Collapse
Affiliation(s)
- Mayuko Watase
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine
- Department of Respiratory Medicine, National Hospital Organization, Tokyo Medical Center
| | - Yusuke Shiraishi
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University
| | - Shotaro Chubachi
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine
| | - Naoya Tanabe
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University
| | - Tomoki Maetani
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University
| | - Takanori Asakura
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine
- Laboratory of Bioregulatory Medicine, Department of Clinical Medicine, Kitasato University School of Pharmacy
- Department of Respiratory Medicine, Kitasato University, Kitasato Institute Hospital
| | - Ho Namkoong
- Department of Infectious Diseases, Keio University School of Medicine
| | - Hiromu Tanaka
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine
| | - Takashi Shimada
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine
| | - Shuhei Azekawa
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine
| | - Shiro Otake
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine
| | - Takahiro Fukushima
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine
| | - Kensuke Nakagawara
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine
| | - Katsunori Masaki
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine
| | - Hideki Terai
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine
| | - Takao Mochimaru
- Department of Respiratory Medicine, National Hospital Organization, Tokyo Medical Center
| | - Mamoru Sasaki
- Department of Respiratory Medicine, Japan Community Health Care Organization (JCHO), Saitama Medical Center
| | - Soichiro Ueda
- Department of Respiratory Medicine, Japan Community Health Care Organization (JCHO), Saitama Medical Center
| | - Yukari Kato
- Department of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine
| | - Norihiro Harada
- Department of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine
| | - Shoji Suzuki
- Department of Pulmonary Medicine, Saitama City Hospital
| | | | - Hiroki Tateno
- Department of Pulmonary Medicine, Saitama City Hospital
| | | | | | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine
- Department of Genome Informatics, Graduate School of Medicine, University of Tokyo
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences
| | - Ryuji Koike
- Health Science Research and Development Center (HeRD), Tokyo Medical and Dental University
| | - Makoto Ishii
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine
| | - Akinori Kimura
- Institute of Research, Tokyo Medical and Dental University
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo
| | - Satoru Miyano
- M&D Data Science Center, Tokyo Medical and Dental University
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Internal Medicine, Keio University School of Medicine
| |
Collapse
|
50
|
Thaweethai T, Selvaggi CA, Ng TC, Cheng D, Cao T, Chibnik LB, Shinnick DJ, Foulkes AS. Biomarker states and risk of death among individuals hospitalized with SARS-CoV-2 infection. BMC Infect Dis 2025; 25:260. [PMID: 39994606 PMCID: PMC11849268 DOI: 10.1186/s12879-025-10651-w] [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: 10/24/2023] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Identifying individuals hospitalized for SARS-CoV-2 infection at increased risk of death is crucial for clinical decision making. Analyses must consider simultaneously the multitude of biomarkers across several domains and how these biomarker profiles change over time. METHODS This electronic health records-based study included individuals hospitalized at a Massachusetts General Brigham hospital for at least 24 h within 5 days prior and 30 days after diagnosis of COVID-19. K-means clustering was used to identify profiles among 20 eligible biomarkers and proportional hazards models were used to model 30-day mortality at hospitalization and 7 days after hospitalization (i.e., landmark models). RESULTS Twelve thousand, nine hundred forty-two individuals were included, among whom 1,198 died within 30 days. Six states were identified, characterized by the following abnormalities: (1) normal/reference, (2) hematologic, (3) inflammatory and hematological, (4) metabolic, (5) kidney, hematologic, and metabolic, and (6) cardio-thrombotic, liver, and metabolic. Risk of death within 30 days was higher in States 3, 4, 5, and 6 (adjusted hazard ratios ranging from 3.6 to 7.8) compared to individuals in State 1 at hospitalization. Landmark model findings were similar. CONCLUSIONS Distinct sub-phenotypes based on biomarker profiles were identified among patients hospitalized with SARS-CoV-2 infection, and certain phenotypes are associated with greater risk of 30-day mortality.
Collapse
Affiliation(s)
- Tanayott Thaweethai
- Massachusetts General Hospital Biostatistics, 399 Revolution Drive Ste 1068, Somerville, MA, 02145, USA.
- Department of Medicine, Harvard Medical School, 25 Shattuck St., Boston, MA, 02115, USA.
| | - Caitlin A Selvaggi
- Massachusetts General Hospital Biostatistics, 399 Revolution Drive Ste 1068, Somerville, MA, 02145, USA
| | - Ta-Chou Ng
- Massachusetts General Hospital Biostatistics, 399 Revolution Drive Ste 1068, Somerville, MA, 02145, USA
| | - David Cheng
- Massachusetts General Hospital Biostatistics, 399 Revolution Drive Ste 1068, Somerville, MA, 02145, USA
- Department of Medicine, Harvard Medical School, 25 Shattuck St., Boston, MA, 02115, USA
| | - Tingyi Cao
- Massachusetts General Hospital Biostatistics, 399 Revolution Drive Ste 1068, Somerville, MA, 02145, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Ave., Boston, MA, 02115, USA
| | - Lori B Chibnik
- Massachusetts General Hospital Biostatistics, 399 Revolution Drive Ste 1068, Somerville, MA, 02145, USA
- Department of Medicine, Harvard Medical School, 25 Shattuck St., Boston, MA, 02115, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave., Boston, MA, 02115, USA
| | - Daniel J Shinnick
- Massachusetts General Hospital Biostatistics, 399 Revolution Drive Ste 1068, Somerville, MA, 02145, USA
| | - Andrea S Foulkes
- Massachusetts General Hospital Biostatistics, 399 Revolution Drive Ste 1068, Somerville, MA, 02145, USA
- Department of Medicine, Harvard Medical School, 25 Shattuck St., Boston, MA, 02115, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 655 Huntington Ave., Boston, MA, 02115, USA
| |
Collapse
|