1
|
Kasem Ali Sliman R, Cohen H, Shehadeh S, Batcir R, Alter YE, Cohen K, Koren I, Halabi I, Sliman H, Saied MH. Pediatric autoimmune diseases in the light of COVID-19 pandemic, A retrospective observational big data study. J Transl Autoimmun 2025; 10:100281. [PMID: 40162434 PMCID: PMC11951201 DOI: 10.1016/j.jtauto.2025.100281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/18/2025] [Accepted: 03/01/2025] [Indexed: 04/02/2025] Open
Abstract
Background The COVID-19 pandemic has raised concerns about potential links between SARS-CoV-2 infection and autoimmune diseases. This study investigated changes in the incidence rate (IR) of autoimmune diseases among children following the pandemic's onset. Methods A retrospective cross-sectional study analyzed data from Clalit Health Services, Israel's largest healthcare provider, examining the IR of different autoimmune diseases in children aged 0-18. The study compared pre-pandemic (2019) with pandemic/post-pandemic periods (2020-2023), encompassing a cohort of over 1.5 million children. Results Significant IR increases were observed across multiple autoimmune diseases. Rheumatic diseases (Juvenile Idiopathic Arthritis, Systemic Lupus Erythematosus, Henoch Schoenlein Purpura (HSP)) showed consistent increases, with HSP demonstrating the most pronounced trend. Endocrine disorders exhibited diverse patterns, with autoimmune thyroid diseases and Type 1 diabetes showing overall increases, while diabetic ketoacidosis exhibited an initial spike followed by a decline. Gastrointestinal diseases displayed heterogeneous patterns; Celiac disease and Ulcerative colitis showed general increases, Crohn's disease showed a downward trend, and autoimmune hepatitis exhibited an initial significant decrease followed by a significant increase. Dermatological conditions, including Psoriasis and Vitiligo, demonstrated consistent elevations throughout 2020-2023. Immune Thrombocytopenia Purpura showed initial decreases followed by significant increases in 2022-2023. Conclusions This comprehensive analysis reveals significant changes in pediatric autoimmune disease incidence following the COVID-19 pandemic, suggesting potential associations between SARS-CoV-2 infection and autoimmune dysregulation. The diverse patterns observed across different conditions highlight the complex interplay between viral infection and autoimmunity, emphasizing the need for continued surveillance and investigation of long-term immunological consequences of COVID-19 in pediatric populations.
Collapse
Affiliation(s)
- Rim Kasem Ali Sliman
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
| | - Hilla Cohen
- Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
| | - Shereen Shehadeh
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Infectious Disease Unit, Carmel Medical Center, Haifa, Israel
| | - Reut Batcir
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Gastroenterology Unit, Carmel Medical Center, Haifa, Israel
| | - Yigal Elenberg Alter
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Gastroenterology Unit, Carmel Medical Center, Haifa, Israel
| | - Keren Cohen
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Endocrine Unit, Carmel Medical Center, Haifa, Israel
| | - Ilana Koren
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Endocrine Unit, Carmel Medical Center, Haifa, Israel
| | - Inbal Halabi
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Pediatric Endocrine Unit, Carmel Medical Center, Haifa, Israel
| | - Hussein Sliman
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Cardiology, Carmel Medical Center, Heart Center, Haifa, Israel
| | - Mohamad Hamad Saied
- Technion Israel Institute of Technology, Rappaport Faculty of Medicine, Haifa 3109601, Israel
- Department of Pediatrics, Clalit Health Care Organization, Carmel Medical Center, Haifa, Israel
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital/University Medical Center, Utrecht, the Netherlands
| |
Collapse
|
2
|
Yang L, Zhou X, Liu J, Yang G, Yu J, Tan W, Fang X, Li W, He J, Ma Q, Yu L, Lu Z. Liang-Ge-San attenuates virus-induced acute lung injury by targeting FXR-mediated ACE2 downregulation to modulate the formation of the cytokine storm. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156584. [PMID: 40056637 DOI: 10.1016/j.phymed.2025.156584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Traditional Chinese medicine has been recognized for its significant role in treating acute lung injury (ALI) due to its distinct therapeutic advantages. Liang-Ge-San (LGS), a formulation from the ancient "Taiping Huimin Hejiju Fang", is believed to possess beneficial effects for treating ALI. However, LGS's precise mechanisms and efficacy in addressing viral ALI remain inadequately explored. PURPOSE To evaluate LGS's therapeutic effects and underlying mechanisms in treating viral-induced ALI. METHODS The protective effects of LGS were examined in a Polyinosinic-polycytidylic acid [Poly(I:C)]-induced ALI model using real-time quantitative PCR, enzyme-linked immunosorbent assay, and histopathological analysis. A bioinformatics approach combined with network pharmacology was utilized to ascertain the key targets of LGS in viral pneumonia. The pharmacodynamic mechanisms of LGS in viral ALI were further validated through immunofluorescence, overexpression, short hairpin RNA, chromatin immunoprecipitation, and target agonist assays. RESULTS LGS administration resulted in a reduction of IL-1β, IL-6, and TNF-α levels, along with a decrease in macrophage infiltration, pulmonary damage, and pneumonedema following the Poly(I:C) challenge. Bioinformatics and network pharmacology analyses suggested that Farnesyl X receptor (FXR) and angiotensin converting enzyme 2 (ACE2) are potential therapeutic targets for LGS in viral pneumonia. Further experiments revealed that LGS suppressed the expression of FXR, ACE2, and NF-κB-p65 in Poly(I:C)-infected cells. Notably, overexpression of FXR counteracted the repressive effects of LGS, while ACE2 expression remained unchanged in FXR-knockdown RAW264.7 cells upon treatment with Poly(I:C) or LGS. Additionally, LGS inhibited the interaction between FXR and ACE2 transcriptional promoters. In vivo, LGS attenuated the Poly(I:C)-induced upregulation of FXR, ACE2, IL-1β, IL-6, and TNF-α in ALI zebrafish and mice models, effects that could be reversed by chenodeoxycholic acid (CDCA), an FXR agonist. Moreover, LGS markedly alleviated weight loss, improved survival rates, reduced lung index, diminished viral load, and inhibited lung pathological changes in H1N1-PR8-induced ALI mice. IL-1β, IL-6, TNF-α, INF-γ, FXR, ACE2, small heterodimer partner, and NF-κB-p65 levels were markedly reduced by LGS, with these effects being reversed by CDCA. CONCLUSION This investigation provides the first evidence that FXR/ACE2 signaling is pivotal in acute respiratory viral infections, while LGS demonstrates antiviral activity against viral-induced ALI. LGS inhibits ACE2 expression induced by viral infection via FXR inhibition and modulates the cytokine storm, thus alleviating viral ALI. These findings suggest that LGS may be a promising treatment strategy for treating viral ALI.
Collapse
Affiliation(s)
- Liling Yang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Xiangjun Zhou
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan 523808, PR China
| | - Junshan Liu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Guangli Yang
- Department of Central Laboratory, The Binhaiwan Central Hospital of Dongguan, Dongguan 523808, PR China
| | - Jingtao Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Weifu Tan
- Department of Neonatology, The Binhaiwan Central Hospital of Dongguan, Dongguan 523808, PR China
| | - Xiaochuan Fang
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China
| | - Wei Li
- Department of Neonatology, The Binhaiwan Central Hospital of Dongguan, Dongguan 523808, PR China
| | - Jiayang He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510030, PR China
| | - Qinhai Ma
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510030, PR China.
| | - Linzhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China.
| | - Zibin Lu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
3
|
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
|
4
|
Eltobgy M, Klamer B, Farkas D, Londino JD, Englert JA, Horowitz JC, Mallampalli RK, Brock G, Bednash JS. Plasma proteomic profiles correlate with organ dysfunction in COVID-19 ARDS. Physiol Rep 2025; 13:e70300. [PMID: 40170544 PMCID: PMC11962209 DOI: 10.14814/phy2.70300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 03/18/2025] [Indexed: 04/03/2025] Open
Abstract
Severe COVID-19 is often complicated by hypoxemic respiratory failure and acute respiratory distress syndrome (ARDS). Mechanisms governing lung injury and repair in ARDS remain poorly understood. We hypothesized that plasma proteomics may uncover protein biomarkers correlated with COVID-19 ARDS severity. We analyzed the plasma proteome from 32 patients with ARDS and COVID-19 using an aptamer-based platform of 7289 proteins, and correlated protein measurements with sequential organ failure assessment (SOFA) scores at days 1 and 7 of ICU admission. We identified 184 differentially abundant proteins correlated with SOFA at day 1 and 46 proteins at day 7. In a longitudinal analysis, we correlated dynamic changes in protein abundance and SOFA between days 1 and 7 and identified 40 significant proteins. Pathway analysis of significant proteins identified increased ephrin signaling and acute phase response signaling correlated with increased SOFA scores between days 1 and 7, while pathways related to pulmonary fibrosis signaling and wound healing had a negative correlation. These findings suggest that persistent inflammation may drive disease severity, while repair processes correlate with improvements in organ dysfunction. This approach is generalizable to future ARDS cohorts for identification of biomarkers and disease mechanisms as we strive towards targeted therapies in ARDS.
Collapse
Grants
- K08HL169725 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL142767 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL141195 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01HL114453 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL097376 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL081784 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL096376 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- UM1TR004548 HHS | NIH | National Center for Advancing Translational Sciences (NCATS)
- OSU | College of Medicine Office of Research, Ohio State University (COMOR)
- HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HHS | NIH | National Center for Advancing Translational Sciences (NCATS)
- OSU | College of Medicine Office of Research, Ohio State University (COMOR)
Collapse
Affiliation(s)
- Moemen Eltobgy
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep MedicineThe Ohio State UniversityColumbusOhioUSA
- Dorothy M. Davis Heart and Lung Research Institute (DHLRI), College of Medicine, The Ohio State UniversityColumbusOhioUSA
| | - Brett Klamer
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOhioUSA
| | - Daniela Farkas
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep MedicineThe Ohio State UniversityColumbusOhioUSA
- Dorothy M. Davis Heart and Lung Research Institute (DHLRI), College of Medicine, The Ohio State UniversityColumbusOhioUSA
| | - James D. Londino
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep MedicineThe Ohio State UniversityColumbusOhioUSA
- Dorothy M. Davis Heart and Lung Research Institute (DHLRI), College of Medicine, The Ohio State UniversityColumbusOhioUSA
- The Center for RNA BiologyCollege of Medicine, the Ohio State UniversityColumbusOhioUSA
| | - Joshua A. Englert
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep MedicineThe Ohio State UniversityColumbusOhioUSA
- Dorothy M. Davis Heart and Lung Research Institute (DHLRI), College of Medicine, The Ohio State UniversityColumbusOhioUSA
| | - Jeffrey C. Horowitz
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep MedicineThe Ohio State UniversityColumbusOhioUSA
- Dorothy M. Davis Heart and Lung Research Institute (DHLRI), College of Medicine, The Ohio State UniversityColumbusOhioUSA
| | - Rama K. Mallampalli
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep MedicineThe Ohio State UniversityColumbusOhioUSA
- Dorothy M. Davis Heart and Lung Research Institute (DHLRI), College of Medicine, The Ohio State UniversityColumbusOhioUSA
| | - Guy Brock
- Department of Biomedical InformaticsThe Ohio State UniversityColumbusOhioUSA
| | - Joseph S. Bednash
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep MedicineThe Ohio State UniversityColumbusOhioUSA
- Dorothy M. Davis Heart and Lung Research Institute (DHLRI), College of Medicine, The Ohio State UniversityColumbusOhioUSA
- The Center for RNA BiologyCollege of Medicine, the Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
5
|
Narin Çopur E, Ergün D, Ergün R, Atik S, Türk Dağı H, Körez MK. Risk Factors Affecting the Severity, Mortality, and Intensive Care Unit Admission of COVID-19 Patients: A Series of 1075 Cases. Viruses 2025; 17:429. [PMID: 40143356 PMCID: PMC11946003 DOI: 10.3390/v17030429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND The clinical spectrum of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is broad; it can range from asymptomatic cases to mild upper respiratory tract illness, respiratory failure, and severe multiorgan failure resulting in death. Therefore, it is important to identify the clinical course of the disease and the factors associated with mortality. OBJECTIVE The aim of this study is to identify the risk factors associated with the severity of the disease, intensive care unit admission, and mortality in COVID-19 patients. METHODS A total of 1075 patients with clinical and radiological findings compatible with COVID-19 pneumonia and positive SARS-CoV-2 PCR were selected and retrospectively screened. All included patients were classified according to the 7th edition of the 2019 Coronavirus Disease Guidelines published by the National Health Commission of China. RESULTS It was observed that elevated white blood count (WBC) increased the severity of COVID-19 by 3.26 times and the risk of intensive care unit (ICU) admission by 3.47 times. Patients with high D-dimer levels had a 91% increased risk, and those with high fibrinogen levels had a 2.08 times higher risk of severe disease. High C-reactive protein (CRP) values were found to increase disease severity by 6.89 times, mortality by 12.84 times, and ICU admission by 3.37 times. CONCLUSIONS Identifying the factors associated with disease severity, ICU admission, and mortality in COVID-19 patients could help reduce disability and mortality rates in pandemics.
Collapse
Affiliation(s)
- Ecem Narin Çopur
- Department of Pulmonary Medicine, Dr. Yaşar Eryılmaz Doğubeyazıt State Hospital, Ağrı 04402, Turkey
| | - Dilek Ergün
- Department of Pulmonary Medicine, Faculty of Medicine, Selcuk University, Konya 42130, Turkey;
| | - Recai Ergün
- Department of Pulmonary Medicine, Faculty of Medicine, Selcuk University, Konya 42130, Turkey;
| | - Serap Atik
- Department of Pulmonary Medicine, Iğdır Dr. Nevruz Erez State Hospital, Iğdır 76000, Turkey;
| | - Hatice Türk Dağı
- Department of Medical Microbiology, Faculty of Medicine, Selcuk University, Konya 42130, Turkey;
| | - Muslu Kazım Körez
- Department of Biostatistics, Faculty of Medicine, Selcuk University, Konya 42130, Turkey;
| |
Collapse
|
6
|
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
|
7
|
Wen Y, Zhao J, Zhang Z. Heterogeneity and longitudinal transcriptomic characteristics of Tregs in COVID-19 patients. Front Immunol 2025; 16:1548173. [PMID: 40114921 PMCID: PMC11922936 DOI: 10.3389/fimmu.2025.1548173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction Regulatory T cells (Tregs) play a crucial role in maintaining immune tolerance by suppressing immune responses against pathogens. The fluctuation of Treg proportions in COVID-19 remains a topic of debate, and the mechanisms triggering Treg activation in COVID-19 are still unclear. Understanding these issues is essential for better managing immune responses in COVID-19 patients. Methods We collected a cohort of COVID-19 patients with varying disease severity and stage to explore the transcriptomic and functional traits of Tregs in these individuals. Using transcriptomic analysis, we evaluated the proportion and functionality of different Treg subsets, specifically HLA_DR+ Tregs, across different stages of COVID-19 patients. Results Our analysis revealed that the proportion of CCR7 + Tregs decreased as the disease advanced, while the cell proportion of HLA_DR+ regs escalated with the severity of the disease. Moreover, the transcription actor CARHSP1 exhibited apositive correlation with the proportion of HLA_DR+ Tregs. Notably, the heightened suppressive function of HLA_DR+ Tregs in severe COVID-19 patients, with interactions between PF4 and CXCR3, contributed to the homeostasis of HLA_DR+ Tregs in severe COVID-19 patients. Furthermore, we observed that Tregs in COVID-19 patients exhibited weakened TCR clonotype expansion, and the suppression of HLA_DR+ Tregs with expanded TCR clonotypes in severe COVID-19 cases did not show a significant increase compared to asymptomatic and mild COVID-19 groups. The findings indicate that Tregs may be activated through the bystander effect, as evidenced by the analysis of TCR clonotype characteristics. Discussion Our research delineates the diversity of dynamic alterations in Tregs and sheds light on potential mechanisms underlying Treg activation, providing a theoretical foundation and offering treatment strategies for managing COVID-19 patients.
Collapse
Affiliation(s)
- Yanling Wen
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Juanjuan Zhao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
8
|
Hwang Y, Kang SJ, Kang J, Choi J, Kim SJ, Jang S. DNA repair and disease: insights from the human DNA glycosylase NEIL family. Exp Mol Med 2025; 57:524-532. [PMID: 40033009 PMCID: PMC11958798 DOI: 10.1038/s12276-025-01417-0] [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: 08/01/2024] [Revised: 11/27/2024] [Accepted: 12/11/2024] [Indexed: 03/05/2025] Open
Abstract
The base excision repair pathway protects DNA from base damage via oxidation, deamination, alkylation and methylation. DNA glycosylases are key enzymes that recognize damaged bases in a lesion-specific manner and initiate the base excision repair process. Among these, the endonuclease VIII-like 1-3 (NEIL1-3) family, which is found in mammalian genomes, is a homolog of bacterial DNA glycosylases known as Fpg/Nei. NEIL enzymes have similar structures and substrates but with slight differences. When repair proteins are impaired, the accumulation of damaged bases can lead to increased genomic instability, which is implicated in various pathologies, including cancer and neurodegeneration. Notably, mutations in these proteins also influence a range of other diseases and inflammation. This review focuses on the influence of the NEIL family on human health across different organ systems. Investigating the relationship between NEIL mutations and diseases can improve our understanding of how these enzymes affect the human body. This information is crucial for understanding the basic mechanisms of DNA repair and enabling the development of novel inhibitors or gene therapies that target only these enzymes. Understanding the role of the NEIL family provides insights into novel therapies and improves our ability to combat genetic diseases.
Collapse
Affiliation(s)
- Yuna Hwang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Su-Jin Kang
- College of Pharmacy, Dongduk Women's University, Seoul, Republic of Korea
| | - Jieun Kang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Jeongwoo Choi
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Seung-Jin Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Republic of Korea.
| | - Sunbok Jang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, Republic of Korea.
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Fatima U, Ahmed H, Singh G, Giri K, Azmi MS, Meenakshi A, Jandrajupalli SB, Chandolu S, Nayyar AS. Non-Neoformans Cryptococcal Infections in the Post-Coronavirus Disease-19 (COVID-19) Era: Are We Ready to Face the Emerging Challenge? Asian J Neurosurg 2025; 20:190-195. [PMID: 40041573 PMCID: PMC11875713 DOI: 10.1055/s-0044-1791998] [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] [Indexed: 03/06/2025] Open
Abstract
Coronaviruses are a large group of viruses that infect animals as well as humans, while it is also suggested that, rarely, coronaviruses that infect animals can evolve and infect humans. Current evidence suggests that severe acute respiratory syndrome (SARS) coronavirus-2 leads to coronavirus disease-19 (COVID-19), the respiratory illness responsible for COVID-19 pandemic, while it has a zoonotic origin, closely related to the bat-origin SARS-like coronavirus. Also, as per the current knowledge, the disease may induce significant and persistent lymphopenia which in turn may increase the risk for various opportunistic infections. Cryptococcus laurentii is one such rare, but serious fungal infection which has been reported in post-COVID-19 disease and is a rising cause of concern since it can turn out to be fatal. The infection is caused by a non-neoformans rare human pathogen. The present case report describes the case of a 45-years old male patient who reported to the Outpatient Department (OPD) for a routine dental complaint with a grossly destructed tooth in left lower back tooth region due to extensive carious involvement, while, simultaneously, presenting with fever since 5 days in the post-COVID-19 phase, and was later diagnosed as being positive for C. laurentii infection on urine culture sensitivity test.
Collapse
Affiliation(s)
- Umayra Fatima
- Department of Dentistry, Princess Esra Hospital, Deccan College of Medical Sciences, Hyderabad, Telangana, India
| | - Hina Ahmed
- Department of Conservative Dental Sciences, Ibn Sina National College for Medical Studies, Jeddah, Kingdom of Saudi Arabia
| | - Gautam Singh
- Department of Conservative Dental Sciences, Ibn Sina National College for Medical Studies, Jeddah, Kingdom of Saudi Arabia
| | - K.Y. Giri
- Department of Oral and Maxillofacial Surgery, Drs Sudha and Nageswara Rao Siddhartha Institute of Dental Sciences, Gannavaram, Andhra Pradesh, India
| | - Md. Sania Azmi
- Department of Oral and Maxillofacial Pathology, Care Dental College, Guntur, Andhra Pradesh, India
| | - Archana Meenakshi
- Department of Periodontology, Ragas Dental College and Hospital, Chennai, Tamil Nadu, India
| | - Suresh Babu Jandrajupalli
- Division of Periodontology, Department of Preventive Dental Sciences, College of Dentistry, University of Ha'il, Ha'il, Kingdom of Saudi Arabia
| | - Swarnalatha Chandolu
- Division of Periodontology, Department of Preventive Dental Sciences, College of Dentistry, University of Ha'il, Ha'il, Kingdom of Saudi Arabia
| | - Abhishek Singh Nayyar
- Department of Oral Medicine and Radiology, Saraswati Dhanwantari Dental College and Hospital and Post-graduate Research Institute, Parbhani, Maharashtra, India
| |
Collapse
|
10
|
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
|
11
|
Li G, Hao Z, Wang H, Wang C, Liu D, Chen L, Sun Y. Pharmacological mechanism of action of Lianhua Qingwen in the treatment of COVID-19 and facial neuritis. World J Otorhinolaryngol Head Neck Surg 2025; 11:102-115. [PMID: 40070503 PMCID: PMC11891286 DOI: 10.1002/wjo2.185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/13/2024] [Accepted: 04/10/2024] [Indexed: 03/14/2025] Open
Abstract
Objective Coronavirus disease-2019 (COVID-19) can cause not only respiratory symptoms but also facial paralysis. Lianhua Qingwen (LHQW) has been reported to have therapeutic effects on COVID-19 and facial neuritis (FN). We explored the potential mechanism of LHQW in the treatment of COVID-19 and FN through a network-pharmacology approach. Methods Active compounds and relevant targets of LHQW were obtained from the databases of Traditional Chinese Medicine Systems Pharmacology Database, HERB, UniProt Knowledge Base, SwissADME, and Swiss Target Prediction. Disease targets of COVID-19 and FN were acquired from Gene Cards. Database For Annotation, Visualization And Integrated Discovery and Metascape were used to search the biological functions of intersecting targets. After identifying the core targets and their corresponding ingredients, KEGG Mapper analyzes the localization of core targets in key pathways. AutoDock were employed to conduct molecular docking of the core targets and their corresponding ingredients. Results We obtained four core genes: interleukin (IL)-8, IL-1B, IL-6, and tumor necrosis factor (TNF)-α. Database searching revealed the anti-inflammatory and antiviral effects of LHQW may be related to the action of aleo-emodin, hyperforin, kaempferol, luteolin, and quercetin on these four genes by regulating the pathways of IL-17 and NOD-like receptor. The molecular-docking results of the four core targets and their corresponding active ingredients showed good binding activity between receptors and ligands. Conclusions We uncovered the active ingredients, potential targets, and biological pathways of LHQW for COVID-19 and FN coinfection. Our data provide a theoretical basis for further exploration of the mechanism of action of LHQW in treatment of COVID-19 and FN.
Collapse
Affiliation(s)
- Guang‐Jin Li
- Department of OtorhinolaryngologyThe Second Affiliated Hospital of Guilin Medical UniversityGuilinGuangxiChina
- Department of Otorhinolaryngology Head and Neck SurgeryThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiShandongChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiShandongChina
| | - Zhi‐Hong Hao
- Department of Otorhinolaryngology Head and Neck SurgeryThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiShandongChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiShandongChina
- School of Clinical MedicineShandong Second Medical University (Weifang Medical University)WeifangShandongChina
| | - Han‐Jing Wang
- Department of Otorhinolaryngology Head and Neck SurgeryThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiShandongChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiShandongChina
- School of Clinical MedicineShandong Second Medical University (Weifang Medical University)WeifangShandongChina
| | - Chen Wang
- Department of Otorhinolaryngology Head and Neck SurgeryThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiShandongChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiShandongChina
- School of Clinical MedicineShandong Second Medical University (Weifang Medical University)WeifangShandongChina
| | - Da‐Wei Liu
- Department of Otorhinolaryngology Head and Neck SurgeryThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiShandongChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiShandongChina
| | - Liang Chen
- Department of Otorhinolaryngology Head and Neck SurgeryThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiShandongChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiShandongChina
| | - Yan Sun
- Department of Otorhinolaryngology Head and Neck SurgeryThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiShandongChina
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic DiseasesYantaiShandongChina
| |
Collapse
|
12
|
Augustus AR, Radhakrishnan Y, Bhaskar JP, Ramamurthi S, Shunmugiah KP. Tannic acid modulates SARS-CoV-2 pathogenesis by curbing key host receptors and oxidative stress. Toxicol In Vitro 2025; 103:105971. [PMID: 39551113 DOI: 10.1016/j.tiv.2024.105971] [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: 01/04/2024] [Revised: 10/23/2024] [Accepted: 11/12/2024] [Indexed: 11/19/2024]
Abstract
The novel coronavirus SARS-CoV-2, which wrecked havoc around the world in the recent years through COVID-19, gains entry into the host cell through various receptors. Development of therapies targeting host-pathogen interaction will be a key to curb the infection as it potentially suppresses viral attachment and entry into the host. Boundless bioactives abundant in natural resources are the important source of new as well as safer alternatives. Tannic acid, a polyphenolic compound found abundantly in various plant sources, has gained much attention owing to its multifaceted pharmacological properties. This research paper presents a comprehensive investigation on antioxidant, anti-inflammatory and anti-viral abilities of tannic acid, substantiated through a triad of methodologies: in silico, in vitro and in vivo approaches. In vitro experiments, confirmed the antioxidant and anti-inflammatory efficacy as well as the host receptor modulating potential of tannic acid. In silico docking analyses elucidated the molecular interactions between tannic acid and key host receptors involved in inflammation and viral pathogenesis. Furthermore, the in vivo studies involving Danio rerio provided a holistic understanding of the systemic impact of tannic acid, including its antioxidant effects by mitigating the oxidative stress.
Collapse
Affiliation(s)
- Akshaya Rani Augustus
- Department of Biotechnology, Alagappa University, Karaikudi 630 003, Tamil Nadu, India
| | | | | | | | | |
Collapse
|
13
|
Jiang RD, Luo YZ, Lin HF, Zheng XS, Zeng WT, Liu MQ, Deng HH, Wang Q, Lai YN, Chen Y, Guo ZS, Zeng Y, Gong QC, Qiu C, Dong M, Wang X, Wang ZY, Ji LN, Hou PP, Li Q, Shen XR, Li B, Gao Y, Zhang AH, Jiang TT, Shi AM, Zhou P, Lin XH, Deng ZQ, Li JM, Shi ZL. Impaired inflammatory resolution with severe SARS-CoV-2 infection in leptin knock out obese hamster. iScience 2025; 28:111837. [PMID: 39981511 PMCID: PMC11841202 DOI: 10.1016/j.isci.2025.111837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/28/2024] [Accepted: 12/13/2024] [Indexed: 02/22/2025] Open
Abstract
Comorbidities, such as obesity, increase the risk of severe COVID-19. However, the mechanisms underlying severe illnesses in individuals with obesity are poorly understood. Here, we used gene-edited leptin knock out (Leptin -/-) obese hamsters to establish a severe infection model. This model exhibits robust viral replication, severe lung lesions, pronounced clinical symptoms, and fatal infection, mirroring severe COVID-19 in patients with obesity. Using single-cell transcriptomics on lung tissues pre- and post-infection, we found that monocyte-derived alveolar macrophages (MD-AM) play a key role in lung hyper-inflammation, including two unique MD-AM cell fate branches specific to Leptin -/- hamsters. Notably, reduced Trem2-dependent efferocytosis pathways in Leptin -/- hamsters indicated weakened inflammation resolution, consistent with the scRNA-seq data from patients with obesity. In summary, our study highlights the obesity-associated mechanisms underlying severe SARS-CoV-2 infections and establishes a reliable preclinical animal model for developing obesity-specific therapeutics for critical COVID-19.
Collapse
Affiliation(s)
- Ren-Di Jiang
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun-Zhe Luo
- BGI Research, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hao-Feng Lin
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Shuang Zheng
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Wen-Tao Zeng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Mei-Qin Liu
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Hao-Hao Deng
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Qi Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ya-Na Lai
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ying Chen
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Zi-Shuo Guo
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ya Zeng
- BGI Research, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qian-Chun Gong
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Qiu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Mei Dong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Xi Wang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zi-Yi Wang
- National Engineering Research Center of Neuromodulation, School of Aerospace Engineering, Tsinghua University, Beijing, China
| | - Li-Na Ji
- School of Life Sciences, Inner Mongolia University, Hohhot, China
- Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, China
| | - Pan-Pan Hou
- Guangzhou National Laboratory, Guangzhou, China
| | - Qian Li
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Xu-Rui Shen
- Guangzhou National Laboratory, Guangzhou, China
| | - Bei Li
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yun Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ai-Hua Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Ting-Ting Jiang
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ai-Min Shi
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Peng Zhou
- The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Xin-Hua Lin
- State Key Laboratory of Genetic Engineering, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
- School of Life Sciences, Inner Mongolia University, Hohhot, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, China
- Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, China
| | - Zi-Qing Deng
- BGI Research, Beijing, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI-Shenzhen, Shenzhen, China
| | - Jian-Min Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Jiangsu Animal Experimental Center of Medicine and Pharmacy, Animal Core facility, Key Laboratory of Model Animal, Department of Cell Biology, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zheng-Li Shi
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
- Guangzhou National Laboratory, Guangzhou, China
| |
Collapse
|
14
|
Yasmin S, Ansari MY. A detailed examination of coronavirus disease 2019 (COVID-19): Covering past and future perspectives. Microb Pathog 2025; 203:107398. [PMID: 39986548 DOI: 10.1016/j.micpath.2025.107398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 01/07/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
The COVID-19 disease has spread rapidly across the world within just six months, affecting 169 million people and causing 3.5 million deaths globally (2021). The most affected countries include the USA, Brazil, India, and several European countries such as the UK and Russia. Healthcare professionals face new challenges in finding better ways to manage patients and save lives. In this regard, more comprehensive research is needed, including genomic and proteomic studies, personalized medicines and the design of suitable treatments. However, finding novel molecular entities (NME) using a standard or de novo strategy to drug development is a time-consuming and costly process. Another alternate strategy is discovering new therapeutic uses for old/existing/available medications, known as drug repurposing. There are a variety of computational repurposing methodologies, and some of them have been used to counter the coronavirus disease pandemic of 2019 (COVID-19). This review article compiles recently published data on the origin, transmission, pathogenesis, diagnosis, and management of the coronavirus by drug repurposing and vaccine development approach. We have attempted to screen probable drugs in clinical trials by using literature survey. This systematic review aims to create priorities for future research of drugs repurposed and vaccine development for COVID-19.
Collapse
Affiliation(s)
- Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, Saudi Arabia.
| | - Mohammad Yousuf Ansari
- MM College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, 133207, India; Ibne Seena College of Pharmacy, Azmi Vidya Nagri Anjhi Shahabad, Hardoi, Uttar Pradesh (U.P.) 241124, India.
| |
Collapse
|
15
|
El Azhary K, Ghazi B, Kouhen F, El Bakkouri J, Chamlal H, El Ghanmi A, Badou A. Clinical Impact of Neutrophil Variation on COVID-19 Complications. Diagnostics (Basel) 2025; 15:457. [PMID: 40002608 PMCID: PMC11854688 DOI: 10.3390/diagnostics15040457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Corona virus disease 2019 (COVID-19) poses a threat to global public health. The early identification of critical cases is crucial to providing timely treatment to patients. Here, we investigated whether the neutrophil levels could predict COVID-19 complications. Methods: We performed a retrospective study of patients with COVID-19, admitted to the Cheikh Khalifa International University Hospital, Casablanca, Morocco. Laboratory test results collected upon admission and during hospitalization were analyzed based on clinical information. Results: Our study revealed that a rise in neutrophil "PNN" levels was associated with respiratory deterioration and intubation. They were positively correlated with the procalcitonin and C-reactive protein levels. Interestingly, PNN (polynuclear neutrophil) levels on day 5 proved to be a better predictor of intubation, acute respiratory distress syndrome (ARDS), and mortality than the initial PNN counts, C-reactive protein, or procalcitonin. Moreover, binary logistic regression with stratified PNN-day 5 data revealed that a PNN level on day 5 > 7.7 (109/L) was an independent risk factor for mortality and ARDS. Finally, the PNN levels on day 5 and proinflammatory cytokine IL-6 were positively correlated. Conclusions: Our data showed that neutrophilia proved to be an excellent predictor of complications and mortality during hospitalization and could be used to improve the management of patients with COVID-19.
Collapse
Affiliation(s)
- Khadija El Azhary
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca 20250, Morocco;
| | - Bouchra Ghazi
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco; (B.G.); (J.E.B.); (A.E.G.)
- Department of Gynecology and Obstetrics, Mohammed VI International University Hospital, Bouskoura 27182, Morocco
| | - Fadila Kouhen
- Laboratory of Neurosciences and Oncogenetics, Neurooncology and Oncogenetic Team, Faculty of Medicine, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco;
- Department of Radiotherapy, International University Hospital Cheikh Khalifa, Casablanca 82403, Morocco
| | - Jalila El Bakkouri
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco; (B.G.); (J.E.B.); (A.E.G.)
- Cheikh Khalifa International University Hospital, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco
| | - Hasna Chamlal
- Computer Science and Systems Laboratory (LIS), Faculty of Sciences Ain Chock, Hassan II University of Casablanca, Casablanca 20250, Morocco;
| | - Adil El Ghanmi
- Immunopathology-Immunotherapy-Immunomonitoring Laboratory, Faculty of Medicine, Mohammed VI University of Health and Sciences (UM6SS), Casablanca 82403, Morocco; (B.G.); (J.E.B.); (A.E.G.)
- Department of Gynecology and Obstetrics, Mohammed VI International University Hospital, Bouskoura 27182, Morocco
| | - Abdallah Badou
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy, Hassan II University of Casablanca, Casablanca 20250, Morocco;
| |
Collapse
|
16
|
Sezak N, Karaca B, Balik R, Aksun M. Prognostic Value of Neutrophil/Lymphocyte, Lymphocyte/C-reactive protein, Platelet/ Lymphocyte Rates in Covid-19 Cases Monitored in the Intensive Care Unit. Angiology 2025:33197251318094. [PMID: 39927475 DOI: 10.1177/00033197251318094] [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: 02/11/2025]
Abstract
Coronavirus 2019 (COVID-19) infection has a significant mortality rate. Despite the disease's extensive effects, little is known about the prognostic indicators that can be used. We aimed to assess the prognostic value of neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-C-reactive protein ratio (LCR) and platelet-to- lymphocyte ratio (PLR) in predicting mortality of intensive care unit (ICU) patients. Demographic data, underlying diseases, laboratory parameters were evaluated. The study included 222 cases. The mortality rate was 57.65%. No significant differences in terms of sex, age, or underlying disease were observed between the two groups with and without mortality. Obesity, oxygen therapy, invasive mechanical ventilation (IMV) rates and high SOFA (Sequential Organ Failure Assessment) scores were found to be significantly higher in the group with a mortal course. The mortality rate was significantly higher in patients with lung involvement over 50%, with a low lymphocyte count at ICU admission. In this patient group, NLR was found to be higher, and LCR was found to be lower (P = .001). Although there was no significant difference in PLR between the two groups in univariate analysis, multivariate analysis revealed that PLR was independently associated with mortality. High NLR and low LCR values at ICU admission might serve as early warning signs for healthcare providers, allowing them to identify patients at higher risk of mortality.
Collapse
Affiliation(s)
- Nurbanu Sezak
- Department of Infectious Disease and Clinical Microbiology, Izmir Democracy University Faculty of Medicine, Izmir, Turkiye
| | - Banu Karaca
- Department of Infectious Disease and Clinical Microbiology, Izmir Katip Celebi University Faculty of Medicine, Izmir, Turkiye
| | - Recep Balik
- Department of Infectious Disease and Clinical Microbiology, Goztepe Prof. Dr. Suleyman Yalcin City Hospital, Istanbul, Turkiye
| | - Murat Aksun
- Department of Anesthesiology and Reanimation, Izmir Katip Celebi University Faculty of Medicine, Izmir, Turkiye
| |
Collapse
|
17
|
Nalinthasnai N, Thammasudjarit R, Tassaneyasin T, Eksombatchai D, Sungkanuparph S, Boonsarngsuk V, Sutherasan Y, Junhasavasdikul D, Theerawit P, Petnak T. Unsupervised machine learning clustering approach for hospitalized COVID-19 pneumonia patients. BMC Pulm Med 2025; 25:70. [PMID: 39923003 PMCID: PMC11807335 DOI: 10.1186/s12890-025-03536-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: 10/18/2024] [Accepted: 01/28/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Identification of distinct clinical phenotypes of diseases can guide personalized treatment. This study aimed to classify hospitalized COVID-19 pneumonia subgroups using an unsupervised machine learning approach. METHODS We included hospitalized COVID-19 pneumonia patients from July to September 2021. K-means clustering, an unsupervised machine learning method, was performed to identify clinical phenotypes based on clinical and laboratory variables collected within 24 hours of admission. Variables were normalized before clustering to ensure equal contribution to the analysis. The optimal number of clusters was determined using the elbow method and Silhouette scores. Cox proportional hazard models were used to compare the risk of intubation and 90-day mortality across the identified clusters. RESULTS Three clinically distinct clusters were identified among 538 hospitalized COVID-19 pneumonia patients. Cluster 1 (N = 27) consisted predominantly of males and showed significantly elevated serum liver enzymes and LDH levels. Cluster 2 (N = 370) was characterized by lower chest x-ray scores and higher serum albumin levels. Cluster 3 (N = 141) was characterized by older age, diabetes mellitus, higher chest x-ray scores, more severe vital signs, higher creatinine levels, lower hemoglobin levels, lower lymphocyte counts, higher C-reactive protein, higher D-dimer, and higher LDH levels. When compared to cluster 2, cluster 3 was significantly associated with increased risk of 90-day mortality (HR, 6.24; 95% CI, 2.42-16.09) and intubation (HR, 5.26; 95% CI 2.37-11.72). In contrast, cluster 1 had a 100% survival rate with a non-significant increase in intubation risk compared to cluster 2 (HR, 1.40, 95% CI, 0.18-11.04). CONCLUSIONS We identified three distinct clinical phenotypes of COVID-19 pneumonia patients, with cluster 3 associated with an increased risk of respiratory failure and mortality. These findings may guide tailored clinical management strategies.
Collapse
Affiliation(s)
- Nuttinan Nalinthasnai
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | | | - Tanapat Tassaneyasin
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
- Faculty of Medicine Ramathibodi Hospital, Chakri Naruebodindra Medical Institute, Mahidol University, Samutprakan, Thailand
| | - Dararat Eksombatchai
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Somnuek Sungkanuparph
- Faculty of Medicine Ramathibodi Hospital, Chakri Naruebodindra Medical Institute, Mahidol University, Samutprakan, Thailand
| | - Viboon Boonsarngsuk
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Yuda Sutherasan
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Detajin Junhasavasdikul
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Pongdhep Theerawit
- Division of Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Tananchai Petnak
- Division of Pulmonary and Pulmonary Critical Care Medicine, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand.
| |
Collapse
|
18
|
Mataix RP, Morillo JSG, Martín JMS. Hepatic phenomena associated with SARS-CoV-2: Acute liver injury, autoimmune hepatitis and post-vaccination. Med Clin (Barc) 2025:S0025-7753(25)00011-9. [PMID: 39909769 DOI: 10.1016/j.medcli.2024.12.006] [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: 10/09/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 02/07/2025]
Abstract
The infection with SARS-CoV-2, primarily recognized for its respiratory effects, reveals itself as a multifaceted clinical phenomenon, extending beyond the pulmonary realm. Accompanied by gastrointestinal, neurological, thromboembolic, cardiovascular, and immune-related manifestations, the complexity of the systemic repercussions of the disease becomes apparent. Genetic predisposition is a significant factor in the development of autoimmune hepatitis, as both viruses, such as SARS-CoV-2, and drugs, including vaccines, can act as triggers in genetically susceptible individuals. A profound understanding of these mechanisms is essential to effectively address the clinical complexity of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Roberto Pertusa Mataix
- Unit of Systemic and Rare Autoimmune Diseases in Adults, Internal Medicine Service, Virgen del Rocío University Hospital, Manuel Siurot Avenue, S/n, 41013 Sevilla, Spain.
| | - José Salvador García Morillo
- Unit of Systemic and Rare Autoimmune Diseases in Adults, Internal Medicine Service, Virgen del Rocío University Hospital, Manuel Siurot Avenue, S/n, 41013 Sevilla, Spain
| | - José Manuel Sousa Martín
- Digestive Department, Virgen del Rocío University Hospital, Manuel Siurot Avenue, S/n, 41013 Sevilla, Spain
| |
Collapse
|
19
|
Jiang X, Hu J, Jiang Q, Zhou T, Yao F, Sun Y, Zhou C, Ma Q, Zhao J, Shi K, Yang W, Zhou X, Wang Y, Liu S, Xin X, Fan L. CT-based whole lung radiomics nomogram to identify middle-aged and elderly COVID-19 patients at high risk of progressing to critical disease. J Appl Clin Med Phys 2025; 26:e14562. [PMID: 39611805 DOI: 10.1002/acm2.14562] [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: 05/11/2024] [Revised: 08/19/2024] [Accepted: 09/16/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND COVID-19 remains widespread and poses a threat to people's physical and mental health, especially middle-aged and elderly individuals. Early identification of COVID-19 patients at high risk of progressing to critical disease helps improve overall patient outcomes and healthcare efficiency. PURPOSE To develop a radiomics nomogram to predict the risk of newly admitted middle-aged and elderly COVID-19 patients progressing to critical disease. METHODS A total of 794 patients (aged 40 years or above) were retrospectively included in the study from two institutions, all of them were with non-critical COVID-19 on admission. At follow-up, patients were divided into non-critical group and critical group. About 443 patients (384 non-critical and 59 critical) from the first hospital were randomly assigned to the training (n = 311) and internal validation (n = 132) set in a 7:3 ratio. Additionally, an independent external cohort of 351 patients (292 non-critical and 59 critical) from another hospital was evaluated. Radiomics signatures and clinical indicators were used to build a radiomics model and a clinical model after computed tomography (CT) image processing, CT whole-lung segmentation, feature extraction, and feature selection. The radiomics nomogram model integrated radiomics model and clinical model. The receiver operating characteristic curve (AUC) was used to assess the performance of the proposed models. Calibration curves and decision curve analysis were used to assess the performance of the radiomics nomogram. RESULTS For the training, internal validation, and external validation sets, the AUC values of the radiomic nomogram for the prediction of COVID-19 progression were 0.916, 0.917, and 0.890, respectively. Calibration curves indicated that there was no significant departure between prediction and observation in three sets. The decision curve image demonstrated the clinical utility of the nomogram model. CONCLUSIONS Our nomogram model incorporates radiomics features and clinical indicators, it provides a new pathway to increase predictive accuracy or clinical utility, further helping to provide personalized management for middle-aged and elderly patients with COVID-19.
Collapse
Affiliation(s)
- Xin'ang Jiang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jun Hu
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Qinling Jiang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Taohu Zhou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
- School of Medical Imaging, Weifang Medical University, Weifang, Shandong, China
| | - Fei Yao
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Yi Sun
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Chao Zhou
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Qianyun Ma
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jingyi Zhao
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Kang Shi
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Wen Yang
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xiuxiu Zhou
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yun Wang
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shiyuan Liu
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiaoyan Xin
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Li Fan
- Department of Radiology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
20
|
Amoroso D, Bongo S, Copponi A, Rossi V, Di Giorgio R, Bernardini S, Ippoliti L, Morello M. A Review of the Hematological Picture of Severe COVID-19 Infection. Cureus 2025; 17:e78797. [PMID: 39931501 PMCID: PMC11808344 DOI: 10.7759/cureus.78797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2025] [Indexed: 02/13/2025] Open
Abstract
Numerous hematological abnormalities have been documented in COVID-19 patients. We conducted an analysis of 82 articles from PubMed, focusing on the hematological characteristics observed in survivors (S) and non-survivors (NS) with moderate and severe COVID-19 symptoms, respectively. Our review underlines neutrophilia, lymphopenia, and thrombocytopenia as hallmark features of the disease. In severe cases, blood cell microscopy revealed the following abnormalities: i) an increased number of neutrophils, often displaying granularity, toxic granulation, and vacuolization; ii) lymphocytes with a notably blue cytoplasm; iii) several monocytes that contain vacuoles; iv) platelet aggregation; and v) basophilic stippling in red blood cells. Furthermore, scattergram analysis of COVID-19 patients revealed two common features: i) an increased neutrophil population and ii) the presence of a distinctive "sandglass pattern". This review underscores the critical role of hematochemical and cytomorphological blood cell analysis in COVID-19 patients, aiding clinicians in better recognizing and understanding the indicators of disease severity.
Collapse
Affiliation(s)
- Dominga Amoroso
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Stefania Bongo
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Anna Copponi
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Vanessa Rossi
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Roberta Di Giorgio
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Sergio Bernardini
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Lorenzo Ippoliti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, ITA
| | - Maria Morello
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| |
Collapse
|
21
|
Yu C, Wang W, Zhang Q, Jin Z. Autoimmune hepatitis under the COVID-19 veil: an analysis of the nature of potential associations. Front Immunol 2025; 16:1510770. [PMID: 39958350 PMCID: PMC11825795 DOI: 10.3389/fimmu.2025.1510770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 01/14/2025] [Indexed: 02/18/2025] Open
Abstract
In recent years, the novel coronavirus infectious disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has led to over 670 million infections and nearly 7 million deaths worldwide. The global pandemic of COVID-19 has precipitated a significant public health crisis. The prevalence of liver function abnormalities associated with SARS-CoV-2 is as high as 53% among healthy individuals or patients with autoimmune hepatitis (AIH) and shows a positive correlation with disease severity; moreover, specific adaptive immune responses can influence the trajectory and outcomes of COVID-19. For instance, SARS-CoV-2 may impact autoimmunity through mechanisms such as excessive stimulation of immune responses and molecular mimicry, particularly in genetically predisposed individuals. Currently, the overall mutational trend of SARS-CoV-2 indicates heightened infectivity and immune evasion capabilities. Consequently, vaccination remains crucial for universal protection against this disease. Nevertheless, alongside the widespread implementation of vaccination programs globally, an increasing number of cases have been documented where COVID-19 vaccination appears to trigger new-onset autoimmune hepatitis; yet definitive evidence is still pending elucidation regarding causality. In this review, we analyse the clinical-immunological characteristics, risks associated with severe disease progression, and prognosis for AIH patients infected with SARS-CoV-2; discuss the detrimental effects exerted by SARS-CoV-2 on hepatic function; summarise the mechanisms and attributes leading to new-onset AIH; as well as provide insights into how vaccination may interfere with autoimmunity processes. We continue to underscore the significance of vaccination while aiming to enhance awareness concerning potential risks associated with it-this could facilitate better management strategies for autoimmune diseases along with appropriate adjustments in vaccination protocols. Although the precise triggering mechanism linking COVID-19-related events to AIH remains unclear, existing evidence suggests that this relationship is far from coincidental.
Collapse
Affiliation(s)
| | | | | | - Zhenjing Jin
- Department of Hepatopancreatobiliary Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
22
|
Matsuura Y, Murashita M, Oyasu T, Kodate A, Sadamoto Y, Endo A, Sageshima H, Tsuchida T. Severe invasive streptococcal infection in a patient with COVID-19: A case report. IDCases 2025; 39:e02169. [PMID: 39989948 PMCID: PMC11847517 DOI: 10.1016/j.idcr.2025.e02169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 08/09/2024] [Accepted: 01/27/2025] [Indexed: 02/25/2025] Open
Abstract
A 63-year-old male with a history of hypertension had contracted Coronavirus Disease-2019 (COVID-19) five days before visiting our hospital and was recovering at home. He was brought to the hospital for emergency care because of bilateral leg pain, diarrhea, and shortness of breath. On arrival to the hospital, the patient was already suffering from multiple organ failure and was admitted to the intensive care unit. Streptococcus dysgalactiae subsp. equisimilis (SDSE) was detected in blood and sputum cultures, and we started antimicrobial therapy for septic shock/streptococcal toxic shock syndrome. However, multiple organ failure progressed, and the patient died one day after admission. This case suggests the possibility of immunosuppression due to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The patient had no history of immunodeficiency, and COVID-19 may have contributed to the poor outcome. COVID-19 combined with SDSE sepsis has not been previously reported. Therefore, this case was considered rare.
Collapse
Affiliation(s)
- Yuki Matsuura
- Department of Emergency and Critical Care Medicine, Sapporo City General Hospital, Sapporo, Japan
| | - Mone Murashita
- Department of Emergency and Critical Care Medicine, Sapporo City General Hospital, Sapporo, Japan
| | - Takayoshi Oyasu
- Department of Emergency and Critical Care Medicine, Sapporo City General Hospital, Sapporo, Japan
| | - Akira Kodate
- Department of Emergency and Critical Care Medicine, Sapporo City General Hospital, Sapporo, Japan
| | - Yoshihiro Sadamoto
- Department of Emergency and Critical Care Medicine, Sapporo City General Hospital, Sapporo, Japan
| | - Akio Endo
- Department of Emergency and Critical Care Medicine, Sapporo City General Hospital, Sapporo, Japan
| | - Hisako Sageshima
- Department of Emergency and Critical Care Medicine, Sapporo City General Hospital, Sapporo, Japan
| | - Takumi Tsuchida
- Department of Emergency and Critical Care Medicine, Sapporo City General Hospital, Sapporo, Japan
- Division of Acute and Critical Care Medicine, Department of Anesthesiology and Critical Care Medicine, Hokkaido University Faculty of Medicine, Sapporo, Japan
| |
Collapse
|
23
|
Zubovskaia A, Vazquez JA. Invasive Aspergillosis in the Intensive Care Unit. J Fungi (Basel) 2025; 11:70. [PMID: 39852489 PMCID: PMC11766804 DOI: 10.3390/jof11010070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Invasive aspergillosis (IA) is a fungal infection, which has traditionally been associated with neutropenia and immunosuppressive therapies. Our understanding of invasive aspergillosis has been evolving and, in the past few decades, IA among ICU patients has been recognized as a common infection and has become more widely recognized. The diagnosis and management of invasive aspergillosis in the ICU is particularly challenging, due to the unstable clinical condition of the patients, lack of diagnostic markers, increased risk of further clinical deterioration, multiple comorbidities, and a need for early assessment and treatment. In this article, we will discuss the challenges and pitfalls of the diagnosis and management of invasive aspergillosis in an ICU setting, along with a review of the current literature that is pertinent and specific to this population.
Collapse
Affiliation(s)
| | - Jose A. Vazquez
- Division of Infectious Diseases, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| |
Collapse
|
24
|
Drago F, Herzum A, Varesano S, Serviddio G, Broccolo F, Ciccarese G. Herald Patch as the Only Evidence of Pityriasis Rosea: Clinical, Laboratory and Pathogenetic Features. Viruses 2025; 17:119. [PMID: 39861908 PMCID: PMC11768941 DOI: 10.3390/v17010119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/09/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Pityriasis rosea (PR) is a self-limited exanthem associated with the endogenous systemic reactivation of human herpesvirus (HHV)-6 and HHV-7. The disease typically begins with a single erythematous patch on the trunk (herald patch), followed by a secondary eruption of smaller papulosquamous lesions. Rarely, the herald patch may be the only cutaneous manifestation of PR. The present work aimed to examine the clinical and laboratory features of the PR cases characterized by the herald patch as the sole cutaneous manifestation and to compare them with the classic form of PR. An observational, retrospective study was conducted on patients presenting with herald patch as the only sign of PR (cases) and on a series of age- and sex-matched patients who presented with a typical PR pattern (controls). The records of the patients were extracted from a PR registry, which collected data on patients with PR diagnosed from 2003 to 2023 by at least two dermatologists from the same study team. Nineteen patients (eleven males, eight females) with a mean age of 27.1 years presented the herald patch as the only cutaneous manifestation of PR. Nineteen age- and sex-matched patients were considered controls. In the cases, the exanthem duration was shorter than in controls, and the mean HHV-6 and HHV-7 DNA plasma load was lower than in controls. This rare variant of PR might be considered an abortive form of the exanthem that occurs when the HHV-6/7 reactivation from latency is contrasted by a more robust immunological response than in classic PR.
Collapse
Affiliation(s)
- Francesco Drago
- Dermatology Clinic, Villa Montallegro Health Clinic, Via Monte Zovetto 27, 16145 Genoa, Italy
| | - Astrid Herzum
- Section of Dermatology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147 Genoa, Italy;
| | - Serena Varesano
- Hygiene Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genoa, Italy
| | - Gaetano Serviddio
- Internal Medicine, Liver Unit, C.U.R.E. (University Centre for Liver Disease Research and Treatment), Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy;
| | - Francesco Broccolo
- Department of Experimental Medicine, University of Salento, Piazza Tancredi 7, 73100 Lecce, Italy;
| | - Giulia Ciccarese
- Section of Dermatology, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy;
| |
Collapse
|
25
|
Zhu Z, Zhang Y, Chen H, Zhang H. Cell-cell crosstalk in the pathogenesis of acute lung injury and acute respiratory distress syndrome. Tissue Barriers 2025:2452082. [PMID: 39798076 DOI: 10.1080/21688370.2025.2452082] [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: 07/08/2024] [Revised: 12/21/2024] [Accepted: 01/05/2025] [Indexed: 01/13/2025] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are the result of an exaggerated inflammatory response triggered by a variety of pulmonary and systemic insults. The lung tissues are comprised of a variety of cell types, including alveolar epithelial cells, pulmonary vascular endothelial cells, macrophages, neutrophils, and others. There is mounting evidence that these diverse cell populations within the lung interact to regulate lung inflammation in response to both direct and indirect stimuli. The aim of this review is to provide a summary and discussion of recent advances in the understanding of the importance of cell-cell crosstalk in the pathogenesis of ALI/ARDS, with a specific focus on the cell-cell interactions that may offer prospective therapeutic avenues for ALI/ARDS.
Collapse
Affiliation(s)
- Zhenzhen Zhu
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| | - Ying Zhang
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| | - Huan Chen
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| | - Huali Zhang
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Department of Pathophysiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, PR China
| |
Collapse
|
26
|
Yasseen BA, Elkhodiry AA, El-sayed H, Zidan M, Kamel AG, Badawy MA, Hamza MS, El-Messiery RM, El Ansary M, Abdel-Rahman EA, Ali SS. The role of neutrophilia in hyperlactatemia, blood acidosis, impaired oxygen transport, and mortality outcome in critically ill COVID-19 patients. Front Mol Biosci 2025; 11:1510592. [PMID: 39834785 PMCID: PMC11743367 DOI: 10.3389/fmolb.2024.1510592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 12/03/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction COVID-19 severity and high in-hospital mortality are often associated with severe hypoxemia, hyperlactatemia, and acidosis, yet the key players driving this association remain unclear. It is generally assumed that organ damage causes toxic acidosis, but since neutrophil numbers in severe COVID-19 can exceed 80% of the total circulating leukocytes, we asked if metabolic acidosis mediated by the glycolytic neutrophils is associated with lung damage and impaired oxygen delivery in critically ill patients. Methods Based on prospective mortality outcome, critically ill COVID-19 patients were divided into ICU- survivors and ICU-non-survivors. Samples were analyzed to explore if correlations exist between neutrophil counts, lung damage, glycolysis, blood lactate, blood pH, hemoglobin oxygen saturation, and mortality outcome. We also interrogated isolated neutrophils, platelets, and PBMCs for glycolytic activities. Results Arterial blood gas analyses showed remarkable hypoxemia in non-survivors with no consistent differences in PCO2 or [HCO3 -]. The hemoglobin oxygen dissociation curve revealed a right-shift, consistent with lower blood-pH and elevated blood lactate in non-survivors. Metabolic analysis of different blood cells revealed increased glycolytic activity only when considering the total number of neutrophils. Conclusion This indicates the role of neutrophilia in hyperlactatemia and lung damage, subsequently contributing to mortality outcomes in severe SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Basma A. Yasseen
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
| | - Aya A. Elkhodiry
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
| | - Hajar El-sayed
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
| | - Mona Zidan
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
| | - Azza G. Kamel
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
| | | | - Marwa S. Hamza
- Department of Clinical Pharmacy Practice, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Riem M. El-Messiery
- Infectious Disease Unit, Internal Medicine Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Mohamed El Ansary
- Department of Intensive Care, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Engy A. Abdel-Rahman
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
- Pharmacology Department, Faculty of Medicine, Assuit University, Assuit, Egypt
| | - Sameh S. Ali
- Research Department, Children’s Cancer Hospital Egypt, Cairo, Egypt
| |
Collapse
|
27
|
Cui S, Han Q, Zhang R, Zeng S, Shao Y, Li Y, Li M, Liu W, Zheng J, Wang H. Integration of metabolomics methodologies for the development of predictive models for mortality risk in elderly patients with severe COVID-19. BMC Infect Dis 2025; 25:10. [PMID: 39748307 PMCID: PMC11697755 DOI: 10.1186/s12879-024-10402-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND The rapid evolution of the COVID-19 pandemic and subsequent global immunization efforts have rendered early metabolomics studies potentially outdated, as they primarily involved non-exposed, non-vaccinated populations. This paper presents a predictive model developed from up-to-date metabolomics data integrated with clinical data to estimate early mortality risk in critically ill COVID-19 patients. Our study addresses the critical gap in current research by utilizing current patient samples, providing fresh insights into the pathophysiology of the disease in a partially immunized global population. METHODS One hundred elderly patients with severe COVID-19 infection, including 46 survivors and 54 non-survivors, were recruited in January-February 2023 at the Second Hospital affiliated with Harbin Medical University. A predictive model within 24 h of admission was developed using blood metabolomics and clinical data. Differential metabolite analysis and other techniques were used to identify relevant characteristics. Model performance was assessed by comparing the area under the receiver operating characteristic curve (AUROC). The final prediction model was externally validated in a cohort of 50 COVID-19 elderly critically ill patients at the First Hospital affiliated with Harbin Medical University during the same period. RESULTS Significant disparities in blood metabolomics and laboratory parameters were noted between individuals who survived and those who did not. One metabolite indicator, Itaconic acid, and four laboratory tests (LYM, IL-6, PCT, and CRP), were identified as the five variables in all four models. The external validation set demonstrated that the KNN model exhibited the highest AUC of 0.952 among the four models. When considering a 50% risk of mortality threshold, the validation set displayed a sensitivity of 0.963 and a specificity of 0.957. CONCLUSIONS The prognostic outcome of COVID-19 elderly patients is significantly influenced by the levels of Itaconic acid, LYM, IL-6, PCT, and CRP upon admission. These five indicators can be utilized to assess the mortality risk in affected individuals.
Collapse
Affiliation(s)
- Shanpeng Cui
- Department of Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Qiuyuan Han
- Department of Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China
| | - Ran Zhang
- School of Measurement-Control and Communication Engineering, Harbin University of Science and Technology, Harbin, 150080, Heilongjiang Province, China
| | - Siyao Zeng
- Department of Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China
| | - Ying Shao
- Interventional vascular department, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Yue Li
- Department of Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China
| | - Ming Li
- Department of Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China
| | - Wenhua Liu
- Department of Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China.
| | - Junbo Zheng
- Department of Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China.
| | - Hongliang Wang
- Department of Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, Heilongjiang Province, China.
| |
Collapse
|
28
|
Bekbossynova M, Akhmaltdinova L, Dossybayeva K, Tauekelova A, Smagulova Z, Tsechoeva T, Turebayeva G, Sailybayeva A, Kalila Z, Mirashirova T, Muratov T, Poddighe D. Prospective and Longitudinal Analysis of Lymphocyte Subpopulations in SARS-CoV-2 Positive and Negative Pneumonia: Potential Role of Decreased Naïve CD8 + in COVID-19 Patients. Viruses 2024; 17:41. [PMID: 39861830 PMCID: PMC11768816 DOI: 10.3390/v17010041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/23/2024] [Accepted: 12/25/2024] [Indexed: 01/27/2025] Open
Abstract
Background: During the acute phase of COVID-19, a number of immunological abnormalities have been reported, but few studies longitudinally analyzed the specific subsets of peripheral blood lymphocytes. Methods: In this observational, prospective, and longitudinal study, adult patients developing acute pneumonia during the COVID-19 pandemic have been followed up for 12 months. Peripheral blood lymphocyte subsets were assessed (with a specific focus on the memory markers) at 6 time points after the disease onset until 12 months. Results: A total of 76 patients with acute pneumonia (characterized by a prevalently interstitial pattern of lung inflammation) at the hospital admission (who completed the 12-month follow-up period) were recruited in this study. They were divided into two groups, namely positive (n = 31) and negative (n = 45) patients for the SARS-CoV-2 PCR test. In the acute phase, the general lymphocyte immunophenotyping profile was comparable for most parameters between these groups, except for B cells. When B and T cells were analyzed according to the expression of memory markers, a significant decrease in naïve CD8+ T cells was observed in the SARS-CoV-2-positive pneumonia group during the acute phase. Notably, this aspect was maintained during the follow-up period for at least 9 months. Conclusions: COVID-19 pneumonia seems to be associated with a lower number of naïve CD8+ T cells compared to pneumonia patients negative for this virus. This alteration can persist in the convalescent phase.
Collapse
Affiliation(s)
| | | | - Kuanysh Dossybayeva
- National Research Cardiac Surgery Center, Astana 010000, Kazakhstan
- School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
| | - Ainur Tauekelova
- National Research Cardiac Surgery Center, Astana 010000, Kazakhstan
| | - Zauresh Smagulova
- Department of Infectious Diseases and Clinical Epidemiology, Astana Medical University, Astana 010000, Kazakhstan
| | - Tatyana Tsechoeva
- Department of Infectious Diseases and Clinical Epidemiology, Astana Medical University, Astana 010000, Kazakhstan
| | - Gulsimzhan Turebayeva
- Department of Infectious Diseases and Clinical Epidemiology, Astana Medical University, Astana 010000, Kazakhstan
| | | | - Zhanar Kalila
- National Research Cardiac Surgery Center, Astana 010000, Kazakhstan
| | | | - Timur Muratov
- Department of Public Health of Astana, Astana 010000, Kazakhstan
| | - Dimitri Poddighe
- School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
- University Medical Center (UMC), Astana 010000, Kazakhstan
- College of Health Sciences, VinUniversity, Gia Lam District, Hanoi 100000, Vietnam
| |
Collapse
|
29
|
Héja M, Fekete I, Márton S, Horváth L, Fekete K. Impact of COVID-19 pandemic on acute stroke care in a tertiary stroke centre. Sci Rep 2024; 14:31408. [PMID: 39733029 PMCID: PMC11682369 DOI: 10.1038/s41598-024-83016-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
The aim of this study was to evaluate how COVID-19 affected acute stroke care and outcome in patients with acute ischemic or hemorrhagic stroke. We performed a retrospective analysis on patients who were admitted with acute ischemic (AIS) or hemorrhagic (ICH) stroke from September 2020 to May 2021 with and without COVID-19. We recorded demographic and clinical data, imaging parameters, functional outcome and mortality at one year. Beside descriptive statistics we performed χ2-probe, Mann-Witney U-test, Student t-probe and multivariate testing. We found a 29%-reduction in the number of AIS cases during the pandemic. The number of the large vessel occlusions /LVOs/ (N = 83, 41.7%), from them 37 (17.7%) had mechanical thrombectomy (MT), was higher than before the COVID-19 period (p = 0.02 and p = 0.001, respectively). From all patients needing acute revascularization therapy (N = 137) 118 patients received it, among them 20 (16.9%) had COVID-19. Those positive for COVID-19 were more likely to have a higher median NIHSS score at baseline and at 24 h (p = 0.02 and p = 0.03, respectively). They also had a lower rate of favourable outcome at discharge (15% vs. 41.8%; p = 0.024) and at three months (25% vs. 52%, p = 0.02), longer median hospitalization (p < 0.0001), and a higher mortality rate (52% vs. 25%; p = 0.03). The incidence of symptomatic intracerebral hemorrhage (sICH) did not differ between the groups. Regarding the ICH patients, NIHSS score at 24 h (p = 0.036), mortality at 3 months (p = 0.004) and at one year (p = 0.00) were higher in the COVID-19 group. We concluded that the pandemic resulted fewer admission due to AIS with an increased number of LVOs and MTs. AIS patients with concomitant SARS-CoV-2 infection have more severe strokes and unfavorable long term outcome. The risk of sICH was not increased in COVID-19 positive patients therefore reperfusion therapies appear to be safe and beneficial for some individuals. Patients with ICH and comorbid COVID-19 have a very poor prognosis.
Collapse
Affiliation(s)
- Máté Héja
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - István Fekete
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Sándor Márton
- Faculty of Arts, Institute of Political Science and Sociology, University of Debrecen, Debrecen, Hungary
| | - László Horváth
- Department of Pharmaceutical Surveillance and Economics, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Klára Fekete
- Department of Neurology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
30
|
Li X, Zhu H, Xu P, Zhang J, Wang Z, He H, Shen F, Jiang Y, Shen L, Xiang J, Yang L, Yang C, Jiang H, Gao G, Jin J, Shen H, Wang Y, Wu L, Qian C, Liu D, Qiu W, Li Q, Chen Y, Lin F, Liu Y. A comprehensive immune repertoire signature distinguishes pulmonary infiltration in SARS-CoV-2 Omicron variant infection. Front Immunol 2024; 15:1486352. [PMID: 39742285 PMCID: PMC11685115 DOI: 10.3389/fimmu.2024.1486352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/27/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction The coronavirus disease 2019 (COVID-19) global pandemic has been the most severe public health emergency since 2019. Currently, the Omicron variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been the most dominant. The most prominent symptom of SARS-CoV-2 infection is respiratory. Meanwhile, the fatality of COVID-19 was mainly from pneumonia. However ,in patients with SARS-CoV-2 infection who have pneumonia and those who do not, the differences in the immune repertoire still require further investigation. Methods We conducted seven-chain adaptome immune repertoire analyses on patients with SARS-CoV-2 Omicron infection, both with and without pulmonary infiltration. Results Patients with pulmonary infiltration exhibit lymphopenia, a decreased proportion of the overall TCR repertoire alongside an increased BCR repertoire, reduced IGHD and IGHM isotype expression, a shorter mean CDR3 length for TRG, and a longer mean length for TRD, as well as diminished clonality and diversity in the TCR/BCR repertoire. Meanwhile, patients with pulmonary infiltration have distinct V-J gene usage and unique CDR3 signature, as well as BCR class switch recombination pattern. Finally, prior vaccination triggered less BCR IGHM/IGHD somatic hypermutation response, preserved the diversity of the entire adaptive immune repertoire, and provided clinical protection against severe or critical conditions following Omicron infection. Discussion We report a unique, comprehensive adaptive immune system signature in patients with pulmonary infiltration, which may serve as potential immunological biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Xuechuan Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Hongyi Zhu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Peipei Xu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jie Zhang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Zhe Wang
- Tilcure Biotherapeutics, Shanghai, China
| | - Hui He
- Department of Training Department, China Medical University Benxi Central Hospital Postgraduate Training Workstation, Shanghai, China
| | - Fang Shen
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yi Jiang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lijuan Shen
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jing Xiang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Linhua Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Chao Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Hao Jiang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Ganglong Gao
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Junshuo Jin
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Huojian Shen
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yinping Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Linshi Wu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Changlin Qian
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Dejun Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Weiqing Qiu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Qiwei Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Yuanwen Chen
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Fujun Lin
- Renal Division, Department of Internal Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun Liu
- Shanghai Cancer Institute, Shanghai, China
| |
Collapse
|
31
|
Leite GGF, Sousa MB, Rodrigues LDOCP, Brunialti MKC, Medina-Pestana J, Butler JM, Peters-Sengers H, Requião-Moura L, Salomão R. Proteomic profiling of peripheral blood mononuclear cells reveals immune dysregulation and metabolic alterations in kidney transplant recipients with COVID-19. Front Immunol 2024; 15:1508110. [PMID: 39737170 PMCID: PMC11683116 DOI: 10.3389/fimmu.2024.1508110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
The COVID-19 pandemic has significantly impacted global health, especially in vulnerable populations like kidney transplant recipients (KTRs). Recently, mass spectrometry-based proteomics has emerged as a powerful tool to shed light on a broad spectrum of dysregulated biological processes in KTRs with COVID-19. In this study, we prospectively collected blood samples from 17 COVID-19-positive KTRs and 10 non-infected KTRs between May and September 2020. Using tandem mass tag-based quantitative proteomics, we analyzed peripheral blood mononuclear cells (PBMCs), plasma protein biomarkers, and lymphocyte counts, followed by bioinformatics analysis. Our results revealed significant proteomic alterations in COVID-19-infected KTRs, particularly in pathways related to glycolysis, glucose metabolism, and neutrophil degranulation. Additionally, we observed an altered immune response characterized by elevated cytokines and decreased lymphocyte counts. Notably, KTRs with AKI exhibited worse clinical outcomes, including higher rates of ICU admission and mechanical ventilation. Comparative analysis of PBMC proteomic profiles between AKI and non-AKI patients identified distinct immune-related pathways, with AKI patients showing marked changes in innate immune responses, particularly neutrophil degranulation. Furthermore, we observed a negative correlation between T cell counts and neutrophil degranulation, suggesting a role for immune dysregulation in COVID-19. Our findings provide critical insights into the immune and metabolic responses in COVID-19-infected KTRs, especially those with AKI, highlighting the need for focused research and therapeutic strategies targeting immune dysregulation in this high-risk population.
Collapse
Affiliation(s)
- Giuseppe G. F. Leite
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Mônica Bragança Sousa
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Milena Karina Colo Brunialti
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - José Medina-Pestana
- Division of Nephrology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Hospital do Rim, Fundação Oswaldo Ramos, São Paulo, Brazil
| | - Joe M. Butler
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands
| | - Hessel Peters-Sengers
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC, location University of Amsterdam, Amsterdam, Netherlands
- Department of Epidemiology and Data Science, Amsterdam UMC, location Vrije Universiteit, Amsterdam, Netherlands
| | - Lúcio Requião-Moura
- Division of Nephrology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Hospital do Rim, Fundação Oswaldo Ramos, São Paulo, Brazil
| | - Reinaldo Salomão
- Division of Infectious Diseases, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
32
|
Wang J, Li X, Ren J, Rao Y, Qiao Y, Sun L, Liang Y, Chang C, Zhou Q, Sun Y. The Association of Blood Eosinophils and Neutrophils Expressing Eosinophilic Surface Markers with the Severity and Outcome of COVID-19. Microorganisms 2024; 12:2503. [PMID: 39770705 PMCID: PMC11727756 DOI: 10.3390/microorganisms12122503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 01/16/2025] Open
Abstract
(1) Background: The implication of type 2 (T2) inflammatory response in COVID-19 remains controversial. This study aimed to evaluate the association of eosinophils, neutrophils expressing eosinophilic surface markers and T2 cytokines with the severity and outcome of COVID-19. (2) Methods: Patients who were admitted to hospital due to COVID-19 from 18 December 2022 to 31 January 2023 were enrolled. Peripheral blood WBC and differentials, T2 cellular markers (subsets of eosinophils and neutrophils expressing eosinophilic surface markers) and cytokines at admission were measured and compared between subjects with different disease severities and outcomes. (3) Results: Ten mild-to-moderate and 22 severe-to-very severe cases were enrolled for analysis. Of these patients, seven died of severe-to-very severe disease. The severe-to-very severe patients showed a higher number of neutrophils, but lower numbers of eosinophils, lymphocytes cells and neutrophils expressing eosinophilic surface markers. Similarly, deceased cases were also characterized by increased neutrophils, but decreased eosinophils and neutrophils expressing eosinophilic surface markers. The levels of T2 cytokines failed to demonstrate a significant correlation with the severity or outcome of COVID-19. (4) Conclusions: Eosinophils and neutrophils expressing eosinophilic surface markers were associated with milder disease and better outcomes of COVID-19, suggesting that a T2 inflammatory response may confer a potential protective effect against the disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Research Center for Chronic Airway Diseases, Peking University Health Science Center, Beijing 100191, China; (J.W.)
| |
Collapse
|
33
|
Ceylan B, Olmuşçelik O, Karaalioğlu B, Ceylan Ş, Şahin M, Aydın S, Yılmaz E, Dumlu R, Kapmaz M, Çiçek Y, Kansu A, Duger M, Mert A. Predicting Severe Respiratory Failure in Patients with COVID-19: A Machine Learning Approach. J Clin Med 2024; 13:7386. [PMID: 39685844 DOI: 10.3390/jcm13237386] [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: 09/18/2024] [Revised: 10/23/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: Studies attempting to predict the development of severe respiratory failure in patients with a COVID-19 infection using machine learning algorithms have yielded different results due to differences in variable selection. We aimed to predict the development of severe respiratory failure, defined as the need for high-flow oxygen support, continuous positive airway pressure, or mechanical ventilation, in patients with COVID-19, using machine learning algorithms to identify the most important variables in achieving this prediction. Methods: This retrospective, cross-sectional study included COVID-19 patients with mild respiratory failure (mostly receiving oxygen through a mask or nasal cannula). We used XGBoost, support vector machines, multi-layer perceptron, k-nearest neighbor, random forests, decision trees, logistic regression, and naïve Bayes methods to accurately predict severe respiratory failure in these patients. Results: A total of 320 patients (62.1% male; average age, 54.67 ± 15.82 years) were included in this study. During the follow-ups of these cases, 114 patients (35.6%) required high-level oxygen support, 67 (20.9%) required intensive care unit admission, and 43 (13.4%) died. The machine learning algorithms with the highest accuracy values were XGBoost, support vector machines, k-nearest neighbor, logistic regression, and multi-layer perceptron (0.7395, 0.7395, 0.7291, 0.7187, and 0.75, respectively). The method that obtained the highest ROC-AUC value was logistic regression (ROC-AUC = 0.7274). The best predictors of severe respiratory failure were a low lymphocyte count, a high computed tomography score in the right and left upper lung zones, an elevated neutrophil count, a small decrease in CRP levels on the third day of admission, a high Charlson comorbidity index score, and a high serum procalcitonin level. Conclusions: The development of severe respiratory failure in patients with COVID-19 could be successfully predicted using machine learning methods, especially logistic regression, and the best predictors of severe respiratory failure were the lymphocyte count and the degree of upper lung zone involvement.
Collapse
Affiliation(s)
- Bahadır Ceylan
- Department of Infectious Diseases and Clinical Microbiology, Medical Faculty, Istanbul Medipol University, Istanbul 34214, Türkyie
| | - Oktay Olmuşçelik
- Department of Internal Medicine, Medical Faculty, Istanbul Medipol University, Istanbul 34214, Türkyie
| | - Banu Karaalioğlu
- Department of Radiology, Medical Faculty, Istanbul Medipol University, Istanbul 34214, Türkyie
| | - Şule Ceylan
- Department of Nuclear Medicine, University of Health Science, Gaziosmanpaşa Training ve Research Hospital, Istanbul 34668, Türkyie
| | - Meyha Şahin
- Department of Infectious Diseases and Clinical Microbiology, Medical Faculty, Istanbul Medipol University, Istanbul 34214, Türkyie
| | - Selda Aydın
- Department of Infectious Diseases and Clinical Microbiology, Medical Faculty, Istanbul Medipol University, Istanbul 34214, Türkyie
| | - Ezgi Yılmaz
- Department of Infectious Diseases and Clinical Microbiology, Medical Faculty, Istanbul Medipol University, Istanbul 34214, Türkyie
| | - Rıdvan Dumlu
- Department of Infectious Diseases and Clinical Microbiology, Medical Faculty, Istanbul Medipol University, Istanbul 34214, Türkyie
| | - Mahir Kapmaz
- Department of Infectious Diseases and Clinical Microbiology, Medical Faculty, Istanbul Medipol University, Istanbul 34214, Türkyie
| | - Yeliz Çiçek
- Department of Infectious Diseases and Clinical Microbiology, Medical Faculty, Istanbul Medipol University, Istanbul 34214, Türkyie
| | - Abdullah Kansu
- Department of Chest Diseases, Medical Faculty, Istanbul Medipol University, Istanbul 34214, Türkyie
| | - Mustafa Duger
- Department of Chest Diseases, Medical Faculty, Istanbul Medipol University, Istanbul 34214, Türkyie
| | - Ali Mert
- Department of Internal Medicine, Medical Faculty, Istanbul Medipol University, Istanbul 34214, Türkyie
| |
Collapse
|
34
|
Valderrábano RJ, Wipper B, Pencina KM, Migaud M, Shang YV, Latham NK, Montano M, Cunningham JM, Wilson L, Peng L, Memish‐Beleva Y, Bhargava A, Swain PM, Lehman P, Lavu S, Livingston DJ, Bhasin S. Dysregulated nicotinamide adenine dinucleotide metabolome in patients hospitalized with COVID-19. Aging Cell 2024; 23:e14326. [PMID: 39354697 PMCID: PMC11634700 DOI: 10.1111/acel.14326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 10/03/2024] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) depletion has been postulated as a contributor to the severity of COVID-19; however, no study has prospectively characterized NAD+ and its metabolites in relation to disease severity in patients with COVID-19. We measured NAD+ and its metabolites in 56 hospitalized patients with COVID-19 and in two control groups without COVID-19: (1) 31 age- and sex-matched adults with comorbidities, and (2) 30 adults without comorbidities. Blood NAD+ concentrations in COVID-19 group were only slightly lower than in the control groups (p < 0.05); however, plasma 1-methylnicotinamide concentrations were significantly higher in patients with COVID-19 (439.7 ng/mL, 95% CI: 234.0, 645.4 ng/mL) than in age- and sex-matched controls (44.5 ng/mL, 95% CI: 15.6, 73.4) and in healthy controls (18.1 ng/mL, 95% CI 15.4, 20.8; p < 0.001 for each comparison). Plasma nicotinamide concentrations were also higher in COVID-19 group and in controls with comorbidities than in healthy control group. Plasma concentrations of 2-methyl-2-pyridone-5-carboxamide (2-PY), but not NAD+, were significantly associated with increased risk of death (HR = 3.65; 95% CI 1.09, 12.2; p = 0.036) and escalation in level of care (HR = 2.90, 95% CI 1.01, 8.38, p = 0.049). RNAseq and RTqPCR analyses of PBMC mRNA found upregulation of multiple genes involved in NAD+ synthesis as well as degradation, and dysregulation of NAD+-dependent processes including immune response, DNA repair, metabolism, apoptosis/autophagy, redox reactions, and mitochondrial function. Blood NAD+ concentrations are modestly reduced in COVID-19; however, NAD+ turnover is substantially increased with upregulation of genes involved in both NAD+ biosynthesis and degradation, supporting the rationale for NAD+ augmentation to attenuate disease severity.
Collapse
Affiliation(s)
- Rodrigo J. Valderrábano
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Benjamin Wipper
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Karol Mateusz Pencina
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Marie Migaud
- Department of Pharmacology, Mitchell Cancer InstituteUniversity of South AlabamaMobileAlabamaUSA
| | - Yili Valentine Shang
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Nancy K. Latham
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Monty Montano
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - James M. Cunningham
- Division of Hematology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Lauren Wilson
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Liming Peng
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Yusnie Memish‐Beleva
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Avantika Bhargava
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | | | - Phoebe Lehman
- Metro International BiotechWorcesterMassachusettsUSA
| | - Siva Lavu
- Metro International BiotechWorcesterMassachusettsUSA
| | | | - Shalender Bhasin
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
35
|
Bianconi V, Mannarino MR, Figorilli F, Ricciutelli F, De Carlo S, Zullo V, Corba M, Sahebkar A, Greco A, Lombardini R, Paltriccia R, Pirro M. Treatment with proton pump inhibitors is associated with secondary bacterial infections and sepsis in patients with COVID-19: a retrospective analysis of their joint impact on in-hospital prognosis. Ann Med 2024; 56:2399761. [PMID: 39475004 PMCID: PMC11616760 DOI: 10.1080/07853890.2024.2399761] [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: 12/28/2023] [Revised: 03/29/2024] [Accepted: 04/27/2024] [Indexed: 11/10/2024] Open
Abstract
Introduction and objectives. Secondary bacterial infections (SBIs) contribute to worse in-hospital outcomes in patients with Coronavirus disease 2019 (COVID-19). Treatment with proton pump inhibitors (PPIs) is associated with an increased risk of bacterial infections in different clinical settings. However, the association between PPI treatment prior to hospital admission and the occurrence of either SBIs or secondary bacterial sepsis (SBS) as well as their joint impact on clinical outcomes of patients hospitalized for COVID-19 are not clarified. Patients and methods. We retrospectively analyzed preadmission PPI use, in-hospital occurrence of SBIs and SBS, and in-hospital outcomes of a cohort of patients hospitalized for COVID-19. Results. Among 1087 patients, 447 (41%) were on PPI treatment prior to hospital admission. During the hospital stay, 197 (18%) and 223 (20%) patients were diagnosed with SBIs and SBS, respectively. The composite endpoint of intensive care unit (ICU) admission/in-hospital death was met by 214 (20%) patients. Preadmission PPI treatment was independently associated with up to a 2.1-fold and 1.7-fold increased risk of SBIs and SBS, respectively. The occurrence of SBS was independently associated with up to a 2.2-fold increased risk of ICU admission/in-hospital death. A significant preadmission PPI treatment x SBS interaction emerged in predicting ICU admission/in-hospital death (F = 5.221, pinteraction = 0.023). Conclusions. PPI treatment prior to hospital admission for COVID-19 is associated with an increased risk of SBIs and SBS. In addition, it interacts with SBS in predicting in-hospital prognosis. An appropriate use of PPIs may attenuate the risk of adverse clinical outcomes during hospitalization for COVID-19.
Collapse
Affiliation(s)
- Vanessa Bianconi
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Massimo R. Mannarino
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Filippo Figorilli
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Federica Ricciutelli
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Stefania De Carlo
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Valentina Zullo
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Martina Corba
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alessia Greco
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rita Lombardini
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rita Paltriccia
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
36
|
Menotti S, di Filippo L, Terenzi U, Chiloiro S, De Marinis L. Hypophysitis in COVID-19: a systematic review. Pituitary 2024; 27:874-888. [PMID: 39404935 DOI: 10.1007/s11102-024-01462-4] [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] [Accepted: 09/24/2024] [Indexed: 12/12/2024]
Abstract
PURPOSE This systematic review aims to collect and examine recent research findings regarding hypophysitis in COVID-19 patients. METHOD We conducted a comprehensive literature review in English on the topic "Hypophysitis in COVID-19," using the MEDLINE (PubMed) database in July 2024. The selected articles were systematically tabulated and we have assessed in this review patient demographics, symptom presentation, imaging results, diagnosis, clinical management, and outcomes. RESULTS Seven reported cases of post-COVID-19 hypophysitis were identified, comprising 4 (57%) females and 3 (43%) males, with a median age of 37 years. The interval between COVID-19 infection symptoms and the onset of hypophysitis ranged from 2 to 3 weeks. Initial symptoms included frontal headache in 4 (57%) cases and polyuria and polydipsia in 3 (43%) cases. Anterior or posterior hypopituitarism was observed in 6 (85%) patients. Radiological findings varied: 2 (28.5%) cases showed panhypophysitis, 3 (43%) cases exhibited gland enlargement with homogeneous contrast enhancement on magnetic resonance imaging (MRI), 1 case involved the loss of the posterior pituitary bright spot, and 1 case involved pituitary apoplexy/enlargement of the gland and infundibulum. No pituitary biopsies were performed. Four (57%) patients received glucocorticoid (GC) treatment. Long-term follow-up was documented in only one case, a 16-year-old female followed for 2 years reporting complete clinical and radiological resolution. CONCLUSION Although rare, hypophysitis related to COVID-19 is documented in the literature exhibiting distinct characteristics such as a homogeneous gender prevalence, an average age of onset around 35 years, and primary symptoms of headache, polyuria, and polydipsia which are indicative of angiotensin-vasopressin deficiency. This is in contrast with primary autoimmune hypophysitis characterized by a female prevalence and typical symptoms with headache and visual impairment. Longer-term follow-up of these patients is needed to better understand the potential lasting impact on pituitary function and radiological improvement. Future research should also explore the presence of anti-pituitary antibodies and the other possible pathophysiological mechanisms potentially involved in these cases.
Collapse
Affiliation(s)
- Sara Menotti
- Institute of Endocrine and Metabolic Sciences, San Raffaele Vita-Salute University and IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milan, Italy.
| | - Luigi di Filippo
- Institute of Endocrine and Metabolic Sciences, San Raffaele Vita-Salute University and IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milan, Italy
| | - Umberto Terenzi
- Institute of Endocrine and Metabolic Sciences, San Raffaele Vita-Salute University and IRCCS San Raffaele Hospital, Via Olgettina 60, 20132, Milan, Italy
| | - Sabrina Chiloiro
- Pituitary Unit, Department of Endocrinology and Diabetes, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Sacred Heart Catholic University, Rome, Italy
| | - Laura De Marinis
- Pituitary Unit, Department of Endocrinology and Diabetes, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
- Department of Translational Medicine and Surgery, Sacred Heart Catholic University, Rome, Italy
| |
Collapse
|
37
|
Zhao WJ, Tan RZ, Gao J, Su H, Wang L, Liu J. Research on the global trends of COVID-19 associated acute kidney injury: a bibliometric analysis. Ren Fail 2024; 46:2338484. [PMID: 38832469 PMCID: PMC11262241 DOI: 10.1080/0886022x.2024.2338484] [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: 11/22/2023] [Accepted: 03/29/2024] [Indexed: 06/05/2024] Open
Abstract
Critically ill COVID-19 patients may exhibit various clinical symptoms of renal dysfunction including severe Acute Kidney Injury (AKI). Currently, there is a lack of bibliometric analyses on COVID-19-related AKI. The aim of this study is to provide an overview of the current research status and hot topics regarding COVID-19 AKI. The literature was retrieved from the Web of Science Core Collection (WoSCC) database. Subsequently, we utilized Microsoft Excel, VOSviewer, Citespace, and Pajek software to revealed the current research status, emerging topics, and developmental trends pertaining to COVID-19 AKI. This study encompassed a total of 1507 studies on COVID-19 AKI. The United States, China, and Italy emerged as the leading three countries in terms of publication numbers, contributing 498 (33.05%), 229 (15.20%), and 140 (9.29%) studies, respectively. The three most active and influential institutions include Huazhong University of Science and Technology, Wuhan University and Harvard Medical School. Ronco C from Italy, holds the record for the highest number of publications, with a total of 15 papers authored. Cheng YC's work from China has garnered the highest number of citations, totaling 470 citations. The co-occurrence analysis of author keywords reveals that 'mortality', 'intensive care units', 'chronic kidney disease', 'nephrology', 'renal transplantation', 'acute respiratory distress syndrome', and 'risk factors' emerge as the primary areas of focus within the realm of COVID-19 AKI. In summary, this study analyzes the research trends in the field of COVID-19 AKI, providing a reference for further exploration and research on COVID-19 AKI mechanisms and treatment.
Collapse
Affiliation(s)
- Wen-jing Zhao
- Department of Nephrology of the Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Rui-zhi Tan
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jing Gao
- Department of Nephrology of the Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Hongwei Su
- Department of Urology, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Li Wang
- Research Center of Intergated Traditional Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Jian Liu
- Department of Nephrology of the Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University
- Department of Nephrology of the Affiliated Hospital of Southwest Medical University
| |
Collapse
|
38
|
Wu M, Sun C, Shi Q, Luo Y, Wang Z, Wang J, Qin Y, Cui W, Yan C, Dai H, Wang Z, Zeng J, Zhou Y, Zhu M, Liu X. Dry eye disease caused by viral infection: Past, present and future. Virulence 2024; 15:2289779. [PMID: 38047740 PMCID: PMC10761022 DOI: 10.1080/21505594.2023.2289779] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023] Open
Abstract
Following viral infection, the innate immune system senses viral products, such as viral nucleic acids, to activate innate defence pathways, leading to inflammation and apoptosis, control of cell proliferation, and consequently, threat to the whole body. The ocular surface is exposed to the external environment and extremely vulnerable to viral infection. Several studies have revealed that viral infection can induce inflammation of the ocular surface and reduce tear secretion of the lacrimal gland (LG), consequently triggering ocular morphological and functional changes and resulting in dry eye disease (DED). Understanding the mechanisms of DED caused by viral infection and its potential therapeutic strategies are crucial for clinical interventional advances in DED. This review summarizes the roles of viral infection in the pathogenesis of DED, applicable diagnostic and therapeutic strategies, and potential regions of future studies.
Collapse
Affiliation(s)
- Min Wu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Cuilian Sun
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Qin Shi
- Department of General Medicine, Gongli Hospital, Shanghai, China
| | - Yalu Luo
- Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Ziyu Wang
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Jianxiang Wang
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Yun Qin
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Weihang Cui
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Chufeng Yan
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Huangyi Dai
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Zhiyang Wang
- Medical College, Nantong University, Nantong, Jiangsu, China
| | - Jia Zeng
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Yamei Zhou
- Department of Microbiology Laboratory, Jiaxing Center for Disease Control and Prevention, Jiaxing, Zhejiang, China
| | - Manhui Zhu
- Department of Ophthalmology, Lixiang Eye Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaojuan Liu
- Department of Pathogen Biology, Medical College, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
39
|
Amarilla-Irusta A, Zenarruzabeitia O, Sevilla A, Sandá V, Lopez-Pardo A, Astarloa-Pando G, Pérez-Garay R, Pérez-Fernández S, Meijide S, Imaz-Ayo N, Arana-Arri E, Amo L, Borrego F. CD151 identifies an NK cell subset that is enriched in COVID-19 patients and correlates with disease severity. J Infect 2024; 89:106304. [PMID: 39374860 DOI: 10.1016/j.jinf.2024.106304] [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: 04/22/2024] [Revised: 09/27/2024] [Accepted: 09/29/2024] [Indexed: 10/09/2024]
Abstract
Severe coronavirus disease 2019 (COVID-19) often leads to acute respiratory distress syndrome and multi-organ dysfunction, driven by a dysregulated immune response, including a cytokine storm with elevated proinflammatory cytokine levels. Natural killer (NK) cells are part of the innate immune system with a fundamental role in the defense against viral infections. However, during COVID-19 acute infection, they exhibit an altered phenotype and impaired functionality contributing to the immunopathogenesis of the disease. In this work, we have studied a cohort of patients with COVID-19 (ranging from mild to severe) by analyzing IL-15, TGF-β, PlGF and GDF-15 plasma levels and performing multiparametric flow cytometry studies. Our results revealed that severe COVID-19 patients exhibited high levels of IL-15, PlGF and GDF-15, along with an enrichment of an NK cell subset expressing the CD151 tetraspanin, which correlated with IL-15 plasma levels and disease severity. In patients, these CD151+ NK cells displayed a more activated phenotype characterized by an increased expression of HLA-DR, CD38 and granzyme B, a distinct receptor repertoire, with lower levels of CD160 and CD31 and higher levels of CD55 and, remarkably, a higher expression of tissue-resident markers CD103 and the NK cell decidual marker CD9. Last of all, in individuals with severe disease, we identified an expansion of a CD151brightCD9+ NK cell subset, suggesting that these cells play a specific role in COVID-19. Altogether, our findings suggest that CD151+ NK cells may have a relevant role in COVID-19 immunopathogenesis.
Collapse
Affiliation(s)
| | - Olatz Zenarruzabeitia
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Arrate Sevilla
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Víctor Sandá
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Ainara Lopez-Pardo
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain
| | | | - Raquel Pérez-Garay
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Clinical Analysis Service, Cruces University Hospital, OSI Ezkerraldea-Enkarterri-Cruces, Barakaldo, Spain
| | - Silvia Pérez-Fernández
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Susana Meijide
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Natale Imaz-Ayo
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Eunate Arana-Arri
- Scientific Coordination Facility, Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Laura Amo
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Francisco Borrego
- Immunopathology Group, Biobizkaia Health Research Institute, Barakaldo, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
40
|
Li Y, Zhang X, Yi J, Chen Y, Liang J, Wang L, Ma J, Zhu R, Zhang X, Hu D, Jia Y, Yu X, Wang Y. Synergistic evolution: The dynamic adaptation of SARS-CoV-2 and human protective immunity in the real world. J Infect 2024; 89:106310. [PMID: 39393556 DOI: 10.1016/j.jinf.2024.106310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024]
Abstract
OBJECTIVES SARS-CoV-2 is continually evolving with new variants to evade protective immunity and cause new infections. This study aimed to assess infection-acquired immunity and hybrid immunity against re-infection or severe COVID-19. METHODS During 2020-2023, we collected 890 serum samples from individuals infected with SARS-CoV-2 variants including wild type, D614G, Alpha, Delta, BA.1, BA.2, BA.2.76, BA.5.2, BF.7, XBB, and EG.5. The levels of serum neutralizing antibodies (NAbs) against 18 diverse SARS-CoV-2 variants were determined using a bead-based high-throughput broad neutralizing-antibody assay. RESULTS In the initial wave of the COVID-19 pandemic, >75% of the patients demonstrated robust NAb responses against the ancestral SARS-CoV-2, during a period when vaccines were not yet available. After the emergence of the Omicron variant, the seroprevalence of anti-Omicron NAbs among the patients increased rapidly. By April 2023, when XBB variant was predominant, approximately 80% of the patients demonstrated >50% neutralization against the highly immune-evasive XBB lineages. Three serotypes of SARS-CoV-2, namely non-Omicron, Omicron, and XBB serotypes, were identified, with the strong likelihood of further changes occurring as the virus mutating. Generally, NAbs elicited by a previous serotype could not typically effectively protect against another serotype that emerges later in the evolutionary stages. CONCLUSION Our results firstly demonstrated the synergistic evolution between host immunity and SARS-CoV-2 variants in the real world, which would be helpful to develop future vaccines and public health strategies.
Collapse
Affiliation(s)
- Yunhui Li
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xiaohan Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jingkun Yi
- Department of Biomedical Informatics, State Key Laboratory of Vascular Homeostasis and Remodeling, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yuan Chen
- Department of Clinical Laboratory, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Jing Liang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Li Wang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Jiayue Ma
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Renlong Zhu
- Department of Clinical Laboratory, Peking University Ditan Teaching Hospital, Beijing 100015, China
| | - Xiaomei Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Di Hu
- ProteomicsEra Medical Co., Ltd., Beijing 102206, China
| | - Yan Jia
- ProteomicsEra Medical Co., Ltd., Beijing 102206, China
| | - Xiaobo Yu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing 102206, China.
| | - Yajie Wang
- Department of Clinical Laboratory, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China.
| |
Collapse
|
41
|
Ayako RM, Patel K, Ndede I, Nordgren J, Larrson M, Mining SK. Inflammatory, Hematological, and Biochemical Biomarkers in COVID-19 Patients. Immun Inflamm Dis 2024; 12:e70078. [PMID: 39641395 PMCID: PMC11621974 DOI: 10.1002/iid3.70078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
INTRODUCTION There are few accurate prognostic indications of the illness's development and severity for COVID-19, despite certain biomarkers having been investigated. The unexpected nature of COVID-19's course, which can quickly progress from asymptomatic to life-threatening symptoms, lies at the heart of the disease's intricacy. Predicting SARS-CoV-2 pathogenicity through laboratory biomarkers and as such, identifying the patients' illness severity at the time of their initial admission would be crucial in improving patient care. In this study, we sought to evaluate the potential of hematological, biochemical, and inflammatory biomarkers in predicting the course of COVID-19 at a tertiary hospital in western Kenya. METHODS This cross-sectional study involved 48 COVID-19 patients (16 asymptomatic; 16 moderate symptomatic; and 16 severe symptomatic) and 48 age-sex-matched COVID-19-negative clients attending the Moi Teaching and Referral Hospital, Kenya. Demographic information, self-reported chronic illnesses, symptoms, and laboratory results were collected at recruitment. RESULTS Significantly, the severity of COVID-19 was associated with; hemoglobin (p < 0.0001), white blood cells (p = 0.0022), hematocrit (p < 0.0001), blood urea nitrogen (p = 0.01), blood sodium (p = 0.0002), potassium (p = 0.0483), C-reactive protein (p = 0.0002), and Lactate Dehydrogenase (p < 0.0001). Regression analysis of CRP revealed a strong positive correlation (p = 0.0006) whereas LDH revealed a weak positive correlation (p < 0.0001) with COVID-19 disease severity. Discriminative accuracy was highest when asymptomatic was compared to severe COVID-19 for CRP and LDH (AUC: 0.8867, 95% CI: 0.7532-1.000) and (AUC: 1.000, 95% CI: 1.000-1.000) respectively. CONCLUSION The hematological indices, inflammatory and biochemical biomarkers studied have the potential to predict the course of COVID-19. These parameters may be useful in helping design appropriate care for COVID-19 patients.
Collapse
Affiliation(s)
- Rebeccah M. Ayako
- Department of PathologyMoi UniversityEldoretKenya
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and VirologyLinköping UniversityLinköpingSweden
| | | | - Isaac Ndede
- Department of PathologyMoi UniversityEldoretKenya
| | - Johan Nordgren
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and VirologyLinköping UniversityLinköpingSweden
| | - Marie Larrson
- Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and VirologyLinköping UniversityLinköpingSweden
| | | |
Collapse
|
42
|
Regina Malveste Ito C, Santos MO, de Oliveira Cunha M, de Araújo KM, de Souza GRL, Rézio GS, de Brito PN, Rezende APC, Fonseca JG, Wastowski IJ, Gonçalves Vieira JD, Gomes Avelino MA, Carneiro LC. Rhinovirus infection and co-infection in children with severe acute respiratory infection during the COVID-19 pandemic period. Virulence 2024; 15:2310873. [PMID: 38384141 PMCID: PMC10885176 DOI: 10.1080/21505594.2024.2310873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 01/22/2024] [Indexed: 02/23/2024] Open
Abstract
Rhinovirus causes respiratory tract infections in children and is found in co-infections. The objective of this research was to study the clinical profile of rhinovirus infection and co-infection in children with severe acute respiratory infection (SARI) during the COVID-19 pandemic period. We included 606 children ranging in age from 0.1 to 144 months of age from March 2020 to December 2021, hospitalized in the Pediatric Intensive Care Unit (PICU). The samples were collected by secretion from the nasopharynx region. A total of 259 children were tested positive for viral infection, 153 (59.07%) of them had a single rhinovirus infection and, 56 (36.6%) were aged between 60.1 and 144 months. Nine types of co-infections were identified and were found coinfection with three or more viruses (22/104, 21.15%). Observing the seasonality, the number of cases was similar between 2020 (49.53%) and 2021 (51.47%). Patients with a single infection (86.88%) and coinfection (67.30%) were more likely to have coughed. Patients with co-infection required the use of O2 for longer than those with a single rhinovirus infection. Hemogram results obtained from individuals with a single infection had higher levels of urea when compared to patients with co-infection with and other respiratory viruses. Multiple correspondence analyses indicated different clinical symptoms and comorbidities in patients with co-infection compared to those with single infection. The results found that the rhinovirus was much prevalent virus during the pandemic period and was found in co-infection with other virus types, what is important to diagnostic for the correct treatment of patients.
Collapse
Affiliation(s)
- Célia Regina Malveste Ito
- Microorganism Biotechnology Laboratory of Institute of Tropical Pathology and Public Health, Federal University of Goiás– 235 St. Leste Universitário neighborhood, Goiânia, Goiás, Brazil
| | - Mônica Oliveira Santos
- Microorganism Biotechnology Laboratory of Institute of Tropical Pathology and Public Health, Federal University of Goiás– 235 St. Leste Universitário neighborhood, Goiânia, Goiás, Brazil
| | - Marcos de Oliveira Cunha
- Microorganism Biotechnology Laboratory of Institute of Tropical Pathology and Public Health, Federal University of Goiás– 235 St. Leste Universitário neighborhood, Goiânia, Goiás, Brazil
| | - Kelliane Martins de Araújo
- Microorganism Biotechnology Laboratory of Institute of Tropical Pathology and Public Health, Federal University of Goiás– 235 St. Leste Universitário neighborhood, Goiânia, Goiás, Brazil
| | - Guilherme Rocha Lino de Souza
- Biochemistry and Molecular Biology Laboratory, Biologic Science Institute, Federal University of Goiás, Samambaia Camp, Goiânia, Goiás, Brazil
| | - Geovana Sôffa Rézio
- State Emergency Hospital of the Northwest Region of Goiânia Governador Otávio Lage de Siqueira (HUGOL), Goiânia, Goiás, Brazil
| | - Pollyanna Neta de Brito
- State Emergency Hospital of the Northwest Region of Goiânia Governador Otávio Lage de Siqueira (HUGOL), Goiânia, Goiás, Brazil
| | - Alana Parreira Costa Rezende
- State Emergency Hospital of the Northwest Region of Goiânia Governador Otávio Lage de Siqueira (HUGOL), Goiânia, Goiás, Brazil
| | - Jakeline Godinho Fonseca
- State Emergency Hospital of the Northwest Region of Goiânia Governador Otávio Lage de Siqueira (HUGOL), Goiânia, Goiás, Brazil
| | - Isabela Jubé Wastowski
- Molecular Immunology Laboratory of Goiás State University, Laranjeiras Unity Prof. Alfredo de Castro neighborhood, Goiânia, Goiás, Brazil
| | - José Daniel Gonçalves Vieira
- Microorganism Biotechnology Laboratory of Institute of Tropical Pathology and Public Health, Federal University of Goiás– 235 St. Leste Universitário neighborhood, Goiânia, Goiás, Brazil
| | - Melissa Ameloti Gomes Avelino
- Department of Pediatrics, Federal University of Goiás, Universitaria Avenue, Leste Universitário neighborhood, Goiânia, Goiás, Brazil
| | - Lilian Carla Carneiro
- Microorganism Biotechnology Laboratory of Institute of Tropical Pathology and Public Health, Federal University of Goiás– 235 St. Leste Universitário neighborhood, Goiânia, Goiás, Brazil
| |
Collapse
|
43
|
Koyama A, Li L, Yamamoto T, Taira H, Sugimoto E, Ito Y, Mizuno Y, Awaji K, Tateishi S, Kanda H, Sato S, Shibata S. Psoriasis treatment and biologic switching: The association with clinical characteristics and laboratory biomarkers over a 13-year retrospective study. J Dermatol 2024; 51:1572-1578. [PMID: 39269210 PMCID: PMC11624150 DOI: 10.1111/1346-8138.17465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/29/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024]
Abstract
The advent of biologics has greatly improved patient outcomes, yet some patients are compelled to switch therapies. Predicting these therapeutic failures is important; however, the factors associated with switching biologics have not been fully explored. This study examined patterns and determinants of biologics switching in psoriasis treatment retrospectively over 13 years. We focused on the association between clinical characteristics, basal laboratory data, and frequency of biologics switching. The findings revealed that elevated Psoriasis Area Severity Index scores and the presence of arthritis were observed in patients who experienced two or more treatment switches compared with those without treatment switches. Moreover, neutrophil to lymphocyte ratio was associated with higher biologics switching rates, indicating that systemic inflammation significantly impacts treatment adherence. A treatment approach, taking into account both the clinical presentation and inflammatory biomarkers, may be important for optimizing patient management in psoriasis.
Collapse
Affiliation(s)
- Asumi Koyama
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Lixin Li
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Toyoki Yamamoto
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Haruka Taira
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Eiki Sugimoto
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Yukiko Ito
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Yuka Mizuno
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Kentaro Awaji
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Shoko Tateishi
- Department of Allergy and Rheumatology, Graduate School of MedicineThe University of TokyoTokyoJapan
- Division for Health Service PromotionUniversity of TokyoTokyoJapan
| | - Hiroko Kanda
- Department of Allergy and Rheumatology, Graduate School of MedicineThe University of TokyoTokyoJapan
- Immune‐Mediated Diseases Therapy Center, Graduate School of MedicineUniversity of TokyoTokyoJapan
| | - Shinichi Sato
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Sayaka Shibata
- Department of Dermatology, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
44
|
Salemi N, Shojaie B, Bolourinejad P, Sherkat R, Zamanifar A, Ghaedrahmati F, Azizi M, Aria H. Diversity in the Clinical Course and Outcome of COVID-19 in Patients with Different Inborn Errors of Immunity can be Associated with the Type of Error. Adv Biomed Res 2024; 13:112. [PMID: 39717240 PMCID: PMC11665184 DOI: 10.4103/abr.abr_134_23] [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: 04/17/2023] [Revised: 03/30/2024] [Accepted: 04/30/2024] [Indexed: 12/25/2024] Open
Abstract
Background The relationship between inborn errors of immunity (IEIs) and COVID-19 severity and incidence rates remains unclear due to limited and diverse data. This study aimed to address this gap by identifying specific IEIs associated with an increased risk of severe COVID-19 or a predisposition to severe disease before vaccination. Materials and Methods Data were collected from the medical records of 15 patients with various IEIs, supplemented by interviews with individuals from an IEIs registry who had experienced COVID-19 before vaccination. Results Among the participants, only three patients (20%) experienced severe-prolonged COVID-19. Notably, this severity was predominantly observed in two male patients with Bruton's disease (BD) and one female patient with autosomal recessive hypogammaglobinemia. Moderate and severe COVID-19 cases were equally distributed (13.33%). In the female subgroup, one patient with common variable immunodeficiency and another with combined immunodeficiency experienced moderate and severe COVID-19, respectively. Conversely, both male patients with moderate and severe COVID-19 had BD. Conclusion Despite the limited number of severe cases, the absence of cytokine storm manifestation suggests potential protective mechanisms, possibly due to intravenous immunoglobulin therapy and inherent deficiencies within cytokine-producing cells (B and T cells). While IEIs may not be significant risk factors for COVID-19, they offer promising avenues for further research into therapeutic strategies targeting specific immune system components to mitigate severe COVID-19.
Collapse
Affiliation(s)
- Negin Salemi
- Immunodeficiency Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behrokh Shojaie
- Immunodeficiency Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Paria Bolourinejad
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roya Sherkat
- Immunodeficiency Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Aryana Zamanifar
- Immunodeficiency Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahdieh Azizi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Aria
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Noncommunicable Disease Research Center, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
45
|
Liu Q, Miao H, Shi C, Hu P, An S. Association between the lowest level of serum albumin during hospitalization and adverse outcomes in older adults with COVID-19. Medicine (Baltimore) 2024; 103:e40734. [PMID: 39612427 PMCID: PMC11608705 DOI: 10.1097/md.0000000000040734] [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: 02/20/2024] [Accepted: 11/11/2024] [Indexed: 12/01/2024] Open
Abstract
Serum albumin on admission has been investigated among inpatients with COVID-19. However, studies on the lowest level of serum albumin during hospitalization and adverse outcomes are limited. This research aimed to explore association between them in older adults with COVID-19. A retrospective study was conducted with 300 patients aged 60 or older with first confirmed COVID-19 from January to February 2023. An adverse outcome was defined as development of acute respiratory failure, shock, or death. Data on demographics, comorbidities, laboratory parameters, the initial phase of COVID-19, coinfection, sepsis, receipt of antiviral treatment and outcomes were gathered from the electronic medical records. The association between the lowest level of serum albumin and adverse outcomes was analyzed using univariate and multivariate regression models, along with generalized additive models. After adjusting potential confounders, nonlinear relationship with an inflection point of 29.1 g/L was detected between the lowest level of serum albumin and adverse outcomes in the elderly. The effect sizes and the confidence intervals on the left and right sides of the inflection point were 0.667 (0.520, 0.856) and 1.171 (0.875, 1.568), respectively. This demonstrated that the lowest level of serum albumin was negatively correlated with adverse outcomes when albumin was <29.1 g/L. A rise of 1 unit in the lowest level of albumin equated to a 33.3% decrease in the risk of adverse outcomes. The correlation between the lowest level of serum albumin and adverse outcomes of COVID-19 is a nonlinear. this study indicates that serum albumin levels should be sustained above the critical inflection point identified to reduce the risk of adverse outcomes.
Collapse
Affiliation(s)
- Qiaoli Liu
- Department of Infectious Diseases, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou, Zhejiang, China
| | - Haifeng Miao
- Department of Infectious Diseases, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou, Zhejiang, China
| | - Chunwei Shi
- Department of Infectious Diseases, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou, Zhejiang, China
| | - Piao Hu
- Department of Infectious Diseases, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou, Zhejiang, China
| | - Suhong An
- Department of Radiotherapy, Xiaoshan Affiliated Hospital of Wenzhou Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
46
|
Zhang J, Kim MH, Lee S, Park S. Integration of nanobiosensors into organ-on-chip systems for monitoring viral infections. NANO CONVERGENCE 2024; 11:47. [PMID: 39589620 PMCID: PMC11599699 DOI: 10.1186/s40580-024-00455-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/17/2024] [Indexed: 11/27/2024]
Abstract
The integration of nanobiosensors into organ-on-chip (OoC) models offers a promising advancement in the study of viral infections and therapeutic development. Conventional research methods for studying viral infection, such as two-dimensional cell cultures and animal models, face challenges in replicating the complex and dynamic nature of human tissues. In contrast, OoC systems provide more accurate, physiologically relevant models for investigating viral infections, disease mechanisms, and host responses. Nanobiosensors, with their miniaturized designs and enhanced sensitivity, enable real-time, continuous, in situ monitoring of key biomarkers, such as cytokines and proteins within these systems. This review highlights the need for integrating nanobiosensors into OoC systems to advance virological research and improve therapeutic outcomes. Although there is extensive literature on biosensors for viral infection detection and OoC models for replicating infections, real integration of biosensors into OoCs for continuous monitoring remains unachieved. We discuss the advantages of nanobiosensor integration for real-time tracking of critical biomarkers within OoC models, key biosensor technologies, and current OoC systems relevant to viral infection studies. Additionally, we address the main technical challenges and propose solutions for successful integration. This review aims to guide the development of biosensor-integrated OoCs, paving the way for precise diagnostics and personalized treatments in virological research.
Collapse
Affiliation(s)
- Jiande Zhang
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| | - Min-Hyeok Kim
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| | - Seulgi Lee
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
- Department of Biophysics, Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
| |
Collapse
|
47
|
Qiu M, Song X, Zhang Q, Zou S, Pang L, Nian X. Immunophenotyping characteristics and clinical outcome of COVID-19 patients treated with azvudine during the Omicron surge. Front Immunol 2024; 15:1465238. [PMID: 39654887 PMCID: PMC11625784 DOI: 10.3389/fimmu.2024.1465238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
Background Little is known about immunophenotyping characteristics and clinical outcomes of COVID-19 patients treated with azvudine during the Omicron variant surge. Methods This study enrolled patients diagnosed with COVID-19 from December 2022 to February 2023. The primary outcome was defined as all-cause mortality, along with a composite outcome reflecting disease progression. The enrolled patients were followed for a period of 60 days from their admission. Results A total of 268 COVID-19 patients treated with azvudine were enrolled in this retrospective study. The study found that the counts of lymphocyte subsets were significantly reduced in the composite outcome and all-cause mortality groups compared to the non-composite outcome and discharge groups (all p < 0.001). Correlation analysis revealed a negative association between lymphocyte subsets cell counts and inflammatory markers levels. The receiver operating characteristic (ROC) curve analysis identified low CD4+ T cell count as the most significant predictor of disease progression and all-cause mortality among the various lymphocyte subsets. Additionally, both the Kaplan-Meier curve and multivariate regression analysis demonstrated that low CD4+ T cell count level (< 156.00 cells/μl) was closely associated with all-cause mortality in COVID-19 patients treated with azvudine. Conclusions A low CD4+ T cell count may serve as a significant predictive indicator for identifying COVID-19 patients receiving azvudine treatment who are at an elevated risk of experiencing adverse outcomes. These findings may offer valuable insights for physicians in optimizing the administration of azvudine.
Collapse
Affiliation(s)
- Meihua Qiu
- Department of Respiratory and Critical Care Medicine, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaogang Song
- Department of Respiratory and Critical Care Medicine, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| | - Qianqian Zhang
- Department of Imaging, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| | - Shenchun Zou
- Department of Respiratory and Critical Care Medicine, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| | - Lingling Pang
- Department of Respiratory and Critical Care Medicine, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| | - Xueyuan Nian
- Department of Gastroenterology, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, China
| |
Collapse
|
48
|
Singh G, García-Bernalt Diego J, Warang P, Park SC, Chang LA, Noureddine M, Laghlali G, Bykov Y, Prellberg M, Yan V, Singh S, Pache L, Cuadrado-Castano S, Webb B, García-Sastre A, Schotsaert M. Outcome of SARS-CoV-2 reinfection depends on genetic background in female mice. Nat Commun 2024; 15:10178. [PMID: 39580470 PMCID: PMC11585546 DOI: 10.1038/s41467-024-54334-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 11/06/2024] [Indexed: 11/25/2024] Open
Abstract
Antigenically distinct SARS-CoV-2 variants increase the reinfection risk for vaccinated and previously exposed population due to antibody neutralization escape. COVID-19 severity depends on many variables, including host immune responses, which differ depending on genetic predisposition. To address this, we perform immune profiling of female mice with different genetic backgrounds -transgenic K18-hACE2 and wild-type 129S1- infected with the severe B.1.351, 30 days after exposure to the milder BA.1 or severe H1N1. Prior BA.1 infection protects against B.1.351-induced morbidity in K18-hACE2 but aggravates disease in 129S1. H1N1 protects against B.1.351-induced morbidity only in 129S1. Enhanced severity in B.1.351 re-infected 129S1 is characterized by an increase of IL-10, IL-1β, IL-18 and IFN-γ, while in K18-hACE2 the cytokine profile resembles naïve mice undergoing their first viral infection. Enhanced pathology during 129S1 reinfection cannot be attributed to weaker adaptive immune responses to BA.1. Infection with BA.1 causes long-term differential remodeling and transcriptional changes in the bronchioalveolar CD11c+ compartment. K18-hACE2 CD11c+ cells show a strong antiviral defense expression profile whereas 129S1 CD11c+ cells present a more pro-inflammatory response upon restimulation. In conclusion, BA.1 induces cross-reactive adaptive immune responses in K18-hACE2 and 129S1, but reinfection outcome correlates with differential CD11c+ cells responses in the alveolar space.
Collapse
Affiliation(s)
- Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
| | - Juan García-Bernalt Diego
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
| | - Seok-Chan Park
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
| | - Lauren A Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Moataz Noureddine
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriel Laghlali
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Yonina Bykov
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew Prellberg
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vivian Yan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarabjot Singh
- RT-PCR COVID-19 Laboratory, Civil Hospital, Moga, Punjab, India
| | - Lars Pache
- NCI Designated Cancer Center, Sanford-Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Sara Cuadrado-Castano
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Lipschultz Precision Immunology Institute (PrIISM), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brett Webb
- Department of Veterinary Sciences, University of Wyoming, Laramie, WY, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA.
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA.
- Lipschultz Precision Immunology Institute (PrIISM), Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
49
|
Ai N, Zhang Y, Yang J, Zhang Y, Zhao X, Feng H. Genetically predicted blood metabolites mediate the association between circulating immune cells and severe COVID-19: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40509. [PMID: 39560514 PMCID: PMC11575977 DOI: 10.1097/md.0000000000040509] [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: 08/21/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024] Open
Abstract
Investigating the causal relationship between circulating immune cells, blood metabolites, and severe COVID-19 and revealing the role of blood metabolite-mediated circulating immune cells in disease onset and progression. Genetic variation data of 731 circulating immune cells, 1400 blood metabolites, and severe COVID-19 from genome-wide association study open-access database (https://gwas.mrcieu.ac.uk) were used as instrumental variables for bidirectional and two-step Mendelian randomization analysis. The study identified 11 circulating immune cells with unidirectional causality to severe COVID-19. Two-step Mendelian randomization analysis showed 10 blood metabolites were causally associated with severe COVID-19, and blood Myristate and Citrulline to phosphate ratio mediated the association of circulating effector memory double negative % DN and CD8dim natural killer T cell % T cells, respectively, with severe COVID-19 (Myristate mediated effect ratio was 10.20%, P = .011; Citrulline to phosphate ratio mediated effect ratio was -9.21%, P = .017). This study provides genetic evidence assessing the causal relationship between circulating immune cells, blood metabolites, and severe COVID-19, elucidates the role of blood metabolite-mediated circulating immune cells in severe COVID-19 development, and offers new insights into severe COVID-19 etiology and related preventive and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Ning Ai
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Zhang
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Yang
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Zhang
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuejing Zhao
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huifen Feng
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
50
|
Chen L, Jiang YH, Li MY, Huang B, Yuan L, Wan JH, Qin TY, Zeng TT, Chen QG. The Value of Common Laboratory Markers in Predicting the Severity of COVID-19 Patients. Infect Drug Resist 2024; 17:5037-5047. [PMID: 39559342 PMCID: PMC11572049 DOI: 10.2147/idr.s478798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/26/2024] [Indexed: 11/20/2024] Open
Abstract
Purpose The aim of the present study was to identify more effective laboratory markers to assess the severity of corona virus disease 2019 and predict the progression of the disease by collecting more laboratory markers and variables. Patients and Methods In this study, most risk factors, including epidemiological characteristics, blood cell counts, cytokines, and infection markers, were collected from 126 patients with COVID-19 to assess their predictive value. Results The area under curve (AUC) of Albumin (Alb) to fibrinogen (Fib) ratio (AFR) (0.791), Lactate dehydrogenase (LDH) (0.792), myoglobin (MYO) (0.795), C-reactive protein (CRP) (0.801) and lymphocyte count (0.859) were higher than other markers to distinguish severe from non-severe patients in receiver operating characteristic (ROC) analysis. In the univariate logistic regression analysis, thirty-six out of 46 risk factors, including 34 laboratory markers, were significantly associated with increased odds of severe patients. Multivariate logistic regression analysis showed that the CD19+ lymphocyte count, MYO, LDH, and AFR were associated with increased odds of severe disease. Moreover, Lymphocyte count and AFR levels increased, LDH and CRP levels decreased during hospitalization in recovered severe patients, whereas severe lymphocytopenia and continuously increasing LDH levels were observed in deteriorated patients. AFR level increased and CRP level decreased before the disease worsened in the deteriorated patients; however, when the patients deteriorated, AFR decreased and CRP increased significantly. Conclusion CD19+ lymphocyte count, MYO, LDH, and AFR are independent biomarkers for early identification of severe COVID-19. Lymphocyte count, AFR, LDH, and CRP levels were helpful in predicting the clinical progression of the disease.\.
Collapse
Affiliation(s)
- Lian Chen
- Department of Ultrasound Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Yu-Huan Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Mei-Yong Li
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Bo Huang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Lei Yuan
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Jin-Hua Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Ting-Yu Qin
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Ting-Ting Zeng
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Qing-Gen Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| |
Collapse
|