1
|
Takahashi H, Satake Y, Shimizu S, Fujihara S, Takano S, Fukasawa S, Park K, Toba N, Yano T, Nagamatsu H, Hirose R, Toyama-Kousaka M, Ota S, Morikawa M, Shinkai M. Trends in Group A Streptococcus Pharyngitis and Co-Infection with Severe Acute Respiratory Syndrome Coronavirus 2: A Retrospective Observational Study. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:937. [PMID: 40428896 PMCID: PMC12113336 DOI: 10.3390/medicina61050937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 05/02/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025]
Abstract
Background and Objectives: Group A Streptococcus (GAS) is a leading cause of acute pharyngitis with seasonal outbreaks. The coronavirus disease 2019 (COVID-19) pandemic significantly altered respiratory infection trends; however, its impact on GAS pharyngitis (GAS-P) incidence remains unclear. Additionally, data on co-infections with GAS and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are limited. In this study, temporal trends in GAS-P incidence and characteristics of GAS-SARS-CoV-2 co-infections in Japan were examined. Materials and Methods: In this observational study, data from patients who visited the Tokyo Shinagawa Hospital between January 2019 and December 2024 were retrospectively analyzed. Data on GAS and SARS-CoV-2 test results and patient demographics were extracted from medical records. The study period was categorized based on COVID-19-related public health measures as follows: pre-COVID-19 social period (January 2019-April 2020), restricted social period (May 2020-April 2023), and post-restriction period (May 2023-December 2024). GAS incidence stratified by sex, age, and period was calculated. Clinical characteristics of patients co-infected with GAS and SARS-CoV-2 were analyzed. Results: Among 4837 GAS tests, 463 (9.6%) were positive. GAS positivity rates varied significantly: 11.4% (pre-COVID-19), 7.1% (restricted social period), and 12.6% (post-restriction period; p < 0.001). The proportion of pediatric cases decreased significantly during the restricted social period (24.8-5.3%) before rising sharply in the post-restriction period (47.1%, p < 0.001). Among 151 patients tested for GAS and SARS-CoV-2, 14 (9.3%) had co-infections, which were identified exclusively after July 2022. Most patients exhibited mild symptoms, primarily fever and sore throat, with decreased lymphocyte counts despite normal white blood cell counts. Conclusions: In our cohort, the incidence of GAS pharyngitis temporarily declined during COVID-19-related public health measures and subsequently increased, particularly among children, after restrictions were lifted. Limited testing may contribute to the underdiagnosis of GAS-SARS-CoV-2 co-infections. Further large-scale studies are warranted to assess microbial interactions, disease severity, and long-term outcomes.
Collapse
Affiliation(s)
- Hidenori Takahashi
- Department of Respiratory Medicine, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan (N.T.); (S.O.)
- Department of Infection Control, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan; (S.S.); (S.F.); (K.P.)
| | - Yugo Satake
- Department of Respiratory Medicine, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan (N.T.); (S.O.)
| | - Saori Shimizu
- Department of Infection Control, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan; (S.S.); (S.F.); (K.P.)
| | - Satomi Fujihara
- Department of Infection Control, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan; (S.S.); (S.F.); (K.P.)
| | - Syunsuke Takano
- Department of Infection Control, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan; (S.S.); (S.F.); (K.P.)
| | - Suzuko Fukasawa
- Department of Infection Control, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan; (S.S.); (S.F.); (K.P.)
| | - Kaeyong Park
- Department of Infection Control, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan; (S.S.); (S.F.); (K.P.)
| | - Naoya Toba
- Department of Respiratory Medicine, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan (N.T.); (S.O.)
- Department of Infection Control, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan; (S.S.); (S.F.); (K.P.)
| | - Takahiko Yano
- Department of Infection Control, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan; (S.S.); (S.F.); (K.P.)
| | - Hiroki Nagamatsu
- Department of Respiratory Medicine, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan (N.T.); (S.O.)
| | - Ryutaro Hirose
- Department of Respiratory Medicine, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan (N.T.); (S.O.)
| | - Mio Toyama-Kousaka
- Department of Respiratory Medicine, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan (N.T.); (S.O.)
| | - Shinichiro Ota
- Department of Respiratory Medicine, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan (N.T.); (S.O.)
- Department of Infection Control, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan; (S.S.); (S.F.); (K.P.)
| | - Miwa Morikawa
- Department of Respiratory Medicine, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan (N.T.); (S.O.)
| | - Masaharu Shinkai
- Department of Respiratory Medicine, Tokyo Shinagawa Hospital, Tokyo 140-8522, Japan (N.T.); (S.O.)
| |
Collapse
|
2
|
Ghosh S, Das Sarma J. The age-dependent neuroglial interaction with peripheral immune cells in coronavirus-induced neuroinflammation with a special emphasis on COVID-19. Biogerontology 2025; 26:111. [PMID: 40380990 DOI: 10.1007/s10522-025-10252-9] [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: 02/28/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025]
Abstract
Neurodegenerative diseases are chronic progressive disorders that impair memory, cognition, and motor functions, leading to conditions such as dementia, muscle weakness, and speech difficulties. Aging disrupts the stringent balance between pro- and anti-inflammatory cytokines, increasing neuroinflammation, which contributes to neurodegenerative diseases. The aging brain is particularly vulnerable to infections due to a weakened and compromised immune response and impaired integrity of the blood-brain barrier, allowing pathogens like viruses to trigger neurodegeneration. Coronaviruses have been linked to both acute and long-term neurological complications, including cognitive impairments, psychiatric disorders, and neuroinflammation. The virus can induce a cytokine storm, damaging the central nervous system (CNS) and worsening existing neurological conditions. Though its exact mechanism of neuroinvasion remains elusive, evidence suggests it disrupts the blood-brain barrier and triggers immune dysregulation, leading to persistent neurological sequelae in elderly individuals. This review aims to understand the interaction between the peripheral immune system and CNS glial cells in aged individuals, which is imperative in addressing coronavirus-induced neuroinflammation and concomitant neurodegeneration.
Collapse
Affiliation(s)
- Satavisha Ghosh
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohanpur, Kolkata, 741246, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohanpur, Kolkata, 741246, India.
- Department of Ophthalmology, University of Pennsylvania, 19104, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Nehar-Belaid D, Mejías A, Xu Z, Marches R, Yerrabelli R, Chen G, Mertz S, Ye F, Sánchez PJ, Tsang JS, Aydillo T, Miorin L, Cupic A, García-Sastre A, Ucar D, Banchereau JF, Pascual V, Ramilo O. SARS-CoV-2 induced immune perturbations in infants vary with disease severity and differ from adults' responses. Nat Commun 2025; 16:4562. [PMID: 40379618 PMCID: PMC12084365 DOI: 10.1038/s41467-025-59411-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 04/22/2025] [Indexed: 05/19/2025] Open
Abstract
Differences in immune profiles of children and adults with COVID-19 have been previously described. However, no systematic studies have been reported from infants hospitalized with severe disease. We applied a multidimensional approach to decipher the immune responses of SARS-CoV-2 infected infants (n = 26; 10 subacute, 11 moderate and 5 severe disease; median age = 1.6 months) and matched controls (n = 14; median age = 2 months). Single cell (scRNA-seq) profiling of PBMCs revealed substantial alterations in cell composition in SARS-CoV-2 infected infants; with most cell-types switching to an interferon-stimulated gene (ISGhi) state including: (i) CD14+ monocytes co-expressing ISGs and inflammasome-related molecules, (ii) ISGhi naive CD4+ T cells, (iii) ISGhi proliferating cytotoxic CD8+ T cells, and (iv) ISGhi naive and transitional B cells. We observe increased serum concentrations of both interferons and inflammatory cytokines in infected infants. Antibody responses to SARS-CoV-2 are also consistently detected in the absence of anti-IFN autoantibodies. Compared with infected adults, infants display a similar ISG signature in monocytes but a markedly enhanced ISG signature in T and B cells. These findings provide insights into the distinct immune responses to SARS-CoV-2 in the first year of life and underscore the importance of further defining the unique features of early life immunity.
Collapse
Affiliation(s)
| | - Asunción Mejías
- Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Zhaohui Xu
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Radu Marches
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Rushil Yerrabelli
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Guo Chen
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Sara Mertz
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Fang Ye
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Pablo J Sánchez
- Department of Pediatrics, Division of Neonatology and Center for Perinatal Research, Ohio Perinatal Research Network, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - John S Tsang
- Center for Systems and Engineering Immunology, Departments of Immunobiology and Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Chan Zuckerberg Biohub NY, New Haven, CT, USA
| | - Teresa Aydillo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Lisa Miorin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Anastasija Cupic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Pathology, Molecular and Cell-Based Medicine, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Jacques F Banchereau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA.
- Immunoledge LLC, Montclair, NJ, USA.
| | - Virginia Pascual
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| | - Octavio Ramilo
- Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA.
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA.
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
4
|
Anderson W, Gould R, Patil N, Mohr N, Dodd K, Boyce D, Dasher P, Guerin PJ, Khan R, Cheruku S, Kumar VK, Mathé E, Mehta AK, Michelson AP, Williams A, Heavner SF, Podichetty JT. Unveiling sub-populations in critical care settings: a real-world data approach in COVID-19. Front Public Health 2025; 13:1544904. [PMID: 40443932 PMCID: PMC12119499 DOI: 10.3389/fpubh.2025.1544904] [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: 12/13/2024] [Accepted: 03/31/2025] [Indexed: 06/02/2025] Open
Abstract
Background Disease presentation and progression can vary greatly in heterogeneous diseases, such as COVID-19, with variability in patient outcomes, even within the hospital setting. This variability underscores the need for tailored treatment approaches based on distinct clinical subgroups. Objectives This study aimed to identify COVID-19 patient subgroups with unique clinical characteristics using real-world data (RWD) from electronic health records (EHRs) to inform individualized treatment plans. Materials and methods A Factor Analysis of Mixed Data (FAMD)-based agglomerative hierarchical clustering approach was employed to analyze the real-world data, enabling the identification of distinct patient subgroups. Statistical tests evaluated cluster differences, and machine learning models classified the identified subgroups. Results Three clusters of COVID-19 in patients with unique clinical characteristics were identified. The analysis revealed significant differences in hospital stay durations and survival rates among the clusters, with more severe clinical features correlating with worse prognoses and machine learning classifiers achieving high accuracy in subgroup identification. Conclusion By leveraging RWD and advanced clustering techniques, the study provides insights into the heterogeneity of COVID-19 presentations. The findings support the development of classification models that can inform more individualized and effective treatment plans, improving patient outcomes in the future.
Collapse
Affiliation(s)
| | - Ruth Gould
- Centers of Disease Control and Prevention, Atlanta, GA, United States
| | - Namrata Patil
- Brigham and Women’s Hospital, Boston, MA, United States
| | - Nicholas Mohr
- University of Iowa Healthcare, Iowa City, IA, United States
| | - Kenneth Dodd
- Advocate Aurora Health, Downers Grove, IL, United States
| | - Danielle Boyce
- Tufts University School of Medicine, Boston, MA, United States
- Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Pam Dasher
- Critical Path Institute, Tucson, AZ, United States
| | | | - Reham Khan
- Society of Critical Care Medicine, Mount Prospect, IL, United States
| | - Sreekanth Cheruku
- Department of Anesthesiology and Pain Management, University of Texas Southwestern, Dallas, TX, United States
| | - Vishakha K. Kumar
- Society of Critical Care Medicine, Mount Prospect, IL, United States
| | - Ewy Mathé
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, United States
| | - Aneesh K. Mehta
- Department of Medicine, Emory University, Atlanta, GA, United States
| | - Andrew P. Michelson
- Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Andrew Williams
- Tufts University School of Medicine, Boston, MA, United States
| | - Smith F. Heavner
- Critical Path Institute, Tucson, AZ, United States
- Department of Public Health Sciences, Clemson University, Clemson, SC, United States
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC, United States
| | | |
Collapse
|
5
|
Zeidan RK, Al-Bluwi N, AlZubaidi H, Awad M, Hussein A, AlHajjaj M, Saddik B. The predictive role of inflammation indices derived from complete blood count in severe COVID-19 patients: a study from the United Arab Emirates. Front Med (Lausanne) 2025; 12:1565616. [PMID: 40438363 PMCID: PMC12116627 DOI: 10.3389/fmed.2025.1565616] [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: 01/23/2025] [Accepted: 04/29/2025] [Indexed: 06/01/2025] Open
Abstract
Purpose To investigate the predictive effect and determine the cut-off values of complete blood count parameters in severe COVID-19 hospitalized patients in the United Arab Emirates. Methods A retrospective observational analytical study analyzed data from 738 medical records of COVID-19 hospitalized patients across several healthcare centers in the United Arab Emirates between 29 January 2020 and 14 October 2021. Complete blood count ratios and indices on admission were evaluated for COVID-19 severity using receiver operating characteristic curves, sensitivity, and specificity. Results Main complete blood count-based ratios and indices significantly predicting severe COVID-19 were elevated ratios index (optimal cut-off point ≥3; AOR = 2.8, 95% CI: 1.77-4.42), systemic immune-inflammation index (≥1259.95; AOR = 2.4, 95% CI: 1.53-3.87), systemic inflammation response index (≥3.96; AOR = 2.9, 95% CI: 1.79-4.72), aggregate index of systemic inflammation (≥949.02; AOR = 2.3, 95% CI: 1.43-3.77), platelet-to-lymphocyte ratio (≥188.91; AOR = 2.2, 95% CI: 1.39-3.53), derived neutrophil-to-lymphocyte ratio (≥2.91; AOR = 3.0, 95% CI: 1.84-4.87), and neutrophil-to-lymphocyte ratio (≥6.01; AOR = 3.2, 95% CI: 1.98-5.12). Conclusion Identifying hematological markers' predictive effects and their cut-off values can aid healthcare providers in risk classification and the development of tailored treatment plans. It can also provide cheap, quick, and easy guidance for surveillance systems to lessen the impact of any future outbreaks.
Collapse
Affiliation(s)
- Rouba Karen Zeidan
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Najlaa Al-Bluwi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Hamzah AlZubaidi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- School of Medicine, Deakin Rural Health, Deakin University Faculty of Health, Warrnambool, VIC, Australia
| | - Manal Awad
- Department of Orthodontics, Pediatric and Community Dentistry, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Amal Hussein
- Department of Family and Community Medicine and Behavioral Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Mohamed AlHajjaj
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Basema Saddik
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Department of Family and Community Medicine and Behavioral Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- School of Population Health, University of NSW, Sydney, NSW, Australia
| |
Collapse
|
6
|
Liu S, Xu W, Tu B, Xiao Z, Li X, Huang L, Yuan X, Zhou J, Yang X, Yang J, Chang D, Chen W, Wang FS. Early Immunological and Inflammation Proteomic Changes in Elderly COVID-19 Patients Predict Severe Disease Progression. Biomedicines 2025; 13:1162. [PMID: 40426989 PMCID: PMC12108611 DOI: 10.3390/biomedicines13051162] [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: 03/31/2025] [Revised: 04/28/2025] [Accepted: 05/01/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Elderly patients infected with SARS-CoV-2 are at higher risk of developing cytokine storm and severe outcomes; however, specific immunological and proteomic biomarkers for early prediction remain unclear in this vulnerable group. Methods: We enrolled 182 elderly COVID-19 patients from the Chinese PLA General Hospital between November 2022 and April 2023, categorizing them based on progression to respiratory failure requiring mechanical ventilation (defined as severe progression). Olink proteomic analysis was performed on admission serum from 40 propensity score-matched samples, with differentially expressed proteins (DEPs) validated by cytometric bead array (CBA) in 178 patients. To predict severe progression, a model was developed using a 70% training set and validated on a 30% validation set. LASSO regression screened features followed by logistic regression and receiver operating characteristic (ROC) analysis to optimize the model by incrementally incorporating features ranked by random forest importance. Results: Elderly patients progressing to severe COVID-19 exhibited early immune dysregulation, including neutrophilia, lymphopenia, monocytopenia, elevated procalcitonin (PCT), C-reactive protein (CRP), interleukin-6 (IL-6), neutrophil-to-lymphocyte ratio (NLR), and systemic immune-inflammation index (SII), as well as coagulation dysfunction and multi-organ injury. Proteomics identified a set of biomarkers, including tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), and revealed disruptions in signaling pathways, including the mTOR and VEGF signaling pathways. The optimal predictive model, which incorporated PCT, IL-6, monocyte percentage, lymphocyte count, and TRAIL, achieved an area under curve (AUC) of 0.870 (0.729-1.000) during validation. TRAIL levels negatively correlated with fibrinogen (p < 0.05). Conclusions: Elderly COVID-19 patients with severe progression demonstrate early immune dysregulation, hyperinflammation, coagulation dysfunction, and multi-organ injury. The model we proposed effectively predicts disease progression in elderly COVID-19 patients, providing potential biomarkers for early clinical risk stratification in this vulnerable population.
Collapse
Affiliation(s)
- Shiyang Liu
- Medical School of Chinese PLA, Beijing 100853, China;
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Wen Xu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Bo Tu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Zhiqing Xiao
- Department of Pulmonary and Critical Care Medicine at The Seventh Medical Center, College of Pulmonary and Critical Care Medicine of The Eighth Medical Center, Chinese PLA General Hospital, Beijing 100007, China; (Z.X.); (D.C.)
| | - Xue Li
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
- Yu-Yue Pathology Scientific Research Center, Chongqing 401329, China
| | - Lei Huang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Xin Yuan
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Juanjuan Zhou
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Xinxin Yang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Junlian Yang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - De Chang
- Department of Pulmonary and Critical Care Medicine at The Seventh Medical Center, College of Pulmonary and Critical Care Medicine of The Eighth Medical Center, Chinese PLA General Hospital, Beijing 100007, China; (Z.X.); (D.C.)
| | - Weiwei Chen
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100073, China; (W.X.); (B.T.); (X.L.); (L.H.); (X.Y.); (J.Z.); (X.Y.); (J.Y.)
| |
Collapse
|
7
|
Djuric M, Nenadic I, Radisavljevic N, Todorovic D, Dimic N, Bobos M, Bojic S, Savic P, Turnic TN, Stevanovic P, Djukic V. Severe COVID-19 in Non-Smokers: Predictive Factors and Outcomes. Healthcare (Basel) 2025; 13:1041. [PMID: 40361818 PMCID: PMC12071376 DOI: 10.3390/healthcare13091041] [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: 03/19/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Background: The COVID-19 pandemic revealed an unexpected pattern known as the "smoker's paradox", with lower rates of severe disease among smokers compared to non-smokers, highlighting the need for the specific investigation of disease progression in non-smoking populations. Objective: To identify early mortality predictors in non-smoking patients with severe COVID-19 through the evaluation of clinical, laboratory, and oxygenation parameters. Methods: This retrospective observational cohort study included 59 non-smokers hospitalized with COVID-19 between November and December 2020. Clinical parameters, laboratory findings, and respiratory support requirements were analyzed on Days 1 and 7 of hospitalization. ROC curves were constructed to assess the predictive value of the parameters. Results: The overall mortality rate was 54.2%. The seventh-day SOFA score showed the strongest predictive value (AUC = 0.902, p = 0.004), followed by pCO2 (AUC = 0.853, p = 0.012). Significant differences between survivors and non-survivors were observed in acid-base parameters, oxygenation indices, and hematological markers. Mortality rates varied significantly with ventilation type: 84.6% for IMV and 50% for NIMV, with no deaths in HFNC patients. Conclusions: Multiple parameters measured on Day 7 of hospitalization demonstrate significant predictive value for mortality in non-smoking COVID-19 patients, with the SOFA score being the strongest predictor. The type of respiratory support significantly influences outcomes, suggesting the importance of careful ventilation strategy selection.
Collapse
Affiliation(s)
- Marko Djuric
- Clinic for Anesthesiology and Intensive Care, University Clinical Hospital Center “Dr Dragisa Misovic-Dedinje”, No. 1, Str. Heroja Milana Tepica, 11030 Belgrade, Serbia; (I.N.); (N.D.); (M.B.); (S.B.); (P.S.)
- Department of Anesthesiology, Reanimatology and Intensive Care, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Irina Nenadic
- Clinic for Anesthesiology and Intensive Care, University Clinical Hospital Center “Dr Dragisa Misovic-Dedinje”, No. 1, Str. Heroja Milana Tepica, 11030 Belgrade, Serbia; (I.N.); (N.D.); (M.B.); (S.B.); (P.S.)
- Department of Anesthesiology, Reanimatology and Intensive Care, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Nina Radisavljevic
- Institute of Medical Physiology ”Richard Burian”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (N.R.); (D.T.)
| | - Dusan Todorovic
- Institute of Medical Physiology ”Richard Burian”, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (N.R.); (D.T.)
| | - Nemanja Dimic
- Clinic for Anesthesiology and Intensive Care, University Clinical Hospital Center “Dr Dragisa Misovic-Dedinje”, No. 1, Str. Heroja Milana Tepica, 11030 Belgrade, Serbia; (I.N.); (N.D.); (M.B.); (S.B.); (P.S.)
- Department of Anesthesiology, Reanimatology and Intensive Care, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Marina Bobos
- Clinic for Anesthesiology and Intensive Care, University Clinical Hospital Center “Dr Dragisa Misovic-Dedinje”, No. 1, Str. Heroja Milana Tepica, 11030 Belgrade, Serbia; (I.N.); (N.D.); (M.B.); (S.B.); (P.S.)
- Department of Anesthesiology, Reanimatology and Intensive Care, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Suzana Bojic
- Clinic for Anesthesiology and Intensive Care, University Clinical Hospital Center “Dr Dragisa Misovic-Dedinje”, No. 1, Str. Heroja Milana Tepica, 11030 Belgrade, Serbia; (I.N.); (N.D.); (M.B.); (S.B.); (P.S.)
- Department of Anesthesiology, Reanimatology and Intensive Care, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Predrag Savic
- Clinic for Surgery, University Clinical Hospital Center “Dr Dragisa Misovic-Dedinje”, 11030 Belgrade, Serbia; (P.S.); (V.D.)
- Department of Surgery, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Tamara Nikolic Turnic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
- N.A. Semashko Public Health and Healthcare Department, F.F. Erismann Institute of Public Health, I.M. Schenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Predrag Stevanovic
- Clinic for Anesthesiology and Intensive Care, University Clinical Hospital Center “Dr Dragisa Misovic-Dedinje”, No. 1, Str. Heroja Milana Tepica, 11030 Belgrade, Serbia; (I.N.); (N.D.); (M.B.); (S.B.); (P.S.)
- Department of Anesthesiology, Reanimatology and Intensive Care, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Vladimir Djukic
- Clinic for Surgery, University Clinical Hospital Center “Dr Dragisa Misovic-Dedinje”, 11030 Belgrade, Serbia; (P.S.); (V.D.)
- Department of Surgery, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
8
|
Liao M, Zhang LT, Bai LJ, Wang RY, Liu Y, Han J, Liu LH, Qi BL. Xuebijing injection reduces COVID-19 patients' mortality as influenced by the neutrophil to lymphocyte platelet ratio. JOURNAL OF INTEGRATIVE MEDICINE 2025; 23:282-288. [PMID: 40251040 DOI: 10.1016/j.joim.2025.04.002] [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: 05/22/2024] [Accepted: 01/21/2025] [Indexed: 04/20/2025]
Abstract
OBJECTIVE Xuebijing injection has been recommended as a therapeutic approach for individuals with severe and critical COVID-19. This study aims to explore the correlation of neutrophil to lymphocyte platelet ratio (NLPR) with the severity and prognosis of COVID-19, and the effect of XBJ on the prognosis of patients with COVID-19 in different inflammatory states. METHODS This was a retrospective study conducted at Wuhan Union Hospital in China. COVID-19 patients admitted between November 1, 2022 and February 1, 2023 were included. In predicting prognosis for individuals with COVID-19, new inflammatory indicators were used, and their prognostic value was assessed by using Cox regression models and receiver operating characteristic curves. Furthermore, a calculation was made to determine the cutoff value for NLPR. Relative risk and Cox regression models were used to examine the effects of Xuebijing injection on prognosis in patient cohorts that had been stratified by the NLPR cutoff. RESULTS This research included 455 participants with COVID-19, with a mean age of 72 years. Several inflammatory indicators were found to be strongly correlated with prognosis, and NLPR shows the greatest predictive power. Patients with NLPR > 3.29 exhibited a mortality rate of 17.3%, which was 6.2 times higher than in patients with NLPR ≤ 3.29. Importantly, providing Xuebijing injection to patients with NLPR > 3.29 was associated with a lower risk of 60-day all-cause mortality. However, there was no discernible improvement in survival among patients with NLPR ≤ 3.29 who received Xuebijing injection. CONCLUSION NLPR is the most reliable inflammatory marker for predicting prognosis among individuals with COVID-19, and can accurately identify individuals who may benefit from Xuebijing injection. Please cite this article as: Liao M, Zhang LT, Bai LJ, Wang RY, Liu Y, Han J, Liu LH, Qi BL. Xuebijing injection reduces COVID-19 patients mortality as influenced by the neutrophil to lymphocyte platelet ratio. J Integr Med. 2025; 23(3): 282-288.
Collapse
Affiliation(s)
- Man Liao
- Department of General Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Li-Ting Zhang
- Department of General Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Li-Juan Bai
- Department of General Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Rui-Yun Wang
- Department of General Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Yun Liu
- Department of General Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Jing Han
- Department of General Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Li-Hua Liu
- Department of General Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Ben-Ling Qi
- Department of General Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| |
Collapse
|
9
|
Wu Y, Serna R, Gan W, Fan Z. Different patterns of leukocyte immune responses to infection of ancestral SARS-CoV-2 and its variants. Front Cell Infect Microbiol 2025; 15:1508120. [PMID: 40313462 PMCID: PMC12043629 DOI: 10.3389/fcimb.2025.1508120] [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: 10/08/2024] [Accepted: 03/27/2025] [Indexed: 05/03/2025] Open
Abstract
Background Contributions of leukocytes to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) defense have been reported extensively. However, it remains unclear whether there are different leukocyte responses to ancestral SARS-CoV-2 and its variants. Methods We analyzed peripheral blood leukocyte and subtype concentrations from 575 COVID-19 patients and 950 non-COVID-19 subjects registered at the University of Connecticut John Dempsey Hospital between 2020 and 2022, which covers the ancestral strain, Delta, and Omicron variants. Results We found that neutrophils, immature granulocytes, and monocytes were elevated, and lymphocytes were reduced after infection. These hyperactive neutrophils/immature granulocytes and suppressed lymphocytes/monocytes were associated with poorer prognosis in ancestral strain infection. Different from the ancestral strain, hyperactive immature granulocytes were not shown in the decedents of Delta infection, and immature granulocyte concentration was not observed to be associated with mortality. In Omicron infection, suppressed lymphocytes and monocytes were not shown in the decedents, and lymphocyte/monocyte concentrations were not associated with mortality. Conclusions Our findings provided insights into different leukocyte immune responses to ancestral SARS-CoV-2, Delta, and Omicron variants.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Raphael Serna
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Wenqi Gan
- Department of Public Health Sciences, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Zhichao Fan
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
10
|
Mihuta C, Socaci A, Hogea P, Tudorache E, Mihuta MS, Oancea C. Comparative Insights into COVID-19 and Tuberculosis: Clinical Manifestations, Inflammatory Markers, and Outcomes in Pulmonary Versus Extrapulmonary Tuberculosis and SARS-CoV-2 Co-Infection. J Clin Med 2025; 14:2782. [PMID: 40283612 PMCID: PMC12028324 DOI: 10.3390/jcm14082782] [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: 03/22/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Tuberculosis and COVID-19 co-infection poses significant clinical challenges, with pulmonary TB (PTB) and extrapulmonary TB (extraPTB) potentially influencing disease progression and outcomes differently. This study aims to compare the clinical manifestations, inflammatory markers, and outcomes between PTB and extraPTB patients with SARS-CoV-2 co-infection. Methods: A retrospective, cross-sectional study was conducted on 55 hospitalized adults with TB-COVID-19 co-infection from March 2020 to March 2022. Patients were divided into PTB (n = 32) and extraPTB (n = 23) groups. Demographic, clinical, laboratory, and imaging data were collected and analyzed using statistical models, including ANCOVA, LASSO regression, and Random Forest classification, to identify key predictors of hospitalization duration and mortality. Results: PTB patients had significantly lower BMI, worse oxygenation status, and greater lung involvement on CT compared to extraPTB patients. CRP was elevated in PTB, while IL-6 levels were higher in extraPTB. Hospitalization duration was primarily influenced by inflammatory and coagulation markers (IL-6, D-dimer, neutrophil count, systemic inflammatory index), while higher BMI was associated with shorter stays. Mortality risk was strongly correlated with oxygenation impairment (worst SpO2, SpO2 at diagnosis), inflammatory burden (CRP, LDH), and CT severity score, rather than TB localization. Conclusions: TB localization did not independently affect hospitalization duration or mortality risk. Instead, severe lung involvement, systemic inflammation, and hypoxemia were the strongest predictors of poor outcomes. These findings emphasize the importance of early risk stratification based on respiratory and inflammatory markers to optimize patient management. Further research is needed to clarify the long-term impact of TB-COVID-19 co-infection, particularly in extraPTB cases.
Collapse
Affiliation(s)
- Camil Mihuta
- Department of Doctoral Studies, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Clinical Hospital for Infectious Diseases and Pneumology “Dr. Victor Babes”, 300041 Timisoara, Romania; (P.H.); (E.T.); (C.O.)
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases (CRIPMRD), “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Adriana Socaci
- Clinical Hospital for Infectious Diseases and Pneumology “Dr. Victor Babes”, 300041 Timisoara, Romania; (P.H.); (E.T.); (C.O.)
- Department of Biology and Life Sciences, Faculty of Medicine, “Vasile Goldis” Western University of Arad, 310025 Arad, Romania
| | - Patricia Hogea
- Clinical Hospital for Infectious Diseases and Pneumology “Dr. Victor Babes”, 300041 Timisoara, Romania; (P.H.); (E.T.); (C.O.)
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases (CRIPMRD), “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Department of Pulmonology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Emanuela Tudorache
- Clinical Hospital for Infectious Diseases and Pneumology “Dr. Victor Babes”, 300041 Timisoara, Romania; (P.H.); (E.T.); (C.O.)
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases (CRIPMRD), “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Department of Pulmonology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Monica Simina Mihuta
- Department of Pediatrics, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Center of Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Cristian Oancea
- Clinical Hospital for Infectious Diseases and Pneumology “Dr. Victor Babes”, 300041 Timisoara, Romania; (P.H.); (E.T.); (C.O.)
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases (CRIPMRD), “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Department of Pulmonology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
11
|
Lana FCB, Marinho CC, de Paiva BBM, Valle LR, do Nascimento GF, da Rocha LCD, Carneiro M, Batista JDL, Anschau F, Paraiso PG, Bartolazzi F, Cimini CCR, Schwarzbold AV, Rios DRA, Gonçalves MA, Marcolino MS. Unraveling relevant cross-waves pattern drifts in patient-hospital risk factors among hospitalized COVID-19 patients using explainable machine learning methods. BMC Infect Dis 2025; 25:537. [PMID: 40234758 PMCID: PMC12001466 DOI: 10.1186/s12879-025-10766-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/07/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Several studies explored factors related to adverse clinical outcomes among COVID-19 patients but lacked analysis of the impact of the temporal data shifts on the strength of association between different predictors and adverse outcomes. This study aims to evaluate factors related to patients and hospitals in the prediction of in-hospital mortality, need for invasive mechanical ventilation (IMV), and intensive care unit (ICU) transfer throughout the pandemic waves. METHODS This multicenter retrospective cohort included COVID-19 patients from 39 hospitals, from March/2020 to August/2022. The pandemic was divided into waves: 10/03/2020-14/11/2020 (first), 15/11/2020-25/12/2021 (second), 26/12/2021-03/08/2022 (third). Patient-related factors included clinical, demographic, and laboratory data, while hospital-related factors covered funding sources, accreditation, academic status, and socioeconomic characteristics. Shapley additive explanation (SHAP) values derived from the predictions of a light gradient-boosting machine (LightGBM) model were used to assess potential risk factors for death, IMV and ICU. RESULTS Overall, 16,958 adult patients were included (median age 59 years, 54.7% men). LightGBM achieved competitive effectiveness metrics across all periods. Temporal drifts were observed due to a decrease in various metrics, such as the recall for the positive class [ICU: 0.4211 (wave 1) to 0.1951 (wave 3); IMV: 0.2089 (wave 1) to 0.0438 (wave 3); death: 0.2711 (wave 1) to 0.1175 (wave 3)]. Peripheral arterial oxygen saturation to the fraction of inspired oxygen ratio (SatO2/FiO2) at admission had great predictive capacity for all outcomes, with an optimal cut-off value for death prediction of 227.78. Lymphopenia had its association strength increased over time for all outcomes, optimal threshold for death prediction of 643 × 109/L. Thrombocytopenia was the most important feature in wave 2 (ICU); overall, values below 143,000 × 109/L were more related to death. CONCLUSION Data drifts were observed in all scenarios, affecting potential predictive capabilities of explainable machine learning methods. Upon admission, SatO2/FiO2 values, platelet and lymphocyte count were significant predictors of adverse outcomes in COVID-19 patients. Overall, inflammatory response markers were more important than clinical characteristics. Limitations included sample representativeness and confounding factors. Integrating the drift's knowledge into models to improve effectiveness is a challenge, requiring continuous updates and monitoring of performance in real-world applications. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
| | - Carolina Coimbra Marinho
- Department of Internal Medicine, Medical School & Telehealth Center, University Hospital, Universidade Federal de Minas Gerais, Av. Professor Alfredo Balena, Belo Horizonte, 110, Brazil
| | - Bruno Barbosa Miranda de Paiva
- Computer Science Department, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, Belo Horizonte, 6627, Brazil
| | - Lucas Rocha Valle
- Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, Belo Horizonte, 6627, Brazil
| | | | | | - Marcelo Carneiro
- Hospital Santa Cruz. R. Fernando Abott, Santa Cruz do Sul, 174, Brazil
| | | | - Fernando Anschau
- Hospital Nossa Senhora da Conceição, Av. Francisco Trein, Porto Alegre, 326, Brazil
| | | | - Frederico Bartolazzi
- Hospital Santo Antônio, Praça Dr. Márcio Carvalho Lopes Filho, Curvelo, 501, Brazil
| | | | | | | | - Marcos André Gonçalves
- Computer Science Department, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, Belo Horizonte, 6627, Brazil
| | - Milena Soriano Marcolino
- Department of Internal Medicine, Medical School & Telehealth Center, University Hospital, Universidade Federal de Minas Gerais, Av. Professor Alfredo Balena, Belo Horizonte, 110, Brazil
- Institute for Health and Technology Assessment. R. Ramiro Barcelos, Porto Alegre, 2350, Brazil
| |
Collapse
|
12
|
Zhang L, Liu K, Duan X, Zhou S, Jia H, You Y, Han B. CXCL12/CXCR4 axis mediates CD8 + T cell overactivation in the progression of viral myocarditis. J Transl Med 2025; 23:399. [PMID: 40186195 PMCID: PMC11969836 DOI: 10.1186/s12967-025-06394-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/17/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Myocarditis is a common inflammatory heart disease in children and young adults, with fulminant myocarditis (FM) being the most severe form due to its rapid onset and high mortality rate. However, the precise pathological immune subsets and molecular change in myocarditis, particularly FM, remain unknown. METHODS We performed single-cell RNA sequencing of pediatric peripheral blood mononuclear cells during the acute and recovery phases of FM. A viral myocarditis (MC) mouse model was established using CVB3. Deletion and adoptive transfer of CD8+T cells, as well as blockade of CXCR4, were conducted in vivo. CD8+T cells were sorted and cultivated in vitro, then stimulated with CXCL12 and CXCR4 antagonists to investigate the mechanism of CD8+T cell overactivation. RESULTS CD8+T cells show significant activation, amplification, enhanced cytotoxicity, and increased chemotactic ability in FM. Deletion of CD8+T cells alleviates myocardial injury and improves cardiac function in MC mice, while adoptive transfer of CD8+T cells from MC mice aggravates myocardial inflammation and injury. The transcriptomic analysis reveals elevated CXCR4 expression in CD8+T cells in acute FM. In vitro experiments demonstrate that the CXCL12/CXCR4 axis drives the overactivation and cytotoxicity of CD8+T cells. In vivo treatment with a CXCR4 antagonist effectively reduces CD8+T cell accumulation in the heart, alleviates myocardial inflammation, and improves cardiac function in MC mice. CONCLUSIONS These findings provide deeper insights into the immune landscape of pediatric FM, uncovering a novel role of the CXCL12/CXCR4 axis in driving CD8+T cell responses in myocarditis. Furthermore, they highlight the CXCL12/CXCR4 axis as a promising therapeutic target for myocarditis treatment.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Keyu Liu
- Department of Pediatric Cardiology, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiuyun Duan
- Department of Pediatric Cardiology, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shan Zhou
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Hailin Jia
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yingnan You
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Bo Han
- Department of Pediatric Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
- Department of Pediatric Cardiology, Cheeloo College of Medicine, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
- Shandong Provincial Clinical Research Center for Children's Health and Disease office, Shandong Provincial Hospital, Jinan, Shandong, China.
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Medical Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
13
|
Lai C, Lu S, Yang Y, You X, Xu F, Deng X, Lan L, Guo Y, Kuang Z, Luo Y, Yuan L, Meng L, Wu X, Song Z, Jiang N. Myeloid-Driven Immune Suppression Subverts Neutralizing Antibodies and T Cell Immunity in Severe COVID-19. J Med Virol 2025; 97:e70335. [PMID: 40183283 PMCID: PMC11969634 DOI: 10.1002/jmv.70335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025]
Abstract
The objective of this study was to better understand immune failure mechanisms during severe acute respiratory syndrome coronavirus 2, SARS-CoV-2 infection, which are critical for developing targeted vaccines and effective treatments. We collected 34 cases representing different disease severities and performed high-quality single-cell TCR/BCR sequencing to analyze the peripheral immune cell profiles. Additionally, we assessed antibody-neutralizing activity through in vitro experiments. Our integrated multiomics analysis uncovers a profound immune paradox in severe COVID-19: hyperinflammation coexists with immunosuppression, driven by distinct yet interconnected dysregulatory mechanisms. Severe patients develop robust humoral immunity, evidenced by clonally expanded plasma cells producing neutralizing antibodies (e.g., IGHG1-dominated responses) and antigen-specific T cell activation. However, these protective responses are counteracted by myeloid-driven immunosuppression, particularly CD14+ HMGB2+ monocytes exhibiting metabolic reprogramming and HLA-DR downregulation, coupled with progressive T cell exhaustion characterized by IFN-γ/TNF-α hyperactivation and impaired antigen presentation. Importantly, prolonged viral persistence in severe cases arises from a failure to coordinate humoral and cellular immunity-antibody-mediated neutralization cannot compensate for defective cytotoxic T cell function and monocyte-mediated immune suppression. These findings highlight the necessity for therapeutic strategies that simultaneously enhance antibody effector functions (e.g., Fc optimization), restore exhausted T cells, and reverse myeloid suppression. They also highlight the importance of vaccines designed to elicit balanced B cell memory and durable T cell responses, which are critical to preventing severe disease progression. By addressing the dual challenges of hyperinflammation and immunosuppression, such approaches could restore immune coordination and improve outcomes in severe COVID-19.
Collapse
Grants
- This work was supported by the National Key Research and Development Program of China (2021YFC2501800, 2022YFA0806200, 2023YFC0872500, and 2024YFC3044600), the National Natural Science Foundation of China (82072214, 82272198, and 82202373), the Science and Technology of Shanghai Committee (21MC1930400, 22Y11900100, and 23Y31900100), and the Shanghai Municipal Health Commission (2023ZDFC0101).
Collapse
Affiliation(s)
- Cong Lai
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Su Lu
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Yilin Yang
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Xiaoyu You
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Feixiang Xu
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Xinran Deng
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Lulu Lan
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Yuesheng Guo
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Zhongshu Kuang
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Yue Luo
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Li Yuan
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Lu Meng
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Xueling Wu
- Department of Respiratory MedicineShanghai Jiaotong University School of Medicine, Renji HospitalShanghaiChina
| | - Zhenju Song
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| | - Ning Jiang
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| |
Collapse
|
14
|
Yuan L, Stoddard M, Sarkar S, van Egeren D, Mangalaganesh S, Nolan RP, Rogers MS, Hather G, White LF, Chakravarty A. The Impact of Vaccination Frequency on COVID-19 Public Health Outcomes: A Model-Based Analysis. Vaccines (Basel) 2025; 13:368. [PMID: 40333247 PMCID: PMC12031506 DOI: 10.3390/vaccines13040368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/16/2025] [Accepted: 03/27/2025] [Indexed: 05/09/2025] Open
Abstract
Background: While the rapid deployment of SARS-CoV-2 vaccines had a significant impact on the ongoing COVID-19 pandemic, rapid viral immune evasion and waning neutralizing antibody titers have degraded vaccine efficacy. Nevertheless, vaccine manufacturers and public health authorities have a number of options at their disposal to maximize the benefits of vaccination. In particular, the effect of booster schedules on vaccine performance bears further study. Methods: To better understand the effect of booster schedules on vaccine performance, we used an agent-based modeling framework and a population pharmacokinetic model to simulate the impact of boosting frequency on the durability of vaccine protection against infection and severe acute disease. Results: Our work suggests that repeated dosing at frequent intervals (three or more times a year) may offset the degradation of vaccine efficacy, preserving the utility of vaccines in managing the ongoing pandemic. Conclusions: Given the practical significance of potential improvements in vaccine utility, clinical research to better understand the effects of repeated vaccination would be highly impactful. These findings are particularly relevant as public health authorities worldwide have reduced the frequency of boosters to once a year or less.
Collapse
Affiliation(s)
- Lin Yuan
- Fractal Therapeutics, Lexington, MA 02420, USA; (L.Y.); (M.S.)
| | | | - Sharanya Sarkar
- Department of Microbiology and Immunology, Dartmouth College, Hanover, NH 03755, USA;
| | - Debra van Egeren
- Department of Oncology, School of Medicine, Stanford University, Stanford, CA 94305, USA;
| | - Shruthi Mangalaganesh
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3800, Australia;
| | | | - Michael S. Rogers
- Department of Surgery, Harvard Medical School, Boston, MA 02114, USA;
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Greg Hather
- Sage Therapeutics, Cambridge, MA 02142, USA;
| | - Laura F. White
- School of Public Health, Boston University, Boston, MA 02118, USA;
| | | |
Collapse
|
15
|
Zhuang Q, Zhu J, Peng B, Zhu Y, Cheng K, Ming Y. Correlation between peripheral lymphocyte subsets monitoring and COVID-19 pneumonia in kidney transplant recipients. BMC Infect Dis 2025; 25:426. [PMID: 40148763 PMCID: PMC11948920 DOI: 10.1186/s12879-025-10581-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/30/2025] [Indexed: 03/29/2025] Open
Abstract
OBJECTIVES In kidney transplant recipients (KTRs), immune monitoring of peripheral lymphocyte subsets (PLS) reflects the real immune status and aids in the diagnosis of the occurrence and development of infectious diseases, including COVID-19. Exploring the PLS of COVID-19 pneumonia in KTRs is important. METHODS In this study, a total of 103 KTRs were divided into mild pneumonia (MP) and severe pneumonia (SP) groups, as well as a stable group. The clinical information and PLS data were assessed via t or Mann-Whitney test and receiver operating curve analysis. Logistic regression was employed to identify the risk factors, and Spearman's correlation analysis was used to identify correlations. RESULTS Lymphopenia is a common manifestation of COVID-19 in KTRs, and it is positively related to the severity of COVID-19 pneumonia. The CD3 + T-cell count had the highest AUC between the MP and the SP. Multiple PLS measures were found to be independent risk factors for COVID-19 pneumonia progression in KTRs. CONCLUSIONS This study revealed a robust correlation between PLS and severe COVID-19 pneumonia progression in KTRs. PLS monitoring could facilitate real-time diagnosis and therapy for infection, as well as timely and precisive adjustment of immunosuppression instructions, for KTRs with COVID-19.
Collapse
Affiliation(s)
- Quan Zhuang
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Key Laboratory of Translational Research in Transplantation Medicine of National Health Commission, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Clinical Research Center for Infectious Diseases in Hunan Province, Changsha, 410013, China
| | - Jiang Zhu
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Key Laboratory of Translational Research in Transplantation Medicine of National Health Commission, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Clinical Research Center for Infectious Diseases in Hunan Province, Changsha, 410013, China
| | - Bo Peng
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Key Laboratory of Translational Research in Transplantation Medicine of National Health Commission, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Clinical Research Center for Infectious Diseases in Hunan Province, Changsha, 410013, China
| | - Yi Zhu
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Key Laboratory of Translational Research in Transplantation Medicine of National Health Commission, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Clinical Research Center for Infectious Diseases in Hunan Province, Changsha, 410013, China
| | - Ke Cheng
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Key Laboratory of Translational Research in Transplantation Medicine of National Health Commission, Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Clinical Research Center for Infectious Diseases in Hunan Province, Changsha, 410013, China
| | - Yingzi Ming
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, 410013, China.
- Key Laboratory of Translational Research in Transplantation Medicine of National Health Commission, Third Xiangya Hospital, Central South University, Changsha, 410013, China.
- Clinical Research Center for Infectious Diseases in Hunan Province, Changsha, 410013, China.
| |
Collapse
|
16
|
Li L, Feng T, Shen Q, Shi X, Wei Z, Chen W, Yang F, Zhu Y, Zhang C, Zhang S, Zhang Q, Fu S, Wang N, Tian WX, Liu J, Si L. Natural Infection of Omicron BA.5.2 in Patients Provides Broad Immune Responses Against SARS-CoV-2. Microorganisms 2025; 13:746. [PMID: 40284583 PMCID: PMC12029644 DOI: 10.3390/microorganisms13040746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 03/22/2025] [Accepted: 03/23/2025] [Indexed: 04/29/2025] Open
Abstract
The implementation of COVID-19 policy and the rapid development of SARS-CoV-2 vaccines in the early pandemic significantly contained numerous outbreaks and reduced the severity and mortality of COVID-19. However, the population immunity induced by existing vaccines was insufficient to prevent SARS-CoV-2 outbreaks. The host immunity induced by the wide spread of Omicron variants and its influence on emerging SARS-CoV-2 variants are attracting broad attention. In this study, a clinical data analysis of the patients indicated that pre-vaccination reduced inflammatory responses and mitigated the severity of COVID-19 cases caused by natural infection with Omicron BA.5.2. The analysis of adaptive immune responses indicated that natural infection with BA.5.2 induced robust and broad immune responses, including both humoral and T cell-mediated immune responses (IFN-γ) against highly conserved viral antigens, and provided cross-reactive neutralization against various viral variants. Collectively, we report that the natural infection with Omicron BA.5.2 induced broad cross-reactive immunity against SARS-CoV-2 variants, which suggests that the development of a live attenuated SARS-CoV-2 vaccine with desired safety, high efficacy, broad spectrum, and long-term immune persistence is feasible. Therefore, we suggest that herd immunity, achieved through vaccination with attenuated vaccines, combined with booster doses of existing vaccines and antiviral therapy for people with high viral loads, may contribute to the eradication of this virus.
Collapse
Affiliation(s)
- Le Li
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Tang Feng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Quan Shen
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoshan Shi
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhigong Wei
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wanze Chen
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fan Yang
- University of Chinese Academy of Sciences, Beijing 100049, China
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yueting Zhu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chengxin Zhang
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Shuang Zhang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qisi Zhang
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Shengwei Fu
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ning Wang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wen-xia Tian
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong 030801, China
| | - Jiyan Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Longlong Si
- State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
17
|
Perestiuk V, Kosovska T, Volianska L, Boyarchuk O. Association of zinc deficiency and clinical symptoms, inflammatory markers, severity of COVID-19 in hospitalized children. Front Nutr 2025; 12:1566505. [PMID: 40201587 PMCID: PMC11975579 DOI: 10.3389/fnut.2025.1566505] [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: 01/24/2025] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
Introduction Zinc plays an important role in the functioning of the immune system. Zinc deficiency leads to increased susceptibility to inflammatory and infectious diseases. There are few studies investigating the role of zinc in the development and progression of COVID-19 in children, and their findings remain inconsistent. This study aimed to determine the zinc levels in children with COVID-19 and assess their association with symptoms, inflammation markers, and disease progression. Methods A prospective cohort study included hospitalized patients under 18 years who had a confirmed diagnosis of SARS-CoV-2 infection. Serum zinc concentrations were measured using a colorimetric method. Based on zinc levels, the children were divided into two groups: the first group had concentrations below 10.7 μmol/L, indicating zinc deficiency, while the second group had levels above 10.7 μmol/L, which was considered within the optimal range. Results In total, 140 hospitalized patients with COVID-19 were examined. Zinc deficiency was identified in 40 children (28.6%), while optimal levels were found in 100 children (71.4%). Zinc status did not depend on the age of the children. Among the symptoms of acute SARS-CoV-2 infection, children with zinc deficiency showed a trend toward more frequent fever occurrences (p = 0.0654). No significant impact of zinc status was observed on the severity of COVID-19 or the duration of hospitalization. Children with zinc deficiency had higher median values of the neutrophil-to-lymphocyte ratio (NLR) (1.84 vs. 1.09, p = 0.0010), C-reactive protein (CRP) levels (9.65 vs. 3.96 mg/L, p = 0.0053), and fibrinogen levels (2.88 vs. 2.07 g/L, p = 0.0057) compared to those with adequate zinc levels. Additionally, the percentage of patients with a NLR greater than 4, elevated CRP, and fibrinogen levels was higher in the zinc-deficient group (p = 0.0017, p = 0.0107, p = 0.0338, respectively). Conclusion Zinc deficiency was observed in 28.6% of children with COVID-19 and was not dependent on age. Children with hypozincemia had higher levels of inflammation markers, including the neutrophil-to-lymphocyte ratio and CRP.
Collapse
Affiliation(s)
- Vita Perestiuk
- Department of Children’s Diseases and Pediatric Surgery, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | | | | | | |
Collapse
|
18
|
Yildiz Gulhan P, Eroz R, Ozturk CE, Yekenkurul D, Altinsoy HB, Gulec Balbay E, Ercelik M, Davran F, Yildiz S. Determination of both the expression and serum levels of epidermal growth factor and transforming growth factor β1 genes in COVID-19. Sci Rep 2025; 15:9771. [PMID: 40118922 PMCID: PMC11928509 DOI: 10.1038/s41598-025-92304-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/26/2025] [Indexed: 03/24/2025] Open
Abstract
We aimed to evaluate the effects of both the expression and serum levels of Epidermal growth factor (EGF) and Transforming growth factor-β1 (TGF-β1) genes in patients with different degrees of cellular damage as mild, moderate, severe, and critical illness that can lead to fibrosis caused by SARS-CoV-2. Totally 45 individuals (male: 21(46.67%); female: 24(53.33%)) with COVID-19 infection were included in this study. Four groups were constituted as mild (n = 16)], moderate (n = 10), severe (n = 10), and critical (n = 9) according to the severity of the disease. Blood samples were drawn from the patients, and all of the hemograms, EGF and TGFβ1 gene expression, and serum levels were evaluated. The mean age of individuals was 57.311 ± 18.383 (min: 28, max: 94). Significant differences were found among the groups for PLT (χ2 = 9.955; p = 0.019), CRP (χ2 = 7.693; p = 0.053), Ferritin (χ2 = 22.196; p < 0.001), D-dimer (χ2 = 21.982; p = 0.000), LDH (χ2 = 21.807; p < 0.001) and all these parameters (exclude PLT in severe groups) was increased depending on the severity of the disease. Additionally, significant differences were detected for EGF (χ2 = 29.528; p < 0.001), TGFB1 (χ2 = 28.981; p < 0.001) expression (that increased depending on the disease severity), and EGF (χ2 = 7.84; p = 0.049), TGFB1 (χ2 = 17.451; p = 0.001) serum concentration levels (that decreased depending on the disease severity). This study found statistically significant differences for both EGF 2-ΔΔCt. TGFβ1 2-ΔΔCt and EGF, TGFβ1 serum concentration values among all patient groups. As disease severity increased, EGF 2-ΔΔCt. TGFβ1 2-ΔΔCt levels increased, while EGF and TGFβ1 serum concentration levels decreased. Perhaps this study will be useful in managing COVID-19 infection severity and pulmonary fibrosis cases secondary to COVID-19.
Collapse
Affiliation(s)
- Pinar Yildiz Gulhan
- Department of Chest Diseases, Faculty of Medicine, Duzce University, Konuralp Campus, 81010, Duzce, Turkey.
| | - Recep Eroz
- Department of Medical Genetics, Aksaray University Medical Faculty, Aksaray, Turkey
| | | | - Dilek Yekenkurul
- Department of Infection Diseases, Duzce University Medical Faculty, Duzce, Turkey
| | | | - Ege Gulec Balbay
- Department of Chest Diseases, Faculty of Medicine, Duzce University, Konuralp Campus, 81010, Duzce, Turkey
| | - Merve Ercelik
- Department of Chest Diseases, Faculty of Medicine, Duzce University, Konuralp Campus, 81010, Duzce, Turkey
| | - Fatih Davran
- Department of Biochemistry, Faculty of Medicine, Duzce University, Duzce, Turkey
| | - Seyma Yildiz
- Deparment of Hematology, Gazi University, Ankara, Turkey
| |
Collapse
|
19
|
Vita E, Monaca F, Mastrantoni L, Piro G, Moretti G, Sparagna I, Stefani A, Vitale A, Trovato G, Di Salvatore M, Sanguinetti M, Urbani A, Richeldi L, Carbone C, Bria E, Tortora G. COVALENCE STUDY: Immunogenicity and Reactogenicity of a COVID-19 mRNA Vaccine in an Open-Label Cohort of Long-Survivor Patients with Metastatic Lung Cancer. Vaccines (Basel) 2025; 13:273. [PMID: 40266144 PMCID: PMC11946322 DOI: 10.3390/vaccines13030273] [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: 12/30/2024] [Revised: 02/24/2025] [Accepted: 03/01/2025] [Indexed: 04/24/2025] Open
Abstract
Background: As COVID-19 has become an epidemic, we conducted an open-label study aimed to identify immunogenicity and reactogenicity of boosters of the BNT162b2 vaccine in a real-world cohort of long-survivor metastatic lung cancer patients (LS-mLC pts). Methods and Analysis: According to the timing of the booster dose (BD) and SARS-CoV-2 infection (Cov-I) during anticancer treatment (ACT), between October 2021 and February 2022, we prospectively enrolled 166 cancer patients into five parallel cohorts. The primary endpoints were seroprevalence of IgG Anti-spike-RBD (anti-S IgG) at two pre-defined timepoints (T1: +30-90 days after BD; T2: +6 months +/- 4 weeks after BD). As an exploratory endpoint, we compared the median pre-vaccination value of four cytokines (IL-6, IL-2R, TNF-α, IL-10) with post-BD values in immunotherapy-treated pts (IO-pts). Results: The anti-S IgG seropositivity rate was 100% at T1 and 98.8% at T2. After 6 months, hybrid immunisation was associated with a higher median anti-S IgG titre compared to vaccine-alone-induced seroconversion (p < 0.0001). In uninfected pts, the median anti-S IgG titre was significantly lower in IO-pts compared to non-IO-pts (p = 0.02); no difference was found when comparing myelosuppressive or not ACT. Among the 68 IO-pts, 5 pts (7.3%) showed a significant increase (≥1.5 fold) of at least two cytokines in post-BD samples, without reporting ir-AEs. Conclusions: Boosters of the COVID-19 mRNA vaccine were effective and safe. In IO-pts without recent Cov-I, additional BDs should be considered to prolong serological immunity.
Collapse
Affiliation(s)
- Emanuele Vita
- UOSD Oncologia Toraco-Polmonare, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.D.S.); (E.B.)
| | - Federico Monaca
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.M.); (G.P.); (I.S.); (A.S.); (A.V.); (G.T.); (C.C.); (G.T.)
| | - Luca Mastrantoni
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.M.); (G.P.); (I.S.); (A.S.); (A.V.); (G.T.); (C.C.); (G.T.)
| | - Geny Piro
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.M.); (G.P.); (I.S.); (A.S.); (A.V.); (G.T.); (C.C.); (G.T.)
| | - Giacomo Moretti
- UOC Chimica, Biochimica e Biologia Molecolare Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.M.); (A.U.)
| | - Ileana Sparagna
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.M.); (G.P.); (I.S.); (A.S.); (A.V.); (G.T.); (C.C.); (G.T.)
| | - Alessio Stefani
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.M.); (G.P.); (I.S.); (A.S.); (A.V.); (G.T.); (C.C.); (G.T.)
| | - Antonio Vitale
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.M.); (G.P.); (I.S.); (A.S.); (A.V.); (G.T.); (C.C.); (G.T.)
| | - Giovanni Trovato
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.M.); (G.P.); (I.S.); (A.S.); (A.V.); (G.T.); (C.C.); (G.T.)
| | - Mariantonietta Di Salvatore
- UOSD Oncologia Toraco-Polmonare, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.D.S.); (E.B.)
| | - Maurizio Sanguinetti
- UOC Microbiologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Andrea Urbani
- UOC Chimica, Biochimica e Biologia Molecolare Clinica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.M.); (A.U.)
| | - Luca Richeldi
- UOC Pneumologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Carmine Carbone
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.M.); (G.P.); (I.S.); (A.S.); (A.V.); (G.T.); (C.C.); (G.T.)
| | - Emilio Bria
- UOSD Oncologia Toraco-Polmonare, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (M.D.S.); (E.B.)
| | - Giampaolo Tortora
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.M.); (G.P.); (I.S.); (A.S.); (A.V.); (G.T.); (C.C.); (G.T.)
| |
Collapse
|
20
|
Maliha ST, Fatemi R, Akter M, Zheng Q, Araf Y, Tabassum T, Munif MR, Saha S, Xue M, Wang H, Zheng C, Hossain MG. Exploring the dynamics of SARS-CoV-2 and HIV Co-infection: Mutation risks, therapeutic efficacy, and future variant prevention. Diagn Microbiol Infect Dis 2025; 111:116707. [PMID: 39854809 DOI: 10.1016/j.diagmicrobio.2025.116707] [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] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
High mutation rates in SARS-CoV-2, particularly among immunocompromised patients living with HIV, continue to complicate the current COVID-19 pandemic. The threshold for severe COVID-19 and a greater risk of mortality have increased in many immunocompromised individuals due to a weakened immune system. Low CD4+ T-cell counts in people living with both HIV and COVID-19 lead to prolonged disease duration and, therefore, an increased likelihood of viral infection with SARS-CoV-2 mutations in such individuals. These mutations could decrease the efficiency of ongoing vaccines and cause new outbreaks. Recently, the rise of new mutations in this patient population has created increasing concern; however, few data are currently available on the direct association of HIV infection with SARS-CoV-2 mutations. This review highlights the implications of SARS-CoV-2 and HIV co-infection, highlighting the need for extra caution and monitoring of the immune-compromised population during a pandemic. Access to HIV care and COVID-19 treatments, careful surveillance, and adapted health strategies are key to reducing risks and protecting these populations. Further research is required to elucidate the dynamics of mutations and develop intervention methods to manage COVID-19 among immunocompromised patients.
Collapse
Affiliation(s)
- Sumaiya Tasnim Maliha
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, BRAC University, Dhaka, Bangladesh
| | - Rabeya Fatemi
- Department of Genetic Engineering and Biotechnology, East-West University, Dhaka 1212, Bangladesh
| | - Marjana Akter
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Qingcong Zheng
- Department of Spinal Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yusha Araf
- Department of Biotechnology, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Tahani Tabassum
- Biotechnology Program, Department of Mathematics and Natural Sciences, School of Data and Sciences, BRAC University, Dhaka, Bangladesh
| | - Mohammad Raguib Munif
- Department of Surgery and Obstetrics, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Sukumar Saha
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China.
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| | - Md Golzar Hossain
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh 2202, Bangladesh.
| |
Collapse
|
21
|
Ji X, Guo Y, Tang L, Gao C. Identifying and Validating Prognostic Hyper-Inflammatory and Hypo-Inflammatory COVID-19 Clinical Phenotypes Using Machine Learning Methods. J Inflamm Res 2025; 18:3009-3024. [PMID: 40034687 PMCID: PMC11874972 DOI: 10.2147/jir.s504028] [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: 11/22/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025] Open
Abstract
Background COVID-19 exhibits complex pathophysiological manifestations, characterized by significant clinical and biological heterogeneity. Identifying phenotypes may enhance our understanding of the disease's diverse trajectories, benefiting clinical practice and trials. Methods This study included adult patients with COVID-19 from Xinhua Hospital, affiliated with Shanghai Jiao Tong University School of Medicine, between December 15, 2022, and February 15, 2023. The k-prototypes clustering method was employed using 50 clinical variables to identify phenotypes. Machine learning algorithms were then applied to select key classifier variables for phenotype recognition. Results A total of 1376 patients met the inclusion criteria. K-prototypes clustering revealed two distinct subphenotypes: Hypo-inflammatory subphenotype (824 [59.9%]) and Hyper-inflammatory subphenotype (552 [40.1%]). Patients in Hypo-inflammatory subphenotype were younger, predominantly female, with low mortality and shorter hospital stays. In contrast, Hyper-inflammatory subphenotype patients were older, predominantly male, exhibiting a hyperinflammatory state with higher mortality and rates of organ dysfunction. The AdaBoost model performed best for subphenotype prediction (Accuracy: 0.975, Precision: 0.968, Recall: 0.976, F1: 0.972, AUROC: 0.975). "CRP", "IL-2R", "D-dimer", "ST2", "BUN", "NT-proBNP", "neutrophil percentage", and "lymphocyte count" were identified as the top-ranked variables in the AdaBoost model. Conclusion This analysis identified two phenotypes based on COVID-19 symptoms and comorbidities. These phenotypes can be accurately recognized using machine learning models, with the AdaBoost model being optimal for predicting in-hospital mortality. The variables "CRP", "IL-2R", "D-dimer", "ST2", "BUN", "NT-proBNP", "neutrophil percentage", and "lymphocyte count" play a significant role in the prediction of subphenotypes. Use the identified subphenotypes for risk stratification in clinical practice. Hyper-inflammatory subphenotypes can be closely monitored, and preventive measures such as early admission to the intensive care unit or prophylactic anticoagulation can be taken.
Collapse
Affiliation(s)
- Xiaojing Ji
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, People’s Republic of China
| | - Yiran Guo
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, People’s Republic of China
| | - Lujia Tang
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, People’s Republic of China
| | - Chengjin Gao
- Department of Emergency, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, People’s Republic of China
| |
Collapse
|
22
|
Hernández-Blanco C, Al-Akioui-Sanz K, Herrera L, Aguirre-Portolés C, Lozano-Ojalvo D, Pérez-Rodriguez L, Cano-Ochando J, Guerra-García P, Martín-Quirós A, Vicario JL, Santos S, Pérez-Vaquero MÁ, Vesga MÁ, Borobia AM, Carcas AJ, Balas A, Moreno MÁ, Pérez de Diego R, Gasior M, Soria B, Eguizabal C, Pérez-Martínez A. The phase I RELEASE clinical trial to evaluate the safety of NK cells in COVID-19. iScience 2025; 28:111698. [PMID: 39877904 PMCID: PMC11772958 DOI: 10.1016/j.isci.2024.111698] [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: 06/10/2024] [Revised: 10/18/2024] [Accepted: 12/24/2024] [Indexed: 01/31/2025] Open
Abstract
The severity of COVID 19 symptoms has a direct correlation with lymphopenia, affecting natural killer (NK) cells. SARS-CoV-2 specific "memory" NK cells obtained from convalescent donors can be used as cell immunotherapy. In 2022 a phase I, dose-escalation, single center clinical trial was conducted to evaluate the safety and feasibility of the infusion of CD3-/CD56+ NK cells against moderate/severe cases of COVID-19 (NCT04578210). Six participants with pneumonia and/or lymphopenia were infused. Four patients received a single-dose infusion of NK cells of 1×106/kg, and the following two patients a dose of 2×106/kg of NK cells. All participants' clinical status and inflammation markers were monitored. No serious adverse events were reported after infusion. Exploratory outcomes included the donor chimerism, NK-cell immunophenotype evolution, and immune lymphocyte reconstitution. This study provides preliminary evidence supporting the idea that treatment of COVID-19 patients with moderate/severe symptoms using NK from COVID-19 convalescent donors is feasible and safe.
Collapse
Affiliation(s)
- Clara Hernández-Blanco
- Internal Medicine Department, University Hospital La Paz, Madrid, Spain
- Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Karima Al-Akioui-Sanz
- Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- CNIO Pediatric OncoHematology Clinical Research Unit, Madrid, Spain
| | - Lara Herrera
- Cell Therapy, Stem Cells and Tissues Group, Biobizkaia Health Research Institute, Barakaldo, Spain
- Advanced Therapies Unit, Basque Center of Blood Transfusion and Human Tissues, Osakidetza, Galdakao, Spain
- Red Española de Terapias Avanzadas (TERAV), Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS RD21/0017/0024, RICORS RD24/0014/0025, RICORS RD21/0017/0025, RICORS RD24/0014/0024), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Red de Inmunoterapia del Cáncer “REINCA” (RED2022-134831-T), Madrid, Spain
| | - Cristina Aguirre-Portolés
- Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- CNIO Pediatric OncoHematology Clinical Research Unit, Madrid, Spain
| | - Daniel Lozano-Ojalvo
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Food Science Research Institute (CIAL, CSIC-UAM), Madrid, Spain
| | - Leticia Pérez-Rodriguez
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jordi Cano-Ochando
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Microbiology National Centre, Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Guerra-García
- Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- Clinical Trials Unit (UCICEC), University Hospital La Paz, Madrid, Spain
| | | | - José Luis Vicario
- Histocompatibility, Centro de Transfusión de la Comunidad de Madrid, Madrid, Spain
| | - Silvia Santos
- Cell Therapy, Stem Cells and Tissues Group, Biobizkaia Health Research Institute, Barakaldo, Spain
- Advanced Therapies Unit, Basque Center of Blood Transfusion and Human Tissues, Osakidetza, Galdakao, Spain
- Red Española de Terapias Avanzadas (TERAV), Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS RD21/0017/0024, RICORS RD24/0014/0025, RICORS RD21/0017/0025, RICORS RD24/0014/0024), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Red de Inmunoterapia del Cáncer “REINCA” (RED2022-134831-T), Madrid, Spain
| | - Miguel Ángel Pérez-Vaquero
- Cell Therapy, Stem Cells and Tissues Group, Biobizkaia Health Research Institute, Barakaldo, Spain
- Advanced Therapies Unit, Basque Center of Blood Transfusion and Human Tissues, Osakidetza, Galdakao, Spain
- Red Española de Terapias Avanzadas (TERAV), Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS RD21/0017/0024, RICORS RD24/0014/0025, RICORS RD21/0017/0025, RICORS RD24/0014/0024), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Red de Inmunoterapia del Cáncer “REINCA” (RED2022-134831-T), Madrid, Spain
| | - Miguel Ángel Vesga
- Cell Therapy, Stem Cells and Tissues Group, Biobizkaia Health Research Institute, Barakaldo, Spain
- Advanced Therapies Unit, Basque Center of Blood Transfusion and Human Tissues, Osakidetza, Galdakao, Spain
- Red Española de Terapias Avanzadas (TERAV), Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS RD21/0017/0024, RICORS RD24/0014/0025, RICORS RD21/0017/0025, RICORS RD24/0014/0024), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Red de Inmunoterapia del Cáncer “REINCA” (RED2022-134831-T), Madrid, Spain
| | - Alberto M. Borobia
- Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- Clinical Trials Unit (UCICEC), University Hospital La Paz, Madrid, Spain
| | - Antonio J. Carcas
- Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- Clinical Trials Unit (UCICEC), University Hospital La Paz, Madrid, Spain
- Faculty of Medicine Universidad Autónoma de Madrid, Madrid, Spain
| | - Antonio Balas
- Histocompatibility, Centro de Transfusión de la Comunidad de Madrid, Madrid, Spain
| | - Miguel Ángel Moreno
- Histocompatibility, Centro de Transfusión de la Comunidad de Madrid, Madrid, Spain
| | | | - Mercedes Gasior
- Hematology Department, Hospital Universitario La Paz, Madrid, Spain
| | - Bernat Soria
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Institute of Bioengineering, Miguel Hernández University, Elche, Alicante, Spain
| | - Cristina Eguizabal
- Cell Therapy, Stem Cells and Tissues Group, Biobizkaia Health Research Institute, Barakaldo, Spain
- Advanced Therapies Unit, Basque Center of Blood Transfusion and Human Tissues, Osakidetza, Galdakao, Spain
- Red Española de Terapias Avanzadas (TERAV), Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS RD21/0017/0024, RICORS RD24/0014/0025, RICORS RD21/0017/0025, RICORS RD24/0014/0024), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Red de Inmunoterapia del Cáncer “REINCA” (RED2022-134831-T), Madrid, Spain
| | - Antonio Pérez-Martínez
- Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- CNIO Pediatric OncoHematology Clinical Research Unit, Madrid, Spain
- Red Española de Terapias Avanzadas (TERAV), Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS RD21/0017/0024, RICORS RD24/0014/0025, RICORS RD21/0017/0025, RICORS RD24/0014/0024), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Pediatric Hemato-Oncology Department, University Hospital La Paz, Madrid, Spain
- Histocompatibility, Centro de Transfusión de la Comunidad de Madrid, Madrid, Spain
- Faculty of Medicine Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
23
|
Ramalho-Pinto CH, Ventura LHA, Camatta GC, Silveira-Nunes G, Gomes MS, Sato HI, Costa MS, Guimarães HC, Barbuto RC, Martins-Filho OA, Amaral LR, Bertarini PLL, Teixeira SMR, Tupinambás U, Teixeira-Carvalho A, Faria AMC. Machine learning algorithm approach to complete blood count can be used as early predictor of COVID-19 outcome. J Leukoc Biol 2025; 117:qiae223. [PMID: 39432758 DOI: 10.1093/jleuko/qiae223] [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: 07/22/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024] Open
Abstract
Although the SARS-CoV-2 infection has established risk groups, identifying biomarkers for disease outcomes is still crucial to stratify patient risk and enhance clinical management. Optimal efficacy of COVID-19 antiviral medications relies on early administration within the initial 5 d of symptoms, assisting high-risk patients in avoiding hospitalization and improving survival chances. The complete blood count (CBC) can be an efficient and affordable option to find biomarkers that predict the COVID-19 prognosis due to infection-induced alterations in various blood parameters. This study aimed to associate hematological parameters with different COVID-19 clinical forms and utilizes them as disease outcome predictors. We performed a CBC in blood samples from 297 individuals with COVID-19 from Belo Horizonte, Brazil. Statistical analysis, as well as ROC Curves and machine learning Decision Tree algorithms were used to identify correlations, and their accuracy, between blood parameters and disease severity. In the initial 4 d of infection, traditional hematological COVID-19 alterations, such as lymphopenia, were not yet apparent. However, the monocyte percentage and granulocyte-to-lymphocyte ratio (GLR) proved to be reliable predictors for hospitalization, even in cases where patients exhibited mild symptoms that later progressed to hospitalization. Thus, our findings demonstrate that COVID-19 patients with monocyte percentages lower than 7.7% and a GLR higher than 8.75 are assigned to the hospitalized group with a precision of 86%. This suggests that these variables can serve as important biomarkers in predicting disease outcomes and could be used to differentiate patients at hospital admission for managing therapeutic interventions, including early antiviral administration. Moreover, they are simple parameters that can be useful in minimally equipped health care units.
Collapse
Affiliation(s)
- Cecília Horta Ramalho-Pinto
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Brazil
| | - Lucas Haniel Araújo Ventura
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Brazil
| | - Giovanna Caliman Camatta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Brazil
| | - Gabriela Silveira-Nunes
- Departamento de Medicina, Universidade Federal de Juiz de Fora, Av. Doutor Raimundo Monteiro Resende, 330, 35010-177, Governador Valadares, Brazil
| | - Matheus Souza Gomes
- Instituto de Biotecnologia, Universidade Federal de Uberlândia, R. Padre Pavoni, 290, 38701-002, Patos de Minas, Brazil
| | - Hugo Itaru Sato
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Brazil
| | - Murilo Soares Costa
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, Av. Alfredo Balena, 190, 30130-100, Belo Horizonte, Brazil
| | | | - Rafael Calvão Barbuto
- Hospital Universitário Risoleta Tolentino Neves, R. das Gabirobas, 1, 31744-012, Belo Horizonte, Brazil
| | - Olindo Assis Martins-Filho
- Instituto de Pesquisa René Rachou, Fundação Oswaldo Cruz - MG, Av. Augusto de Lima, 1715, 30190-002, Belo Horizonte, Brazil
| | - Laurence Rodrigues Amaral
- Faculdade de Computação, Universidade Federal de Uberlândia, R. Padre Pavoni, 290, 38701-002, Patos de Minas, Brazil
| | - Pedro Luiz Lima Bertarini
- Faculdade de Engenharia Elétrica, Universidade Federal de Uberlândia, R. Padre Pavoni, 290, 38701-002, Patos de Minas, Brazil
| | - Santuza Maria Ribeiro Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Brazil
| | - Unaí Tupinambás
- Departamento de Medicina, Universidade Federal de Juiz de Fora, Av. Doutor Raimundo Monteiro Resende, 330, 35010-177, Governador Valadares, Brazil
| | - Andrea Teixeira-Carvalho
- Instituto de Pesquisa René Rachou, Fundação Oswaldo Cruz - MG, Av. Augusto de Lima, 1715, 30190-002, Belo Horizonte, Brazil
| | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, 31270-901, Belo Horizonte, Brazil
| |
Collapse
|
24
|
Naiditch H, Betts MR, Larman HB, Levi M, Rosenberg AZ. Immunologic and inflammatory consequences of SARS-CoV-2 infection and its implications in renal disease. Front Immunol 2025; 15:1376654. [PMID: 40012912 PMCID: PMC11861071 DOI: 10.3389/fimmu.2024.1376654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 12/23/2024] [Indexed: 02/28/2025] Open
Abstract
The emergence of the COVID-19 pandemic made it critical to understand the immune and inflammatory responses to the SARS-CoV-2 virus. It became increasingly recognized that the immune response was a key mediator of illness severity and that its mechanisms needed to be better understood. Early infection of both tissue and immune cells, such as macrophages, leading to pyroptosis-mediated inflammasome production in an organ system critical for systemic oxygenation likely plays a central role in the morbidity wrought by SARS-CoV-2. Delayed transcription of Type I and Type III interferons by SARS-CoV-2 may lead to early disinhibition of viral replication. Cytokines such as interleukin-1 (IL-1), IL-6, IL-12, and tumor necrosis factor α (TNFα), some of which may be produced through mechanisms involving nuclear factor kappa B (NF-κB), likely contribute to the hyperinflammatory state in patients with severe COVID-19. Lymphopenia, more apparent among natural killer (NK) cells, CD8+ T-cells, and B-cells, can contribute to disease severity and may reflect direct cytopathic effects of SARS-CoV-2 or end-organ sequestration. Direct infection and immune activation of endothelial cells by SARS-CoV-2 may be a critical mechanism through which end-organ systems are impacted. In this context, endovascular neutrophil extracellular trap (NET) formation and microthrombi development can be seen in the lungs and other critical organs throughout the body, such as the heart, gut, and brain. The kidney may be among the most impacted extrapulmonary organ by SARS-CoV-2 infection owing to a high concentration of ACE2 and exposure to systemic SARS-CoV-2. In the kidney, acute tubular injury, early myofibroblast activation, and collapsing glomerulopathy in select populations likely account for COVID-19-related AKI and CKD development. The development of COVID-19-associated nephropathy (COVAN), in particular, may be mediated through IL-6 and signal transducer and activator of transcription 3 (STAT3) signaling, suggesting a direct connection between the COVID-19-related immune response and the development of chronic disease. Chronic manifestations of COVID-19 also include systemic conditions like Multisystem Inflammatory Syndrome in Children (MIS-C) and Adults (MIS-A) and post-acute sequelae of COVID-19 (PASC), which may reflect a spectrum of clinical presentations of persistent immune dysregulation. The lessons learned and those undergoing continued study likely have broad implications for understanding viral infections' immunologic and inflammatory consequences beyond coronaviruses.
Collapse
Affiliation(s)
- Hiam Naiditch
- Department of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Michael R. Betts
- Department of Microbiology and Institute of Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - H. Benjamin Larman
- Institute for Cell Engineering, Division of Immunology, Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, United States
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
25
|
Harti GF, Maulida SN, Susandi E, Fadjari TH, Sumardi U, Alisjahbana B, Wijaya I. Comparison of Platelet Indices, Lymphocyte, and Systemic Inflammation Indices on Days 1 and 8 in Surviving and Non-Surviving COVID-19 Patients at Hasan Sadikin General Hospital, Bandung, Indonesia. J Blood Med 2025; 16:61-74. [PMID: 39926111 PMCID: PMC11806915 DOI: 10.2147/jbm.s499023] [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: 11/06/2024] [Accepted: 01/21/2025] [Indexed: 02/11/2025] Open
Abstract
Purpose This study aimed to compare platelet count, platelet indices, lymphocyte, and systemic inflammation indices between surviving and non-surviving COVID-19 patients, measured at admission and on the eighth day of hospitalization. Patients and Methods A retrospective cohort study was conducted on COVID-19 patients hospitalized at Hasan Sadikin General Hospital, Bandung, from March to December 2020. Patient characteristics and laboratory data were sourced from medical records and the Clinical Pathology Laboratory. Bivariate analysis was performed to determine the comparison of platelet indexes between Surviving and Non-Surviving COVID-19 patients depending on data distribution. Significantly correlated variables in Bivariate analysis were included in the ROC analysis, with the AUC used to identify optimal threshold values for laboratory parameters. Results Data from 132 patients were analyzed, with 106 (80.3%) surviving and 32 (19.7%) not surviving. Non-surviving patients had lower platelet count, PLTCT, and lymphocyte levels but higher MPV and PDW compared to survivors. Receiver operating characteristic (ROC) analysis revealed that on day 1, lymphocytes had a higher area under the curve (AUC) than MPV. On day 8, lymphocytes had the highest AUC, followed by platelet count, MPV, PLTCT, and PDW. Conclusion Platelet indices, lymphocyte counts, and systemic inflammation index have the potential to distinguish the severity of COVID-19.
Collapse
Affiliation(s)
- Gusti Fungani Harti
- Division of Hemato and Oncology Medic, Department of Internal Medicine, Hasan Sadikin General Hospital, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Syifa Nur Maulida
- Research Center for Care and Control of Infectious Disease, Universitas Padjadjaran, Bandung, Indonesia
| | - Evan Susandi
- Division of Tropical and Infectious Disease, Department of Internal Medicine, Hasan Sadikin General Hospital, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Trinugroho Heri Fadjari
- Division of Hemato and Oncology Medic, Department of Internal Medicine, Hasan Sadikin General Hospital, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Uun Sumardi
- Division of Tropical and Infectious Disease, Department of Internal Medicine, Hasan Sadikin General Hospital, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Bachti Alisjahbana
- Research Center for Care and Control of Infectious Disease, Universitas Padjadjaran, Bandung, Indonesia
- Division of Tropical and Infectious Disease, Department of Internal Medicine, Hasan Sadikin General Hospital, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| | - Indra Wijaya
- Division of Hemato and Oncology Medic, Department of Internal Medicine, Hasan Sadikin General Hospital, Faculty of Medicine Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
26
|
Shankar-Hari M, Francois B, Remy KE, Gutierrez C, Pastores S, Daix T, Jeannet R, Blood J, Walton AH, Salomao R, Auzinger G, Striker D, Martin RS, Anand NJ, Bosanquet J, Blood T, Brakenridge S, Moldawer LL, Vachharajani V, Yee C, Dal-Pizzol F, Morre M, Berbille F, van den Brink M, Hotchkiss R. A randomized, double-blind, placebo-controlled trial of IL-7 in critically ill patients with COVID-19. JCI Insight 2025; 10:e189150. [PMID: 39903535 PMCID: PMC11949036 DOI: 10.1172/jci.insight.189150] [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: 11/12/2024] [Accepted: 01/31/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Lymphopenia and failure of lymphocytes to mount an early IFN-γ response correlate with increased mortality in COVID-19. Given the essential role of CD4 helper and CD8 cytotoxic cells in eliminating viral pathogens, this profound loss in lymphocytes may impair patients' ability to eliminate the virus. IL-7 is a pleiotropic cytokine that is obligatory for lymphocyte survival and optimal function. METHODS We conducted a prospective, double-blind, randomized, placebo-controlled trial of CYT107, recombinant human IL-7, in 109 critically ill, patients with lymphopenia who have COVID-19. The primary endpoint was to assess CYT107's effect on lymphocyte recovery with secondary clinical endpoints including safety, ICU and hospital length-of-stay, incidence of secondary infections, and mortality. RESULTS CYT107 was well tolerated without precipitating a cytokine storm or worsening pulmonary function. Absolute lymphocyte counts increased in both groups without a significant difference between CYT107 and placebo. Patients with COVID-19 receiving CYT107 but not concomitant antiviral medications, known inducers of lymphopenia, had a final lymphocyte count that was 43% greater than placebo (P = 0.067). There were significantly fewer treatment-emergent adverse events in CYT107 versus placebo-treated patients (P < 0.001), consistent with a beneficial drug effect. Importantly, CYT107-treated patients had 44% fewer hospital-acquired infections versus placebo-treated patients (P = 0.014). CONCLUSION Given that hospital-acquired infections are responsible for a large percentage of COVID-19 deaths, this effect of CYT107 to decrease nosocomial infections could substantially reduce late morbidity and mortality in this highly lethal disease. The strong safety profile of CYT107 and its excellent tolerability provide support for trials of CYT107 in other potential pandemic respiratory viral infections. TRIAL REGISTRATION NCT04379076, NCT04426201, NCT04442178, NCT04407689, NCT04927169. FUNDING Funding for the trial was provided by RevImmune and the Cancer Research Institute.
Collapse
Affiliation(s)
- Manu Shankar-Hari
- Department of Translational Critical Care Medicine, Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Scotland, United Kingdom
| | - Bruno Francois
- Medical-Surgical ICU & Inserm CIC 1435 Centre Hospitalier Universitaire, Limoges, France
| | - Kenneth E. Remy
- Department of Medicine, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Cristina Gutierrez
- Department of Critical Care Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Stephen Pastores
- Department of Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Thomas Daix
- Medical-Surgical ICU & Inserm CIC 1435 Centre Hospitalier Universitaire, Limoges, France
| | - Robin Jeannet
- Medical-Surgical ICU & Inserm CIC 1435 Centre Hospitalier Universitaire, Limoges, France
| | - Jane Blood
- Department of Anesthesiology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Andrew H. Walton
- Department of Anesthesiology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Reinaldo Salomao
- Department of Medicine, Universidade Federal de Sao Paulo (Unifesp), Sao Paulo, Brazil
| | - Georg Auzinger
- Department of Intensive Care Medicine, King’s College Hospital, London, United Kingdom
| | - David Striker
- Department of Critical Care Medicine, Missouri Baptist Medical Center, St. Louis, Missouri, USA
| | - Robert S. Martin
- Department of Critical Care Medicine, Missouri Baptist Medical Center, St. Louis, Missouri, USA
| | - Nitin J. Anand
- Department of Critical Care Medicine, Missouri Baptist Medical Center, St. Louis, Missouri, USA
| | - James Bosanquet
- Department of Critical Care Medicine, Missouri Baptist Medical Center, St. Louis, Missouri, USA
| | - Teresa Blood
- Department of Anesthesiology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | - Scott Brakenridge
- Department of Surgery, Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Lyle L. Moldawer
- Department of Surgery, Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | - Vidula Vachharajani
- Department of Critical Care Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Cassian Yee
- Department of Critical Care Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Felipe Dal-Pizzol
- Department of Medicine, Hospital Sao Jose, Criciuma, Santa Catarina, Brazil
| | | | | | | | - Richard Hotchkiss
- Department of Anesthesiology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
27
|
Schniederova M, Bobcakova A, Grendar M, Markocsy A, Ceres A, Cibulka M, Dobrota D, Jesenak M. Lymphocyte Inhibition Mechanisms and Immune Checkpoints in COVID-19: Insights into Prognostic Markers and Disease Severity. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:189. [PMID: 40005306 PMCID: PMC11857393 DOI: 10.3390/medicina61020189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025]
Abstract
Background and Objectives: Immune checkpoint inhibitors such as PD-1 and TIM-3 play an important role in regulating the host immune response and are proposed as potential prognostic markers and therapeutic targets in severe cases of COVID-19. We evaluated the expression of PD-1 and TIM-3 on T cells, as well as the concentration of sPD-1 in plasma, to clarify the role of these molecules in patients infected with SARS-CoV-2. Materials and Methods: In this retrospective observational study, we analysed the expression of PD-1 and TIM-3 on CD4+ and CD8+ T cells upon admission and after 7 days of hospitalisation in 770 adult patients. We also evaluated sPD-1 levels in the plasma of 145 patients at different stages of COVID-19 and of 11 control subjects. Molecules were determined using conventional flow cytometry and ELISA and the data were statistically processed. Results: We observed a significantly higher expression of PD-1 on CD4+ cells in deceased patients than in those with mild-to-moderate disease. All patients with COVID-19 exhibited a significantly higher expression of TIM-3 on both CD4+ and CD8+ T cells compared to controls. After 1 week of hospitalisation, there was no significant change in PD-1 or TIM-3 expression on CD4+ or CD8+ T cells across the studied groups. sPD-1 concentrations were not significantly different between survivors and non-survivors. Plasma sPD-1 levels did not correlate with PD-1 expression on T cells, but a significant correlation was observed between CD4+ PD-1 and CD8+ PD-1. Using machine-learning algorithms, we supported our observations and confirmed immunological variables capable of predicting survival, with AUC = 0.786. Conclusions: Analysis of the immune response may be useful for monitoring and predicting the course of COVID-19 upon admission. However, it is essential to evaluate complex immune parameters in conjunction with other key clinical and laboratory indicators.
Collapse
Affiliation(s)
- Martina Schniederova
- Institute of Clinical Immunology and Medical Genetics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia; (M.S.); (A.B.); (A.M.); (M.C.)
| | - Anna Bobcakova
- Institute of Clinical Immunology and Medical Genetics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia; (M.S.); (A.B.); (A.M.); (M.C.)
- Department of Pulmonology and Phthisiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia
- Department of Paediatrics and Adolescent Medicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia
| | - Marian Grendar
- Biomed—Centre for Biomedicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03659 Martin, Slovakia
| | - Adam Markocsy
- Institute of Clinical Immunology and Medical Genetics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia; (M.S.); (A.B.); (A.M.); (M.C.)
- Department of Pulmonology and Phthisiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia
| | - Andrej Ceres
- Institute of Clinical Immunology and Medical Genetics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia; (M.S.); (A.B.); (A.M.); (M.C.)
| | - Michal Cibulka
- Institute of Clinical Immunology and Medical Genetics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia; (M.S.); (A.B.); (A.M.); (M.C.)
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03659 Martin, Slovakia;
| | - Dusan Dobrota
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03659 Martin, Slovakia;
- Department of Clinical Biochemisty, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia
| | - Milos Jesenak
- Institute of Clinical Immunology and Medical Genetics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia; (M.S.); (A.B.); (A.M.); (M.C.)
- Department of Pulmonology and Phthisiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia
- Department of Paediatrics and Adolescent Medicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia
| |
Collapse
|
28
|
Paules CI, Nordwall JA, Shaw-Saliba K, Aberg JA, Gardner EM, Goodman AL, Kumarasamy N, Vasudeva S, Vock DM, North CM, Lundgren J, Aggarwal NR. Blood absolute lymphocyte count and trajectory are important in understanding severe COVID-19. BMC Infect Dis 2025; 25:67. [PMID: 39810077 PMCID: PMC11734232 DOI: 10.1186/s12879-024-10428-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Low blood absolute lymphocyte count (ALC) may predict severe COVID-19 outcomes. Knowledge gaps remain regarding the relationship of ALC trajectory with clinical outcomes and factors associated with lymphopenia. METHODS Our post hoc analysis of the Therapeutics for Inpatients with COVID-19 platform trial utilized proportional hazards models to assess relationships between Day (D) 0 lymphopenia (ALC < 0.9 cells/uL), D0 severe lymphopenia (ALC < 0.5 cells/uL) or lymphopenia trajectory between D0 and D5 with mortality and secondary infections, and with sustained recovery using Fine-Gray models. Logistic regression was used to assess relationships between clinical variables and D0 lymphopenia or lymphopenia trajectory. RESULTS D0 lymphopenia (1426/2579) and severe lymphopenia (636/2579) were associated with increased mortality (aHR 1.48; 1.08, 2.05, p = 0.016 and aHR 1.60; 1.20, 2.14, p = 0.001) and decreased recovery (aRRR 0.90; 0.82, 0.99, p = 0.033 and aRRR 0.78; 0.70, 0.87, p < 0.001 respectively). Trial participants with persistent D5 lymphopenia had increased mortality, and increased secondary infections, and participants with persistent or new lymphopenia had impaired recovery, as compared to participants with no lymphopenia. Persistent and new lymphopenia were associated with older age, male sex; prior immunosuppression, heart failure, aspirin use, and normal body mass index; biomarkers of organ damage (renal and lung), and ineffective immune response (elevated IL-6 and viral nucleocapsid antigen levels). Similar results were observed with severe lymphopenia. CONCLUSIONS Lymphopenia was predictive of severe COVID-19 outcomes, particularly when persistent or new during hospitalization. A better understanding of the underlying risk factors for lymphopenia will help illuminate disease pathogenesis and guide management strategies.
Collapse
Affiliation(s)
- Catharine I Paules
- Division of Infectious Diseases, Penn State Health Milton S. Hershey Medical Center, 500 University Drive, Hershey, PA, 17033, USA.
| | - Jacqueline A Nordwall
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Kathryn Shaw-Saliba
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Judith A Aberg
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Anna L Goodman
- MRC Clinical Trials Unit at University College London and CIDR, King's College London and Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - N Kumarasamy
- VHS Infectious Diseases Medical Centre, CART Clinical Research Site, Voluntary Health Services, Chennai, India
| | - Shikha Vasudeva
- Division of Infectious Diseases, VA Medical Center, Salem, VA, USA
| | - David M Vock
- Division of Biostatistics and Health Data Science, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Crystal M North
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jens Lundgren
- CHIP Center of Excellence for Health, Immunity, and Infections, Department of Infectious Diseases, University of Copenhagen, Righospitalet, Copenhagen, Denmark
| | - Neil R Aggarwal
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
29
|
dos Santos Brito WR, de Brito WB, dos Santos Ferreira F, Santana EGM, da Costa Lopes J, da Silva Graça Amoras E, Lima SS, dos Santos EF, da Costa FP, de Sarges KML, Cantanhede MHD, de Brito MTFM, da Silva ALS, de Meira Leite M, de Nazaré do Socorro de Almeida Viana M, Rodrigues FBB, da Silva R, Viana GMR, do Socorro Souza Chaves T, de Oliveira Lameira Veríssimo A, da Silva Carvalho M, Henriques DF, da Silva CP, Nunes JAL, Costa IB, Brasil-Costa I, Quaresma JAS, Cayres-Vallinoto IMV, Reis LO, Falcão LFM, dos Santos EJM, Vallinoto ACR, Queiroz MAF. Polymorphisms Influence the Expression of the Fas and FasL Genes in COVID-19. Int J Mol Sci 2025; 26:666. [PMID: 39859379 PMCID: PMC11765610 DOI: 10.3390/ijms26020666] [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: 11/15/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
The apoptotic molecule Fas and its ligand FasL are involved in the process of T-lymphocyte death, which may lead to lymphopenia, a characteristic of severe coronavirus disease 2019 (COVID-19). In this study, we investigated the influence of polymorphisms in the FAS and FASL genes, FAS and FASL gene expression, and plasma cytokine levels on COVID-19 severity and long COVID occurrence. A total of 116 individuals with severe COVID-19 and 254 with the non-severe form of the disease were evaluated. In the post-COVID-19 period, samples from 196 individuals with long COVID and 67 from people who did not have long COVID were included. Genotyping and quantification of gene expression were performed via real-time PCR, and cytokine measurement was performed via flow cytometry. The AA genotype for FAS rs1800682 (A/G) and the TT genotype for FASL rs763110 (C/T) were associated with increased FAS and FASL gene expression, respectively (p < 0.005). Higher plasma IFN-γ levels were associated with higher FAS and FASL gene expression (p < 0.05). Among individuals with non-severe COVID-19, carriers of the AA genotype for FAS rs1800682 (A/G) had higher levels of FAS expression, more symptoms, and higher IFN-γ levels (p < 0.05). No association of the evaluated markers with long COVID were observed. The AA genotype of FAS rs1800682 (A/G) and the TT genotype of FASL rs763110 (C/T) influence the levels of FAS and FASL gene expression. Higher gene expression of FAS and FASL may lead to greater inflammation in COVID-19 patients, with higher levels of IFN-γ and T lymphocyte death.
Collapse
Affiliation(s)
- Wandrey Roberto dos Santos Brito
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| | - William Botelho de Brito
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
| | - Fabiane dos Santos Ferreira
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| | - Emmanuelle Giuliana Mendes Santana
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
| | - Jeferson da Costa Lopes
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| | - Ednelza da Silva Graça Amoras
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
| | - Sandra Souza Lima
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
| | - Erika Ferreira dos Santos
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Flávia Póvoa da Costa
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Kevin Matheus Lima de Sarges
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Marcos Henrique Damasceno Cantanhede
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Mioni Thieli Figueiredo Magalhães de Brito
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Andréa Luciana Soares da Silva
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Mauro de Meira Leite
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Maria de Nazaré do Socorro de Almeida Viana
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Fabíola Brasil Barbosa Rodrigues
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Rosilene da Silva
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Giselle Maria Rachid Viana
- Laboratory of Basic Research on Malaria, Parasitology Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (G.M.R.V.); (T.d.S.S.C.)
| | - Tânia do Socorro Souza Chaves
- Laboratory of Basic Research on Malaria, Parasitology Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (G.M.R.V.); (T.d.S.S.C.)
- School of Medicine, Institute of Medical Sciences, Federal University of Pará, Belém 66075-110, Brazil
| | | | | | - Daniele Freitas Henriques
- Arbovirology and Hemorrhagic Fevers Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (D.F.H.); (C.P.d.S.)
| | - Carla Pinheiro da Silva
- Arbovirology and Hemorrhagic Fevers Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (D.F.H.); (C.P.d.S.)
| | - Juliana Abreu Lima Nunes
- Laboratory of Immunology, Section of Virology, Instituto Evandro Chagas, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (J.A.L.N.); (I.B.C.); (I.B.-C.)
| | - Iran Barros Costa
- Laboratory of Immunology, Section of Virology, Instituto Evandro Chagas, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (J.A.L.N.); (I.B.C.); (I.B.-C.)
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua 66093-020, Brazil;
| | - Igor Brasil-Costa
- Laboratory of Immunology, Section of Virology, Instituto Evandro Chagas, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (J.A.L.N.); (I.B.C.); (I.B.-C.)
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua 66093-020, Brazil;
| | - Juarez Antônio Simões Quaresma
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua 66093-020, Brazil;
- Center of Biological and Health Sciences, University of the State of Pará, Belém 66087-670, Brazil;
| | - Izaura Maria Vieira Cayres-Vallinoto
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| | - Leonardo Oliveira Reis
- UroScience, Faculty of Medical Sciences, State University of Campinas, Campinas 13083-590, Brazil;
- ImmunOncology, Pontifical Catholic University of Campinas, Campinas 13060-904, Brazil
| | - Luiz Fábio Magno Falcão
- Center of Biological and Health Sciences, University of the State of Pará, Belém 66087-670, Brazil;
| | - Eduardo José Melo dos Santos
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Antonio Carlos Rosário Vallinoto
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua 66093-020, Brazil;
| | - Maria Alice Freitas Queiroz
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| |
Collapse
|
30
|
Tu TH, Bennani FE, Masroori N, Liu C, Nemati A, Rozza N, Grunbaum AM, Kremer R, Milhalcioiu C, Roy DC, Rudd CE. The identification of a SARs-CoV2 S2 protein derived peptide with super-antigen-like stimulatory properties on T-cells. Commun Biol 2025; 8:14. [PMID: 39762551 PMCID: PMC11704208 DOI: 10.1038/s42003-024-07350-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Severe COVID-19 can trigger a cytokine storm, leading to acute respiratory distress syndrome (ARDS) with similarities to superantigen-induced toxic shock syndrome. An outstanding question is whether SARS-CoV-2 protein sequences can directly induce inflammatory responses. In this study, we identify a region in the SARS-CoV-2 S2 spike protein with sequence homology to bacterial super-antigens (termed P3). Computational modeling predicts P3 binding to sites on MHC class I/II and the TCR that partially overlap with sites for the binding of staphylococcal enterotoxins B and H. Like SEB and SEH derived peptides, P3 stimulated 25-40% of human CD4+ and CD8 + T-cells, increasing IFN-γ and granzyme B production. viSNE and SPADE profiling identified overlapping and distinct IFN-γ+ and GZMB+ subsets. The super-antigenic properties of P3 were further evident by its selective expansion of T-cells expressing specific TCR Vα and Vβ chain repertoires. In vivo experiments in mice revealed that the administration of P3 led to a significant upregulation of proinflammatory cytokines IL-1β, IL-6, and TNF-α. While the clinical significance of P3 in COVID-19 remains unclear, its homology to other mammalian proteins suggests a potential role for this peptide family in human inflammation and autoimmunity.
Collapse
Affiliation(s)
- Thai Hien Tu
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
| | - Fatima Ezzahra Bennani
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Nasser Masroori
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Institut Universitaire d'Hématologie-Oncologie & Thérapie Cellulaire de Montréal, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Chen Liu
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
| | - Atena Nemati
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
| | - Nicholas Rozza
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Amichai Meir Grunbaum
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
| | - Richard Kremer
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
| | - Catalin Milhalcioiu
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
- Department of Medical Oncology, McGill University Health Center, Montreal, QC, Canada
| | - Denis-Claude Roy
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Institut Universitaire d'Hématologie-Oncologie & Thérapie Cellulaire de Montréal, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Christopher E Rudd
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada.
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada.
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada.
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco.
- Institut Universitaire d'Hématologie-Oncologie & Thérapie Cellulaire de Montréal, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada.
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada.
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
31
|
Okada K, Tagami T, Otaguro T, Hayakawa M, Yamakawa K, Endo A, Ogura T, Hirayama A, Yasunaga H, Hara Y. Early lymphopenia as a predictor of COVID-19 outcomes: A multicenter cohort study. Acute Med Surg 2025; 12:e70044. [PMID: 40125412 PMCID: PMC11928683 DOI: 10.1002/ams2.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/19/2025] [Accepted: 01/26/2025] [Indexed: 03/25/2025] Open
Abstract
Introduction Lymphopenia is recognized as a biomarker for predicting outcomes in coronavirus disease (COVID-19). However, the optimal timing for its observation remains uncertain. We investigated the association between early lymphopenia and COVID-19 prognosis, as well as the relationship between lymphocyte count trends and disease outcomes. Methods We analyzed data from the J-RECOVER study, a multicenter retrospective cohort study in Japan, encompassing patients with COVID-19 between January and September 2020. The patients were categorized into lymphopenia (LP) (<800 cells/μL) and non-lymphopenia (NL) (≥800 cells/μL) groups based on the lymphocyte counts between days 1 and 4 post-onset. They were further divided into "persistent," "recovered," "exacerbated," and "stable" groups based on lymphocyte counts between days 7 and 10. The primary outcome was the in-hospital mortality. The Cox proportional hazard regression was used for the analysis. Results Of 995 enrolled patients, 212 patients (21.3%) were classified into the LP group. LP was significantly associated with in-hospital mortality (hazard ratio [HR] 2.32, [95% CI 1.39 to 3.87], p-value 0.001). In both the LP and NL groups, lower lymphocyte counts between 7 and 10 days-categorized as the "persistent" and "exacerbated" groups-was associated with in-hospital mortality (HR 4.65, [95% CI 2.07 to 10.47], p-value <0.001, and HR 5.59, [95% CI 2.24 to 13.97], p-value <0.001, respectively). Conclusions Early lymphopenia is predictive of poor prognosis in patients with COVID-19. A declining lymphocyte count trend post-onset further indicates disease deterioration.
Collapse
Affiliation(s)
- Kazuhiro Okada
- Shock and Trauma CenterNippon Medical School Chiba Hokusoh HospitalChibaJapan
- Department of Emergency and Critical Care MedicineNippon Medical SchoolTokyoJapan
| | - Takashi Tagami
- Department of Emergency and Critical Care MedicineNippon Medical SchoolTokyoJapan
- Department of Emergency and Critical Care MedicineNippon Medical School Musashikosugi HospitalKawasakiKanagawaJapan
- Department of Clinical Epidemiology and Health Economics, School of Public HealthThe University of TokyoTokyoJapan
| | - Takanobu Otaguro
- Department of Emergency and Critical Care MedicineNagasaki University HospitalNagasakiJapan
| | - Mineji Hayakawa
- Department of Emergency MedicineHokkaido University HospitalSapporoHokkaidoJapan
| | - Kazuma Yamakawa
- Department of Emergency and Critical Care MedicineOsaka Medical and Pharmaceutical UniversityOsakaJapan
| | - Akira Endo
- Department of Acute Critical Care MedicineTsuchiura Kyodo General HospitalIbarakiJapan
| | - Takayuki Ogura
- Tochigi Prefectural Emergency and Critical Care Center, Imperial Gift Foundation SaiseikaiUtsunomiya HospitalTochigiJapan
| | - Atsushi Hirayama
- Public Health, Department of Social Medicine, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Hideo Yasunaga
- Department of Clinical Epidemiology and Health Economics, School of Public HealthThe University of TokyoTokyoJapan
| | - Yoshiaki Hara
- Shock and Trauma CenterNippon Medical School Chiba Hokusoh HospitalChibaJapan
- Department of Emergency and Critical Care MedicineNippon Medical SchoolTokyoJapan
| |
Collapse
|
32
|
Khan Y, Van den Borre L, Smedt DD, Verhaeghe N, Devleesschauwer B, Deboosere P, Vanthomme K, Gadeyne S. A Nationwide Exploration of Social Inequalities in Cancer Mortality Amidst the COVID-19 Pandemic in Belgium. Cancer Med 2025; 14:e70487. [PMID: 39778068 PMCID: PMC11707617 DOI: 10.1002/cam4.70487] [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: 04/11/2024] [Revised: 11/09/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND The COVID-19 pandemic disrupted global health systems, impacting cancer care and potentially increasing cancer mortality, especially among socioeconomically disadvantaged individuals. We aimed to assess changes in cancer mortality from March 1 to December 31, 2020 relative to the same period in 2019, and to examine potential shifts in cancer mortality's social disparities during the same time frame. METHODS We used nationwide individually linked cancer mortality data from the Belgian National Register, the Census 2011, and the tax register. Analyses were stratified by age group (45-59 years, 60-74 years, 75+ years) and sex across all cancer types, including breast, colorectal, lung, pancreatic, and prostate. Direct age-standardized mortality rates were calculated in 2019 and 2020 to calculate absolute and relative changes in cancer mortality by social indicators. Relative inequalities in cancer mortality by social groups were calculated for both time frames using Poisson regression. Sensitivity analysis considered any mention of specified cancer groups on the Belgian death certificate. RESULTS For both overall and site-specific cancers, our study found decreases in cancer mortality during the pandemic's early stages, particularly among individuals aged 75 and older. These changes did not significantly alter established socioeconomic patterns in cancer mortality. CONCLUSIONS Reductions in reported cancer deaths in 2020 may reflect COVID-19 prioritization in cause-of-death coding and its role as a competing risk, rather than true declines. Persistent educational disparities emphasize the need for continued policy and healthcare collaboration, with future research focused on the pandemic's long-term effects on cancer mortality and social inequalities.
Collapse
Affiliation(s)
- Yasmine Khan
- Department of Public Health and Primary CareGhent UniversityGhentBelgium
- Department of Epidemiology and Public Health, SciensanoBrusselsBelgium
- Interface Demography, Department of SociologyVrije Universiteit BrusselBrusselsBelgium
| | - Laura Van den Borre
- Department of Epidemiology and Public Health, SciensanoBrusselsBelgium
- Interface Demography, Department of SociologyVrije Universiteit BrusselBrusselsBelgium
| | - Delphine De Smedt
- Department of Public Health and Primary CareGhent UniversityGhentBelgium
| | - Nick Verhaeghe
- Department of Public Health and Primary CareGhent UniversityGhentBelgium
| | - Brecht Devleesschauwer
- Department of Epidemiology and Public Health, SciensanoBrusselsBelgium
- Department of Translational Physiology, Infectiology and Public HealthGhent UniversityMerelbekeBelgium
| | - Patrick Deboosere
- Interface Demography, Department of SociologyVrije Universiteit BrusselBrusselsBelgium
| | - Katrien Vanthomme
- Department of Public Health and Primary CareGhent UniversityGhentBelgium
| | - Sylvie Gadeyne
- Interface Demography, Department of SociologyVrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
33
|
Tourki B, Jia M, Karampitsakos T, Vera IM, Arsenault A, Fatima Z, Perrot CY, Allen D, Farsaei F, Rutenberg D, Bandyopadhyay D, Restrepo-Jaramillo R, Qureshi MR, Patel K, Tzouvelekis A, Kapetanaki M, Juan-Guardela B, Kim K, Benos PV, Herazo-Maya JD. Convergent and divergent immune aberrations in COVID-19, post-COVID-19-interstitial lung disease, and idiopathic pulmonary fibrosis. Am J Physiol Cell Physiol 2025; 328:C199-C211. [PMID: 39510135 PMCID: PMC12204034 DOI: 10.1152/ajpcell.00528.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/15/2024]
Abstract
We aimed to study transcriptional and phenotypic changes in circulating immune cells associated with increased risk of mortality in COVID-19, resolution of pulmonary fibrosis in post-COVID-19-interstitial lung disease (ILD), and persistence of idiopathic pulmonary fibrosis (IPF). Whole blood and peripheral blood mononuclear cells (PBMCs) were obtained from 227 subjects with COVID-19, post-COVID-19 interstitial lung disease (ILD), IPF, and controls. We measured a 50-gene signature (nCounter, Nanostring) previously found to be predictive of IPF and COVID-19 mortality along with plasma levels of several biomarkers by Luminex. In addition, we performed single-cell RNA sequencing (scRNA-seq) in PBMCs (10x Genomics) to determine the cellular source of the 50-gene signature. We identified the presence of three genomic risk profiles in COVID-19 based on the 50-gene signature associated with low-, intermediate-, or high-risk of mortality and with significant differences in proinflammatory and profibrotic cytokines. Patients with COVID-19 in the high-risk group had increased expression of seven genes in CD14+HLA-DRlowCD163+ monocytic-myeloid-derived suppressive cells (7Gene-M-MDSCs) and decreased expression of 43 genes in CD4 and CD8 T cell subsets. The loss of 7Gene-M-MDSCs and increased expression of these 43 genes in T cells was seen in survivors with post-COVID-19-ILD. On the contrary, patients with IPF had low expression of the 43 genes in CD4 and CD8 T cells. Collectively, we showed that a 50-gene, high-risk profile, predictive of IPF and COVID-19 mortality is characterized by a genomic imbalance in monocyte and T-cell subsets. This imbalance reverses in survivors with post-COVID-19-ILD highlighting genomic differences between post-COVID-19-ILD and IPF.NEW & NOTEWORTHY Changes in the 50-gene signature, reflective of increase in CD14+HLA-DRlowCD163+ monocytes and decrease in CD4 and CD8 T cells, are associated with increased mortality in COVID-19. A reversal of this pattern can be seen in post-COVID-19-ILD, whereas its persistence can be seen in IPF. Modulating the imbalance between HLA-DRlow monocytes and T cell subsets should be investigated as a potential strategy to treat pulmonary fibrosis associated with severe COVID-19 and progressive IPF.
Collapse
Affiliation(s)
- Bochra Tourki
- Ubben Center for Pulmonary Fibrosis Research, Division of Pulmonary, Critical Care and Sleep Medicine. Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Minxue Jia
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Joint Carnegie Mellon - University of Pittsburgh Computational Biology Ph.D. Program, Pittsburgh, Pennsylvania, USA. Program in Computational Biology, Pittsburgh, PA, USA
| | - Theodoros Karampitsakos
- Ubben Center for Pulmonary Fibrosis Research, Division of Pulmonary, Critical Care and Sleep Medicine. Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Iset M Vera
- Division of infectious Disease, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Alyssa Arsenault
- Ubben Center for Pulmonary Fibrosis Research, Division of Pulmonary, Critical Care and Sleep Medicine. Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Zainab Fatima
- Ubben Center for Pulmonary Fibrosis Research, Division of Pulmonary, Critical Care and Sleep Medicine. Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Carole Y Perrot
- Ubben Center for Pulmonary Fibrosis Research, Division of Pulmonary, Critical Care and Sleep Medicine. Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Dylan Allen
- Division of infectious Disease, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Forouzandeh Farsaei
- Division of infectious Disease, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - David Rutenberg
- Division of infectious Disease, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Debabrata Bandyopadhyay
- Ubben Center for Pulmonary Fibrosis Research, Division of Pulmonary, Critical Care and Sleep Medicine. Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Center for Advanced Lung Disease and Lung Transplant Program. Tampa General Hospital, FL, USA
| | - Ricardo Restrepo-Jaramillo
- Ubben Center for Pulmonary Fibrosis Research, Division of Pulmonary, Critical Care and Sleep Medicine. Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Center for Advanced Lung Disease and Lung Transplant Program. Tampa General Hospital, FL, USA
| | - Muhammad R. Qureshi
- Ubben Center for Pulmonary Fibrosis Research, Division of Pulmonary, Critical Care and Sleep Medicine. Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Center for Advanced Lung Disease and Lung Transplant Program. Tampa General Hospital, FL, USA
| | - Kapilkumar Patel
- Ubben Center for Pulmonary Fibrosis Research, Division of Pulmonary, Critical Care and Sleep Medicine. Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Center for Advanced Lung Disease and Lung Transplant Program. Tampa General Hospital, FL, USA
| | | | - Maria Kapetanaki
- Department of Epidemiology, College of Public Health, and Health Professions, and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Brenda Juan-Guardela
- Ubben Center for Pulmonary Fibrosis Research, Division of Pulmonary, Critical Care and Sleep Medicine. Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Kami Kim
- Division of infectious Disease, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Panayiotis V Benos
- Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Joint Carnegie Mellon - University of Pittsburgh Computational Biology Ph.D. Program, Pittsburgh, Pennsylvania, USA. Program in Computational Biology, Pittsburgh, PA, USA
- Department of Epidemiology, College of Public Health, and Health Professions, and College of Medicine, University of Florida, Gainesville, FL, USA
| | - Jose D. Herazo-Maya
- Ubben Center for Pulmonary Fibrosis Research, Division of Pulmonary, Critical Care and Sleep Medicine. Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Center for Advanced Lung Disease and Lung Transplant Program. Tampa General Hospital, FL, USA
| |
Collapse
|
34
|
Mihuta C, Socaci A, Hogea P, Tudorache E, Mihuta MS, Oancea C. Colliding Challenges Part 2: An Analysis of SARS-CoV-2 Infection in Patients with Extrapulmonary Tuberculosis Versus SARS-CoV-2 Infection Alone. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:2071. [PMID: 39768950 PMCID: PMC11677740 DOI: 10.3390/medicina60122071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/08/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
Background and Objectives: Coinfection with SARS-CoV-2 and extrapulmonary tuberculosis (extraPTB) presents unique clinical challenges due to dual inflammatory responses and potential differences in patient profiles compared to those with SARS-CoV-2 infection alone. This study uniquely contributes to the underexplored interaction between extraPTB and SARS-CoV-2, focusing on systemic inflammation as a critical determinant of outcomes. Materials and Methods: This retrospective, cross-sectional study included 123 patients aged 19-91 years, hospitalized at Victor Babeș Hospital in Timișoara from March 2020 to March 2022. We compared 23 extraPTB and SARS-CoV-2 coinfected patients with 100 age-matched SARS-CoV-2-only patients. Clinical records were examined for demographic, clinical, and laboratory data. Results: The coinfected group was younger, with 65% under 40 years, and presented significantly higher IL-6, PCT, and transaminase levels. Coexisting COPD and type 2 diabetes were independent predictors of coinfection. A higher SpO2 at diagnosis was positively associated with coinfection likelihood (OR = 5.37), while CT scores indicated less pulmonary involvement in coinfected patients. Non-fatal outcomes were more frequent in the coinfection group (95.7% sensitivity), and only one coinfected patient had a fatal outcome versus 17 in the SARS-CoV-2-only group. Low SpO2 and elevated IL-6 were significant predictors of mortality, with severe symptoms tripling fatality odds. Conclusions: Coinfection with extraPTB and SARS-CoV-2 is associated with younger age, heightened systemic inflammation, and longer hospital stays but does not significantly increase mortality risk compared to SARS-CoV-2 alone. These findings underscore the importance of monitoring systemic inflammatory markers and developing tailored management strategies to improve long-term care outcomes for coinfected patients, especially in resource-limited settings.
Collapse
Affiliation(s)
- Camil Mihuta
- Department of Doctoral Studies, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Clinical Hospital for Infectious Diseases and Pneumology “Dr. Victor Babes”, 300041 Timisoara, Romania; (P.H.); (E.T.); (C.O.)
| | - Adriana Socaci
- Clinical Hospital for Infectious Diseases and Pneumology “Dr. Victor Babes”, 300041 Timisoara, Romania; (P.H.); (E.T.); (C.O.)
- Department of Biology and Life Sciences, Faculty of Medicine, “Vasile Goldis” Western University of Arad, 310025 Arad, Romania
| | - Patricia Hogea
- Clinical Hospital for Infectious Diseases and Pneumology “Dr. Victor Babes”, 300041 Timisoara, Romania; (P.H.); (E.T.); (C.O.)
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases (CRIPMRD), “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Department of Pulmonology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Emanuela Tudorache
- Clinical Hospital for Infectious Diseases and Pneumology “Dr. Victor Babes”, 300041 Timisoara, Romania; (P.H.); (E.T.); (C.O.)
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases (CRIPMRD), “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Department of Pulmonology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Monica Simina Mihuta
- Department of Pediatrics, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Center of Molecular Research in Nephrology and Vascular Disease, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Cristian Oancea
- Clinical Hospital for Infectious Diseases and Pneumology “Dr. Victor Babes”, 300041 Timisoara, Romania; (P.H.); (E.T.); (C.O.)
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases (CRIPMRD), “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Department of Pulmonology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| |
Collapse
|
35
|
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
|
36
|
Wu Y, Li Y, Zhou Y, Bai X, Liu Y. Bioinformatics and systems-biology approach to identify common pathogenic mechanisms for COVID-19 and systemic lupus erythematosus. Autoimmunity 2024; 57:2304826. [PMID: 38332666 DOI: 10.1080/08916934.2024.2304826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 01/07/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND The Coronavirus disease 2019 (COVID-19) pandemic has brought a heavy burden to the world, interestingly, it shares many clinical symptoms with systemic lupus erythematosus (SLE). It is unclear whether there is a similar pathological process between COVID-9 and SLE. In addition, we don't know how to treat SLE patients with COVID-19. In this study, we analyse the potential similar pathogenesis between SLE and COVID-19 and explore their possible drug regimens using bioinformatics and systems biology approaches. METHODS The common differentially expressed genes (DEGs) were extracted from the COVID-19 datasets and the SLE datasets for functional enrichment, pathway analysis and candidate drug analysis. RESULT Based on the two transcriptome datasets between COVID-19 and SLE, 325 common DEGs were selected. Hub genes were identified by protein-protein interaction (PPI) analysis. few found a variety of similar functional changes between COVID-19 and SLE, which may be related to the pathogenesis of COVID-19. Besides, we explored the related regulatory networks. Then, through drug target matching, we found many candidate drugs for patients with COVID-19 only or COVID-19 combined with SLE. CONCLUSION COVID-19 and SLE patients share many common hub genes, related pathways and regulatory networks. Based on these common targets, we found many potential drugs that could be used in treating patient with COVID-19 or COVID-19 combined with SLE.
Collapse
Affiliation(s)
- Yinlan Wu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yanhong Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Zhou
- Department of Respiratory and Critical Care Medicine, Chengdu First People's Hospital, Chengdu, China
| | - Xiufeng Bai
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Human Disease and Immunotherapies, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
37
|
Ahmed AEA, Abuhamdah SM, Hassan MH, Rashwan NI, Abd-Elmawgood EA, Mansour H, Sherkawy HS, Rizk SG. Clinical, biochemical, and genetic study of TACE/TNF-α/ACE signaling pathway in pediatric COVID-19 infection. Clin Exp Pediatr 2024; 67:704-717. [PMID: 39600173 PMCID: PMC11621736 DOI: 10.3345/cep.2024.00941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Pediatric patients infected with coronavirus disease 2019 (COVID-19) have unique clinical characteristics. Tumor necrosis factor (TNF) is a proinflammatory cytokine that greatly contributes to tumor pathogenesis. PURPOSE To describe the presenting characteristics of COVID-19 infection among pediatric patients, and investigate the possible role of the TNF-α signaling pathway. METHODS This prospective case-control study included 50 Egyptian pediatric patients with COVID-19 and 50 healthy controls. Clinical, laboratory, and radiological assessments were performed. Serum TNF-alpha (TNF-α), TNF-α-converting enzyme (TACE), and angiotensin-converting enzyme 2 (ACE2) were measured using enzyme-linked immunosorbent assay. ACE (I/D) (rs4646994), ACE2 rs2285666, and TNF-α-308G/A single nucleotide polymorphisms (SNPs) were performed using conventional polymerase chain reaction techniques with or without restriction fragment length polymorphism. RESULTS The median age was 1 year (interquartile range [IQR], 0.31-2.50 years) in the case group and 1.45 years (IQR, 1.00-3.00) in the control group. The main presenting symptoms were fever (92%), dry cough (74%), and dyspnea (72%). The lymphocytic count was normal in 14 patients (28%), decreased in 16 patients (32%), and increased in 20 patients (40%) of the case group. Positive chest computed tomography finding of COVID-19 infection were demonstrated among 40% of patients using COVID-19 Reporting and Data System categories (ground-glass opacity with or without consolidations in the lungs). There were significant increased serum TACE and TNF-α with decreased ACE2 levels among cases versus controls (P< 0.001). The GG genotype and G allele of the TNF-α-308G/A SNP were significantly higher in patients than in controls (P<0.05 for both), with insignificant differences in genotype and allelic frequencies in the ACE (I/D) (rs4646994) and ACE2 rs2285666 SNPs. CONCLUSION The TNF signaling pathway was significantly activated in pediatric COVID-19 infection. Only the TNF-α-308G/A SNP was significantly associated with pediatric COVID-19 infection.
Collapse
Affiliation(s)
- Ahmed El-Abd Ahmed
- Department of Pediatrics, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Sawsan M.A. Abuhamdah
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
- Department of Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Abu Dhabi, UAE
| | - Mohammed H. Hassan
- Department of Medical Biochemistry, Faculty of Medicine, South Valley University, Qena, Egypt
- Department of Medical Biochemistry, Medicine and Surgery Program, South Valley National University, Qena, Egypt
| | - Nagwan I. Rashwan
- Department of Pediatrics, Faculty of Medicine, South Valley University, Qena, Egypt
| | | | - Haggagy Mansour
- Department of Chest Diseases and Tuberculosis, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Hoda S. Sherkawy
- Department of Medical Biochemistry, Faculty of Medicine, Aswan University, Aswan, Egypt
| | - Shymaa G. Rizk
- Department of Pediatrics, Faculty of Medicine, South Valley University, Qena, Egypt
| |
Collapse
|
38
|
Agrawal S, Kasarpalkar N, Ghosh S, Paradkar G, Daund V, Bhowmick S, Chitalia V, Rao P, Sankpal A, Kalsurkar V, Shah K, Khan S, Patil A, Jagtap D, Khandkar O, Kaneria M, Mahale SD, Sachdeva G, Bhor VM, Shastri J, Patel V. Integrated viral and immune monitoring in a prospective COVID-19 cohort from India. J Leukoc Biol 2024; 116:1568-1577. [PMID: 39219468 DOI: 10.1093/jleuko/qiae187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 06/07/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
In this study, we report on longitudinal kinetics of cellular immune subsets following SARS-CoV-2 infection in a cohort of hospitalized individuals and evaluate the interplay of these profiles with infecting viral variants, humoral immunity including neutralizing responses, vaccination history, and clinical outcomes. A cohort of 121 SARS-CoV-2-infected individuals exhibiting varying disease states were prospectively evaluated for lymphopenic profiles, antiviral humoral responses and infecting viral variants for a period of up to 90 d spanning the period of February 2021 to January 2022 (second and third waves of infection). A total of 51 participants received at least 1 vaccine dose of indigenous vaccines (Covishield or Covaxin) prior to recruitment. When stratified in terms of mortality, B and natural killer cells, in contrast to the T cell compartment, did not recover from nadir levels in nonsurvivors who were largely unvaccinated. No discriminatory signature was identified for nonsurvivors in terms of anti-nucleocapsid or anti-S1-RBD IgG chemiluminescent immunoassay profiles including GenScript S1-RBD assays. Evaluation of sVCAM and sMAdCAM revealed opposing dynamics that correlated with disease severity and convalescence respectively. Viral variant analysis revealed Delta and Omicron variants to comprise the majority of the infections, which reflected national transmission kinetics during the period of recruitment. Our results demonstrate the importance of monitoring circulating biomarkers for convalescence as well as mortality in COVID-19 progression. Delta variants of SARS-CoV-2 clearly demonstrated increased pathogenicity and warrants sustained viral surveillance for re-emergence of these strains. Our findings with respect to vaccination advocate for continued vaccine development and administration of COVID-19 vaccines.
Collapse
Affiliation(s)
- Sachee Agrawal
- Department of Microbiology, B.Y.L. Nair Charitable Hospital, Mumbai 400008, India
| | - Nandini Kasarpalkar
- Viral Immunopathogenesis Lab, Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
- Department of Molecular Immunology and Microbiology, Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Sayantani Ghosh
- Viral Immunopathogenesis Lab, Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Gaurav Paradkar
- Department of Molecular Immunology and Microbiology, Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Vaibhav Daund
- Viral Immunopathogenesis Lab, Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Shilpa Bhowmick
- Viral Immunopathogenesis Lab, Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Vidushi Chitalia
- Viral Research and Diagnostic Laboratory, Kasturba Hospital for Infectious Diseases, Mumbai 400011, India
| | - Priyanka Rao
- Topiwala National Medical College and B.Y.L. Nair Charitable Hospital, Mumbai 400008, India
| | - Ashwini Sankpal
- Topiwala National Medical College and B.Y.L. Nair Charitable Hospital, Mumbai 400008, India
| | - Varsha Kalsurkar
- Department of Molecular Immunology and Microbiology, Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Karan Shah
- Viral Immunopathogenesis Lab, Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
- Department of Molecular Immunology and Microbiology, Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Sameen Khan
- Viral Immunopathogenesis Lab, Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Ashwini Patil
- Department of Molecular Immunology and Microbiology, Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Dhanashree Jagtap
- Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Omkar Khandkar
- Topiwala National Medical College and B.Y.L. Nair Charitable Hospital, Mumbai 400008, India
| | - Mala Kaneria
- Topiwala National Medical College and B.Y.L. Nair Charitable Hospital, Mumbai 400008, India
| | - Smita D Mahale
- Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Geetanjali Sachdeva
- Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Vikrant M Bhor
- Department of Molecular Immunology and Microbiology, Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| | - Jayanthi Shastri
- Topiwala National Medical College and B.Y.L. Nair Charitable Hospital, Mumbai 400008, India
- Viral Research and Diagnostic Laboratory, Kasturba Hospital for Infectious Diseases, Mumbai 400011, India
| | - Vainav Patel
- Viral Immunopathogenesis Lab, Indian Council of Medical Research-National Institute for Research in Reproductive and Child Health, Mumbai 400012, India
| |
Collapse
|
39
|
Wang Y, Song Z, Ran P, Xiang H, Xu Z, Xu N, Deng M, Zhu L, Yin Y, Feng J, Ding C, Yang W. Serum proteome reveals distinctive molecular features of H7N9- and SARS-CoV-2-infected patients. Cell Rep 2024; 43:114900. [PMID: 39487987 DOI: 10.1016/j.celrep.2024.114900] [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: 04/29/2024] [Revised: 08/02/2024] [Accepted: 10/07/2024] [Indexed: 11/04/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has reminded us of human infections with the H7N9 virus and has raised questions related to the clinical and molecular pathophysiological diversity between the two diseases. Here, we performed a proteomic approach on sera samples from patients with H7N9-virus or SARS-CoV-2-virus infection and healthy controls. Compared to SARS-CoV-2, H7N9-virus infection caused elevated neutrophil concentrations, T cell exhaustion, and increased cytokine/interleukin secretion. Cell-type deconvolution and temporal analysis revealed that T cells and neutrophils could regulate the core immunological trajectory and influence the prognosis of patients with severe H7N9-virus infection. Elevated tissue-enhanced proteins combined with alterations of clinical biochemical indexes suggested that H7N9 infection induced more severe inflammatory organ injury and dysfunction in the liver and intestine. Further mechanical analysis revealed that the high concentration of neutrophils might impact the intestinal enterocyte cells through cytokine-receptor interaction, leading to intestinal damage in patients with H7N9-virus infection.
Collapse
Affiliation(s)
- Yunzhi Wang
- Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200092, China; Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Zhigang Song
- Institutes of Biomedical Sciences, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou) and Shanghai Public Health Clinical Center, Fudan University, Shanghai 200438, China
| | - Peng Ran
- Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200092, China; Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Hang Xiang
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Ziyan Xu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Ning Xu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Mengjie Deng
- Ruijin Hospital, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingli Zhu
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Yanan Yin
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Jinwen Feng
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200433, China
| | - Chen Ding
- Center for Cell and Gene Therapy, Clinical Research Center for Cell-based Immunotherapy, Shanghai Pudong Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200433, China; Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xingjiang Medical University, Xingjiang Key Laboratory of Translational Biomedical Engineering, Urumqi 830000, P. R. China.
| | - Wenjun Yang
- Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai 200092, China.
| |
Collapse
|
40
|
Wang K, Nie Y, Maguire C, Syphurs C, Sheen H, Karoly M, Lapp L, Gygi JP, Jayavelu ND, Patel RK, Hoch A, Corry D, Kheradmand F, McComsey GA, Fernandez-Sesma A, Simon V, Metcalf JP, Higuita NIA, Messer WB, Davis MM, Nadeau KC, Kraft M, Bime C, Schaenman J, Erle D, Calfee CS, Atkinson MA, Brackenridge SC, Hafler DA, Shaw A, Rahman A, Hough CL, Geng LN, Ozonoff A, Haddad EK, Reed EF, van Bakel H, Kim-Schultz S, Krammer F, Wilson M, Eckalbar W, Bosinger S, Langelier CR, Sekaly RP, Montgomery RR, Maecker HT, Krumholz H, Melamed E, Steen H, Pulendran B, Augustine AD, Cairns CB, Rouphael N, Becker PM, Fourati S, Shannon CP, Smolen KK, Peters B, Kleinstein SH, Levy O, Altman MC, Iwasaki A, Diray-Arce J, Ehrlich LIR, Guan L. Unraveling SARS-CoV-2 Host-Response Heterogeneity through Longitudinal Molecular Subtyping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624784. [PMID: 39651165 PMCID: PMC11623532 DOI: 10.1101/2024.11.22.624784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Hospitalized COVID-19 patients exhibit diverse immune responses during acute infection, which are associated with a wide range of clinical outcomes. However, understanding these immune heterogeneities and their links to various clinical complications, especially long COVID, remains a challenge. In this study, we performed unsupervised subtyping of longitudinal multi-omics immunophenotyping in over 1,000 hospitalized patients, identifying two critical subtypes linked to mortality or mechanical ventilation with prolonged hospital stay and three severe subtypes associated with timely acute recovery. We confirmed that unresolved systemic inflammation and T-cell dysfunctions were hallmarks of increased severity and further distinguished patients with similar acute respiratory severity by their distinct immune profiles, which correlated with differences in demographic and clinical complications. Notably, one critical subtype (SubF) was uniquely characterized by early excessive inflammation, insufficient anticoagulation, and fatty acid dysregulation, alongside higher incidences of hematologic, cardiac, and renal complications, and an elevated risk of long COVID. Among the severe subtypes, significant differences in viral clearance and early antiviral responses were observed, with one subtype (SubC) showing strong early T-cell cytotoxicity but a poor humoral response, slower viral clearance, and greater risks of chronic organ dysfunction and long COVID. These findings provide crucial insights into the complex and context-dependent nature of COVID-19 immune responses, highlighting the importance of personalized therapeutic strategies to improve both acute and long-term outcomes.
Collapse
|
41
|
Chen CC, Lin YA, Liu KT, Huang CY, Shih CM, Lee YT, Pan JL, Lee AW. Navigating SARS-CoV-2-related immunopathology in Crohn's disease: from molecular mechanisms to therapeutic challenges. Virol J 2024; 21:288. [PMID: 39538233 PMCID: PMC11562311 DOI: 10.1186/s12985-024-02529-1] [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: 11/30/2023] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) not only posed major health and economic burdens to international societies but also threatens patients with comorbidities and underlying autoimmune disorders, including Crohn's disease (CD) patients. As the vaccinated population is gradually relieved from the stress of the latest omicron variant of SARS-CoV-2 due to competent immune responses, the anxiety of CD patients, especially those on immunosuppressive treatment, has not subsided. Whether the use of immunosuppressants for remission of CD outweighs the potential risk of severe coronavirus disease 2019 (COVID-19) has long been discussed. Thus, for the best benefit of CD patients, our primary goal in this study was to navigate the clinical management of CD during the COVID pandemic. Herein, we summarized COVID-19 outcomes of CD patients treated with immunosuppressive agents from multiple cohort studies and also investigated possible mechanisms of how SARS-CoV-2 impacts the host immunity with special consideration of CD patients. We first looked into the SARS-CoV-2-related immunopathology, including lymphocytopenia, T-cell exhaustion, cytokine storms, and their possible molecular interactions, and then focused on mechanistic actions of gastrointestinal systems, including interruption of tryptophan absorption, development of dysbiosis, and consequent local and systemic inflammation. Given challenges in managing CD, we summarized up-to-date clinical and molecular evidence to help physicians adjust therapeutic strategies to achieve the best clinical outcomes for CD patients.
Collapse
Affiliation(s)
- Chang-Cyuan Chen
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Medical Education, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yu-An Lin
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kuan-Ting Liu
- Department of General Medicine, Chang Gung Memorial Hospital, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chun-Yao Huang
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chun-Ming Shih
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yuan-Ti Lee
- School of Medicine, Chung Shan Medical University, Taichung City, 40201, Taiwan
- Division of Infectious Diseases, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung City, 40201, Taiwan
| | - Jun-Liang Pan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, 11031, Taiwan.
| | - Ai-Wei Lee
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
42
|
Okabe S, Arai Y, Gotoh A. Vitamin K2 Protects Against SARS-CoV-2 Envelope Protein-Induced Cytotoxicity in Chronic Myeloid Leukemia Cells and Enhances Imatinib Activity. Int J Mol Sci 2024; 25:11800. [PMID: 39519351 PMCID: PMC11546361 DOI: 10.3390/ijms252111800] [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: 09/24/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm characterized by excessive proliferation of myeloid cells. The COVID-19 pandemic has raised concerns regarding the impact of SARS-CoV-2 on patients with malignancies, particularly those with CML. This study aimed to investigate the effects of SARS-CoV-2 proteins on CML cell viability and the protective role of vitamin K2 (VK2) in conjunction with imatinib. Experiments conducted on K562 CML cells demonstrated that the SARS-CoV-2 envelope protein induces cytotoxicity and activates caspase 3/7, which are key markers of apoptosis. VK2 mitigated these cytotoxic effects and decreased cytokine production while inhibiting colony formation. Furthermore, the combination of VK2 with imatinib significantly reduced cellular proliferation, diminished mitochondrial membrane potential, and markedly suppressed colony formation. These findings suggest that VK2 protects CML cells from SARS-CoV-2-induced cytotoxicity and enhances the therapeutic efficacy of imatinib, presenting a potential strategy to improve CML treatment during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Seiichi Okabe
- Department of Hematology, Tokyo Medical University, Tokyo 160-0023, Japan; (Y.A.); (A.G.)
| | | | | |
Collapse
|
43
|
Sun H, Liu K, Yu B, Zhu M, Jia L, Yao W, Chen Z, Hao H, Zhang X, Liu Y, Liu H, Shan C, Huang F, Guan W. Characterization of the Pathogenic Features of Multiple SARS-CoV-2 Pandemic Strains in Different Mouse Models. J Med Virol 2024; 96:e70049. [PMID: 39558699 DOI: 10.1002/jmv.70049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/19/2024] [Accepted: 10/23/2024] [Indexed: 11/20/2024]
Abstract
Elucidating the detailed features of emerging SARS-CoV-2 strains both in vitro and in vivo is indispensable for the development of effective vaccines or drugs against viral infection. We thoroughly characterized the virological and pathogenic features of eight different pandemic SARS-CoV-2 strains, from the WT strain to current circulating sublineage EG.5.1, both in vitro and in vivo. Besides detailed virological features observed in Vero E6 cells, the Omicron variants, from BA.1 to EG.5.1, exhibited enhanced infectious effects to upper respiratory tract in K18 human angiotensin-converting enzyme (ACE2) (K18 hACE2) transgenic mice. Both XBB.1.9.1 and EG.5.1 presented stronger tropism to brain, which could be the main reason for the increased lethal effects on mice. In addition, the pathogenesis comparisons among all these viruses in C57BL/6JGpt mice indicated that Omicron variant BA.1 and two new sublineages XBB.1.9.1 and EG.5.1 possessed dual tropisms to both human and mice, which were further confirmed by subsequent bioinformatic analyses and actual affinity comparison between viral RBDs and mouse or human receptor ACE2. Furthermore, the immunocompromised BKS-db mice were found to be more susceptible to Omicron strains compared to C57BL/6JGpt mice, which revealed that viral infectivity was determined by both its affinity to the host receptor and host immunocompetence. Thus, this study not only contributes to a systematic understanding of the pathogenic features of SARS-CoV-2 in mice, but also provides new insights to combat potential future surges of new SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Huize Sun
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kunpeng Liu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Baocheng Yu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Miao Zhu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lijia Jia
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Weitong Yao
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
| | - Zhen Chen
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Haojie Hao
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Xueyan Zhang
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Yi Liu
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
| | - Haibin Liu
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
| | - Chao Shan
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
| | - Fang Huang
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
| | - Wuxiang Guan
- Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei, China
| |
Collapse
|
44
|
Lima EBDS, Carvalho AFS, Zaidan I, Monteiro AHA, Cardoso C, Lara ES, Carneiro FS, Oliveira LC, Resende F, Santos FRDS, Souza-Costa LP, Chaves IDM, Queiroz-Junior CM, Russo RC, Santos RAS, Tavares LP, Teixeira MM, Costa VV, Sousa LP. Angiotensin-(1-7) decreases inflammation and lung damage caused by betacoronavirus infection in mice. Inflamm Res 2024; 73:2009-2022. [PMID: 39292270 DOI: 10.1007/s00011-024-01948-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/30/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVE Pro-resolving molecules, including the peptide Angiotensin-(1-7) [Ang-(1-7)], have potential adjunctive therapy for infections. Here we evaluate the actions of Ang-(1-7) in betacoronavirus infection in mice. METHODS C57BL/6J mice were infected intranasally with the murine betacoronavirus MHV-3 and K18-hACE2 mice were infected with SARS-CoV-2. Mice were treated with Ang-(1-7) (30 µg/mouse, i.p.) at 24-, 36-, and 48-hours post-infection (hpi) or at 24, 36, 48, 72, and 96 h. For lethality evaluation, one additional dose of Ang-(1-7) was given at 120 hpi. At 3- and 5-days post- infection (dpi) blood cells, inflammatory mediators, viral loads, and lung histopathology were evaluated. RESULTS Ang-(1-7) rescued lymphopenia in MHV-infected mice, and decreased airways leukocyte infiltration and lung damage at 3- and 5-dpi. The levels of pro-inflammatory cytokines and virus titers in lung and plasma were decreased by Ang-(1-7) during MHV infection. Ang-(1-7) improved lung function and increased survival rates in MHV-infected mice. Notably, Ang-(1-7) treatment during SARS-CoV-2 infection restored blood lymphocytes to baseline, decreased weight loss, virus titters and levels of inflammatory cytokines, resulting in improvement of pulmonary damage, clinical scores and lethality rates. CONCLUSION Ang-(1-7) protected mice from lung damage and death during betacoronavirus infections by modulating inflammation, hematological parameters and enhancing viral clearance.
Collapse
Affiliation(s)
- Erick Bryan de Sousa Lima
- Programa de Pós-graduação em Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
- Hospital das Clínicas da Universidade Federal de Minas Gerais/Ebserh, Belo Horizonte, Minas Gerais, Brazil
| | - Antônio Felipe S Carvalho
- Programa de Pós-graduação em Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
- Hospital das Clínicas da Universidade Federal de Minas Gerais/Ebserh, Belo Horizonte, Minas Gerais, Brazil
| | - Isabella Zaidan
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Adelson Héric A Monteiro
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Camila Cardoso
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Edvaldo S Lara
- Programa de Pós-graduação em Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Fernanda S Carneiro
- Programa de Pós-graduação em Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil
| | - Leonardo C Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Filipe Resende
- Programa de Pós-graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Felipe Rocha da Silva Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiz Pedro Souza-Costa
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ian de Meira Chaves
- Programa de Pós-graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Celso M Queiroz-Junior
- Programa de Pós-graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo C Russo
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Robson A S Santos
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, National Institute in Science and Technology in nanobiopharmaceutics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciana P Tavares
- Department of Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Mauro M Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vivian V Costa
- Programa de Pós-graduação em Biologia Celular, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia P Sousa
- Programa de Pós-graduação em Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627 - Pampulha, Belo Horizonte, MG, 31270-901, Brazil.
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
45
|
Chen LM, Li JB, Wu R. Predictors of COVID-19 severity in autoimmune disease patients: A retrospective study during full epidemic decontrol in China. Heart Lung 2024; 68:272-278. [PMID: 39142089 DOI: 10.1016/j.hrtlng.2024.08.009] [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: 04/23/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Early identification of risk factors for adverse COVID-19 progression in patients with autoimmune diseases is crucial for patient management, but data on the Chinese population are scarce. OBJECTIVES The purpose of this study was to identify predictors of severe COVID-19 in patients using blood cell ratios, such as the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), systemic immune-inflammation index (SII), and other inflammatory markers. METHODS A retrospective study of 855 patients (746 females; median age 49 years) with autoimmune diseases and concurrent COVID-19 was conducted from December 2022 to February 2023 at the Rheumatology and Immunology Department of the First Affiliated Hospital of Nanchang University. Disease severity was assessed according to the 8th edition of the National Health Commission of the People's Republic of China's COVID-19 Diagnosis and Treatment Guidelines. The clinical classification criteria group mild and moderate cases as nonsevere cases and severe and critical cases as severe cases. A multivariate logistic regression model was established to evaluate the relationships between COVID-19 severity and demographic characteristics, comorbidities, medication use, and laboratory findings. RESULTS The PLR, NLR, and SII were significantly greater in the severe COVID-19 group than in the nonsevere group (all P < 0.05). In addition to classical independent clinical risk factors, increases in the PLR (OR: 1.004, 95 % CI: 1.001∼1.007, p = 0.001), NLR (OR: 1.180, 95 % CI: 1.041∼1.337, p = 0.010), and SII (OR: 0.999, 95 % CI: 0.998∼1.000, p = 0.005) were identified as risk factors for severe COVID-19 in patients with autoimmune diseases. After adjusting for clinical risk factors, the PLR (AUC: 0.592 vs. 0.865; P < 0.05), NLR (AUC: 0.670 vs. 0.866; P < 0.05), and SII (AUC: 0.616 vs. 0.864; P < 0.05) demonstrated higher predictive values. CONCLUSION Early prediction of severe COVID-19 in patients with autoimmune diseases can be achieved using the NLR, PLR, and SII.
Collapse
Affiliation(s)
- Li-Ming Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17 Yongwaizheng Street, Donghu District, Nanchang City, Jiangxi Province, 330006, China
| | - Jian-Bin Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17 Yongwaizheng Street, Donghu District, Nanchang City, Jiangxi Province, 330006, China
| | - Rui Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No 17 Yongwaizheng Street, Donghu District, Nanchang City, Jiangxi Province, 330006, China.
| |
Collapse
|
46
|
Samet M, Mahdiabadi PR, Tajamolian M, Jelodar MG, Monshizadeh K, Javazm RR, Yazdi M, Abessi P, Hoseini SM. ABO gene polymorphism and COVID-19 severity: The impact on haematological complications, inflammatory markers, and lung lesions. Hum Immunol 2024; 85:111184. [PMID: 39566435 DOI: 10.1016/j.humimm.2024.111184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/01/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
PURPOSE The study aimed to investigate the connection between an intronic variant in the ABO gene (rs657152) and the severity of COVID-19 in terms of clinical symptoms, haematological complications, inflammatory markers, and lung lesions. METHODS After applying exclusion criteria, the study included 240 patients divided into 3 groups: 88 Outpatients, 84 Ward-hospitalized, and 68 ICU-admitted/failed patients. The tetra-ARMS PCR method was used to genotype ABO polymorphism in the patient. Paraclinical tests of patients at the time of admission (before receiving conventional treatments) included levels of C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR), as well as a complete blood count (CBC). Also, the severity of lung lesions was evaluated based on the results of spiral computed tomography (CT) of the chest during admission. RESULTS The statistical analysis using the ANOVA test revealed significant differences in the mean values of allele frequencies (p-value = 0.0020) and genotype proportions (p-value = 0.0017) among clinical groups. The study also found a notable difference in ABO polymorphism across different levels of the inflammatory marker CRP, but not with the ESR levels. Furthermore, the study showed a significant difference in the distribution of lung lesion severity and ABO polymorphism among different clinical groups. CONCLUSION To conclude, our findings supported the substantial impact of ABO polymorphism rs657152 on the severity of COVID-19 in Iranian patients, specifically concerning haematological complications, inflammatory markers, and lung lesions. The study underscored the protective effect of the AC genotype and the detrimental impact of the CC genotype on clinical manifestations.
Collapse
Affiliation(s)
- Mohammad Samet
- Departments of Internal Medicine, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Parvane Raeesi Mahdiabadi
- Departments of Internal Medicine, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoud Tajamolian
- Medical Genetics Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mohsen Gholinataj Jelodar
- Departments of Internal Medicine, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Kimia Monshizadeh
- Medical Genetics Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Reza Rafiei Javazm
- Biotechnology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mehran Yazdi
- Departments of Internal Medicine, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Panteha Abessi
- Biotechnology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mehdi Hoseini
- Hematology and Oncology Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
47
|
Michot JM, Dozio V, Rohmer J, Pommeret F, Roumier M, Yu H, Sklodowki K, Danlos FX, Ouali K, Kishazi E, Naigeon M, Griscelli F, Gachot B, Groh M, Bacciarello G, Stoclin A, Willekens C, Sakkal M, Bayle A, Zitvogel L, Silvin A, Soria JC, Barlesi F, Beeler K, André F, Vasse M, Chaput N, Ackermann F, Escher C, Marabelle A. Circulating Proteins Associated with Anti-IL6 Receptor Therapeutic Resistance in the Sera of Patients with Severe COVID-19. J Proteome Res 2024; 23:5001-5015. [PMID: 39352225 DOI: 10.1021/acs.jproteome.2c00422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Circulating proteomes provide a snapshot of the physiological state of a human organism responding to pathogenic challenges and drug interventions. The outcomes of patients with COVID-19 and acute respiratory distress syndrome triggered by the SARS-CoV2 virus remain uncertain. Tocilizumab is an anti-interleukin-6 treatment that exerts encouraging clinical activity by controlling the cytokine storm and improving respiratory distress in patients with COVID-19. We investigate the biological determinants of therapeutic outcomes after tocilizumab treatment. Overall, 28 patients hospitalized due to severe COVID-19 who were treated with tocilizumab intravenously were included in this study. Sera were collected before and after tocilizumab, and the patient's outcome was evaluated until day 30 post-tocilizumab infusion for favorable therapeutic response to tocilizumab and mortality. Hyperreaction monitoring measurements by liquid chromatography-mass spectrometry-based proteomic analysis with data-independent acquisition quantified 510 proteins and 7019 peptides in the serum of patients. Alterations in the serum proteome reflect COVID-19 outcomes in patients treated with tocilizumab. Our results suggested that circulating proteins associated with the most significant prognostic impact belonged to the complement system, platelet degranulation, acute-phase proteins, and the Fc-epsilon receptor signaling pathway. Among these, upregulation of the complement system by activation of the classical pathway was associated with poor response to tocilizumab, and upregulation of Fc-epsilon receptor signaling was associated with lower mortality.
Collapse
Affiliation(s)
- Jean-Marie Michot
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Vito Dozio
- Biognosys, Wagistrasse 21, Schlieren 8952, Switzerland
| | - Julien Rohmer
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | - Fanny Pommeret
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Mathilde Roumier
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | - Haochen Yu
- Biognosys, Wagistrasse 21, Schlieren 8952, Switzerland
| | | | - François-Xavier Danlos
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Kaissa Ouali
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Edina Kishazi
- Biognosys, Wagistrasse 21, Schlieren 8952, Switzerland
| | - Marie Naigeon
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Laboratoire d'Immunomonitoring en Oncologie, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- Université Paris Saclay, Faculté de Pharmacie, Chatenay-Malabry F-92296, France
| | - Franck Griscelli
- Département de biologie et pathologie, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Bertrand Gachot
- Unité de Pathologie Infectieuse, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Matthieu Groh
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | - Giulia Bacciarello
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Annabelle Stoclin
- Unité de Pathologie Infectieuse, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Christophe Willekens
- Département d'hématologie, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Madona Sakkal
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Arnaud Bayle
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | | | - Aymeric Silvin
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Jean-Charles Soria
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
| | - Fabrice Barlesi
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | | | - Fabrice André
- Département de Médecine, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
- Unité INSERM U981, Gustave Roussy Cancer Campus, Villejuif 94800, France
| | - Marc Vasse
- Université Paris Saclay, Faculté de Pharmacie, Chatenay-Malabry F-92296, France
- Service de biologie clinique, Hôpital Foch, Suresnes 92150, France
- Unité INSERM U1176, Le Kremlin-Bicêtre, Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
| | - Nathalie Chaput
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Laboratoire d'Immunomonitoring en Oncologie, Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
| | - Felix Ackermann
- Service de Médecine Interne, Hôpital Foch, Suresnes 92150, France
| | | | - Aurélien Marabelle
- Département des Innovations Thérapeutiques et des Essais Précoces (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif 94800, France
- INSERM U1015, Gustave Roussy Cancer Campus, Villejuif 94800, France
- Université Paris Saclay, Faculté de Médecine, Le Kremlin-Bicêtre 94270, France
- Centre d'investigation clinique - biothérapie, INSERM CICBT1428, Villejuif 94800, France
| |
Collapse
|
48
|
Aquino A, Zaikova E, Kalinina O, Karonova TL, Rubinstein A, Mikhaylova AA, Kudryavtsev I, Golovkin AS. T Regulatory Cell Subsets Do Not Restore for One Year After Acute COVID-19. Int J Mol Sci 2024; 25:11759. [PMID: 39519310 PMCID: PMC11545974 DOI: 10.3390/ijms252111759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
COVID-19, caused by SARS-CoV-2, triggers a complex immune response, with T regulatory cells (Tregs) playing a crucial role in maintaining immune homeostasis and preventing excessive inflammation. The current study investigates the function of T regulatory cells during COVID-19 infection and the subsequent recovery period, emphasizing their impact on immune regulation and inflammation control. We conducted a comprehensive analysis of Treg subpopulations in peripheral blood samples from COVID-19 patients at different stages: acute infection, early convalescence, and long-term recovery. Flow cytometry was employed to quantify Tregs including "naïve", central memory (CM), effector memory (EM), and terminally differentiated CD45RA+ effector cells (TEMRA). Additionally, the functional state of the Tregs was assessed by the expression of purinergic signaling molecules (CD39, CD73). Cytokine profiles were assessed through multiplex analysis. Our findings indicate a significant decrease in the number of Tregs during the acute phase of COVID-19, which correlates with heightened inflammatory markers and increased disease severity. Specifically, we found a decrease in the relative numbers of "naïve" and an increase in EM Tregs, as well as a decrease in the absolute numbers of "naïve" and CM Tregs. During the early convalescent period, the absolute counts of all Treg populations tended to increase, accompanied by a reduction in pro-inflammatory cytokines. Despite this, one year after recovery, the decreased subpopulations of regulatory T cells had not yet reached the levels observed in healthy donors. Finally, we observed the re-establishment of CD39 expression in all Treg subsets; however, there was no change in CD73 expression among Tregs. Understanding these immunological changes across different T regulatory subsets and adenosine signaling pathways offers important insights into the disease's pathogenesis and provides a broader view of immune system dynamics during recovery.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Alexey S. Golovkin
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia; (A.A.); (A.R.); (I.K.)
| |
Collapse
|
49
|
Li C, He H, Wang Y, Huang L, Chen Z, Zhang Q, Cai Y, Zhai T, Wu X, Zhan Q. Outcomes and inflammation changes in different types of immunocompromised patients with critically ill COVID-19 admitted to ICU: a national multicenter study. BMC Pulm Med 2024; 24:548. [PMID: 39482633 PMCID: PMC11529014 DOI: 10.1186/s12890-024-03362-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Immunocompromised patients face higher risks of Severe Acute Respiratory Syndrome Coronavirus 2 infection and co-infections, leading to a possibility of high disease severity and poor outcomes. Conversely, immunosuppression can mitigate the excessive inflammatory response induced by the virus, potentially reducing disease severity. This study aims to investigate the prognostic differences and early inflammatory response characteristics in various types of immunocompromised patients with severe coronavirus disease 2019 (COVID-19) admitted to intensive care unit (ICU), summarize their clinical features, and explore potential mechanisms. METHODS A retrospective analysis was conducted on critically ill COVID-19 patients admitted to the ICU of 59 medical centers in mainland China during the Omicron outbreak from November 2022 to February 2023. Patients were categorized into two groups based on their immunosuppression status: immunocompromised and immunocompetent. Immunocompromised patients were further subdivided by etiology into cancer patients, solid organ transplant (SOT) patients, and other immunocompromised groups, with immunocompetent patients serving as controls. The mortality rates, respiratory support, complications, and early inflammatory cytokine dynamics upon ICU admission among different populations were analyzed. RESULTS A total of 2030 critically ill COVID-19 patients admitted to ICU were included, with 242 in the immunocompromised group and 1788 in the immunocompetent group. Cancer patients had a higher median age of 69 years (IQR 59, 77), while SOT patients were generally younger and had less severe illness upon ICU admission, with a median APACHE II score of 12.0 (IQR 8.0, 20.0). Cancer patients had a twofold increased risk of death (OR = 2.02, 95% CI 1.18-3.46, P = 0.010) compared to immunocompetent patients. SOT and cancer patients exhibited higher C-reactive protein and serum ferritin levels than the immunocompetent group in their early days of ICU admission. The CD8+ T cells dynamics were inversely correlated in cancer and SOT patients, with Interleukin-6 levels consistently lower in the SOT group compared to both immunocompetent and cancer patients. CONCLUSION Critically ill COVID-19 patients admitted to the ICU exhibit distinct clinical outcomes based on their immunosuppression status, with cancer patients facing the highest mortality rate due to variations in inflammatory responses linked to their immunosuppression mechanisms. Monitoring dynamic changes in inflammatory markers and immune cells, particularly CD8+ T lymphocytes and IL-6, may offer valuable prognostic insights for these patients.
Collapse
Affiliation(s)
- Chunyan Li
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang, China
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, Chinese Academy of Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Hangyong He
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, Chinese Academy of Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Yuqiong Wang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, Chinese Academy of Medical Sciences, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship School of Clinical Medicine, Peking University, Beijing, China
| | - Linna Huang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, Chinese Academy of Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Ziying Chen
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, Chinese Academy of Medical Sciences, China-Japan Friendship Hospital, Beijing, China
- China-Japan Friendship School of Clinical Medicine, Peking University, Beijing, China
| | - Qi Zhang
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, Chinese Academy of Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Ying Cai
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, Chinese Academy of Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Tianshu Zhai
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, Chinese Academy of Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Xiaojing Wu
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, Chinese Academy of Medical Sciences, China-Japan Friendship Hospital, Beijing, China.
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, China.
| | - Qingyuan Zhan
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, Heilongjiang, China.
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, Chinese Academy of Medical Sciences, China-Japan Friendship Hospital, Beijing, China.
- China-Japan Friendship School of Clinical Medicine, Peking University, Beijing, China.
| |
Collapse
|
50
|
Liu Y, Zhang W, Sun M, Liang X, Wang L, Zhao J, Hou Y, Li H, Yang X. The severity assessment and nucleic acid turning-negative-time prediction in COVID-19 patients with COPD using a fused deep learning model. BMC Pulm Med 2024; 24:515. [PMID: 39402509 PMCID: PMC11476205 DOI: 10.1186/s12890-024-03333-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Previous studies have shown that patients with pre-existing chronic obstructive pulmonary diseases (COPD) were more likely to be infected with coronavirus disease (COVID-19) and lead to more severe lung lesions. However, few studies have explored the severity and prognosis of COVID-19 patients with different phenotypes of COPD. PURPOSE The aim of this study is to investigate the value of the deep learning and radiomics features for the severity evaluation and the nucleic acid turning-negative time prediction in COVID-19 patients with COPD including two phenotypes of chronic bronchitis predominant patients and emphysema predominant patients. METHODS A total of 281 patients were retrospectively collected from Hohhot First Hospital between October 2022 and January 2023. They were divided to three groups: COVID-19 group of 95 patients, COVID-19 with emphysema group of 94 patients, COVID-19 with chronic bronchitis group of 92 patients. All patients underwent chest computed tomography (CT) scans and recorded clinical data. The U-net model was pretrained to segment the pulmonary involvement area on CT images and the severity of pneumonia were evaluated by the percentage of pulmonary involvement volume to lung volume. The 107 radiomics features were extracted by pyradiomics package. The Spearman method was employed to analyze the correlation of the data and visualize it through a heatmap. Then we establish a deep learning model (model 1) and a fusion model (model 2) combined deep learning with radiomics features to predict nucleic acid turning-negative time. RESULTS COVID-19 patients with emphysema was lowest in the lymphocyte count compared to COVID-19 patients and COVID-19 companied with chronic bronchitis, and they have the most extensive range of pulmonary inflammation. The lymphocyte count was significantly correlated with pulmonary involvement and the time for nucleic acid turning negative (r=-0.145, P < 0.05). Importantly, our results demonstrated that model 2 achieved an accuracy of 80.9% in predicting nucleic acid turning-negative time. CONCLUSION The pre-existing emphysema phenotype of COPD severely aggravated the pulmonary involvement of COVID-19 patients. Deep learning and radiomics features may provide more information to accurately predict the nucleic acid turning-negative time, which is expected to play an important role in clinical practice.
Collapse
Affiliation(s)
- Yanhui Liu
- Medical Imaging Department, Hohhot First Hospital, Inner Mongolia, P.R. China
| | - Wenxiu Zhang
- Institute of Research and Clinical Innovations, Neusoft Medical Systems Co., Ltd, Shanghai, P.R. China
| | - Mengzhou Sun
- Institute of Research and Clinical Innovations, Neusoft Medical Systems Co., Ltd, Beijing, P.R. China
| | - Xiaoyun Liang
- Institute of Research and Clinical Innovations, Neusoft Medical Systems Co., Ltd, Shanghai, P.R. China
| | - Lu Wang
- Medical Imaging Department, Hohhot First Hospital, Inner Mongolia, P.R. China
| | - Jiaqi Zhao
- Medical Imaging Department, Hohhot First Hospital, Inner Mongolia, P.R. China
| | - Yongquan Hou
- Respiratory and Critical Care Medicine Department, Hohhot First Hospital, Inner Mongolia, P.R. China
| | - Haina Li
- Medical Imaging Department, Hohhot First Hospital, Inner Mongolia, P.R. China
| | - Xiaoguang Yang
- Medical Imaging Department, Hohhot First Hospital, Inner Mongolia, P.R. China.
| |
Collapse
|