1
|
Oba S, Hosoya T, Kawata D, Komiya Y, Iwai H, Koike R, Miyamoto S, Kanno T, Ainai A, Suzuki T, Hasegawa H, Yasuda S. Iguratimod, a promising therapeutic agent for COVID-19 that attenuates excessive inflammation in mouse models. Eur J Pharmacol 2025; 996:177537. [PMID: 40147575 DOI: 10.1016/j.ejphar.2025.177537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
In severe COVID-19 patients, excessive inflammation can lead to multiorgan dysfunction. Current anti-inflammatory treatments like glucocorticoids partially improve the outcomes, while immune systems are compromised. We have identified that SARS-CoV-2-infected obese mice were a good model of the cytokine storm seen in COVID-19. Here, we revealed that iguratimod (IGU), an approved agent for rheumatoid arthritis, improved survival by attenuating inflammation with minimal immune suppression. In this study, C57BL/6 mice were fed a high-fat diet (HFD) or a normal-fat diet (NFD) for ten weeks before being infected with a mouse-adapted SARS-CoV-2. IGU significantly improved survival rates and reduced lung inflammation in HFD-fed mice, with minimal impact on interferon-induced genes and viral load. Meanwhile, dexamethasone (DEX) did not improve survival, while it suppressed various immune reactions with different mechanisms to IGU. Interestingly, IGU-treated mice had fewer SARS-CoV-2 positive cells in the lung, although viral replication was comparable to the control mice. Neither IGU nor DEX inhibited the SARS-CoV-2 infection in Vero-E6 cells, unlike the antiviral agent, remdesivir. Of note, IGU was effective prophylactically and therapeutically in HFD mice, and showed beneficial effects in NFD-fed mice with a lethal dose exposure of SARS-CoV-2. We demonstrated that IGU could be a promising treatment for severe COVID-19, especially in obese patients, by fine-tuning inflammation without compromising antiviral immunity. This study supports the possibility of drug repositioning for IGU COVID-19 beyond autoimmune diseases.
Collapse
Affiliation(s)
- Seiya Oba
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Tadashi Hosoya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan.
| | - Daisuke Kawata
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Yoji Komiya
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Hideyuki Iwai
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan; Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Ryuji Koike
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Sho Miyamoto
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Takayuki Kanno
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Akira Ainai
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 208-0011, Japan
| | - Hideki Hasegawa
- WHO Collaborating Centre for Reference and Research on Influenza, Tokyo, Japan; Research Center for Influenza and Respiratory Virus, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shinsuke Yasuda
- Department of Rheumatology, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Tokyo, Japan.
| |
Collapse
|
2
|
Aguirre-Alvarado C, Cortes-Vázquez MÁ, Pérez-González YS, Meza-Sánchez DE, Nuñez-Enriquez JC, Pinto-Cardoso SM, Bekker-Méndez VC. Elevated IL-6 and IL-10 Levels as Prognostic Biomarkers in COVID-19 Pneumonia: A Comparative Study in Mexican Patients. Healthcare (Basel) 2025; 13:1245. [PMID: 40508859 PMCID: PMC12155521 DOI: 10.3390/healthcare13111245] [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: 03/21/2025] [Revised: 05/15/2025] [Accepted: 05/16/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES Proinflammatory cytokines have been associated with poor prognosis in community-acquired and COVID-19 pneumonia. There is a paucity of reports on the cytokine release syndrome, also called cytokine storm in the Mexican population with pneumonia and COVID-19; therefore, our objective was to compare proinflammatory cytokine levels in Mexican patients without COVID-19 (non-COVID-19) and those with moderate, severe, and critical COVID-19 pneumonia. METHODS This study included 30 patients with non-COVID-19 pneumonia and 57 with COVID-19 pneumonia. Disease diagnosis and severity were determined using the radiographic pulmonary edema assessment (RALE) score. Quantification of IL-6, IL-10, and TNF-α was performed using multiplex immunoassays. A receiver operating characteristic curve was constructed to classify subjects with elevated cytokine levels. Logistic regression was used to find associations between elevated cytokine levels and the presence of COVID-19 pneumonia. RESULTS The severity classification of patients with COVID-19 pneumonia was as follows: moderate (n = 20), severe (n = 19), and critical (n = 18). The proinflammatory cytokines IL-6 and IL-10 were significantly increased in COVID-19 patients compared to non-COVID-19 patients (p < 0.005), while TNF-α levels were lower in critically ill patients with COVID-19 pneumonia. High levels of IL-6 and IL-10, adjusted for age, sex, the presence of comorbidities, and the neutrophil-to-lymphocyte ratio (NLR), showed an elevated risk (OR IL-6 = 4.02; OR IL-10 = 9.36) of presenting pneumonia and COVID-19 compared to pneumonia without COVID-19 in patients. Likewise, 61% of COVID-19 patients with elevated proinflammatory cytokines (IL-6 and IL-10) had a fatal outcome. CONCLUSIONS Elevated levels of both IL-6 and IL-10 are a differential risk factor for developing COVID-19 pneumonia. These elevated levels were more frequently observed in Mexican COVID-19 pneumonia patients who died at the onset of the COVID-19 pandemic. It is important that they are monitored from initial diagnosis as they may be markers of a fatal outcome in severe and critical COVID-19 patients.
Collapse
Affiliation(s)
- Charmina Aguirre-Alvarado
- Laboratorio de Bioquímica Farmacológica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11300, Mexico
- Unidad de Investigación Biomédica en Inmunología e Infectología, Hospital de Infectología “Dr Daniel Méndez Hernández”, Centro Médico Nacional (CMN) “La Raza”, Instituto Mexicano del Seguro Social (IMSS), Mexico City 02990, Mexico
| | - Miguel Ángel Cortes-Vázquez
- Hospital de Infectología “Dr Daniel Méndez Hernández”, CMN “La Raza”, IMSS, Mexico City 02990, Mexico; (M.Á.C.-V.); (Y.S.P.-G.)
| | - Yessica Sara Pérez-González
- Hospital de Infectología “Dr Daniel Méndez Hernández”, CMN “La Raza”, IMSS, Mexico City 02990, Mexico; (M.Á.C.-V.); (Y.S.P.-G.)
| | - David Eduardo Meza-Sánchez
- Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Universidad Nacional Autónoma de México, Mexico City 14080, Mexico;
| | - Juan Carlos Nuñez-Enriquez
- Unidad de Investigación Médica en Epidemiología Clínica, UMAE Hospital de Pediatría, Centro Médico Nacional “Siglo XXI”, IMSS, Mexico City 06729, Mexico;
| | - Sandra María Pinto-Cardoso
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico;
| | - Vilma Carolina Bekker-Méndez
- Unidad de Investigación Biomédica en Inmunología e Infectología, Hospital de Infectología “Dr Daniel Méndez Hernández”, Centro Médico Nacional (CMN) “La Raza”, Instituto Mexicano del Seguro Social (IMSS), Mexico City 02990, Mexico
| |
Collapse
|
3
|
Partouche N, Maumy M, Chamaraux-Tran TN, Bertrand F, Schneider F, Meyer N, Solis M, Fafi-Kremer S, Noll E, Pottecher J. Does the IL-6/KL-6 ratio distinguish different phenotypes in COVID-19 Acute Respiratory Distress Syndrome? An observational study stemmed from prospectively derived clinical, biological, and computed tomographic data. PLoS One 2025; 20:e0321533. [PMID: 40397955 DOI: 10.1371/journal.pone.0321533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 03/07/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND As new SARS-CoV-2 variants emerge and as treatment of COVID-19 ARDS remains exclusively supportive, there is an unmet need to better characterize its different phenotypes to tailor personalized treatments. Clinical, biological, spirometric and CT data hardly allow deciphering of Heavy (H), Intermediate (I) and Light (L) phenotypes of COVID-19 ARDS and the implementation of tailored specific strategies (prone positioning, PEEP settings, recruitment maneuvers). We hypothesized that the ratio of two pivotal COVID-19 biomarkers (interleukin 6 [IL-6] and Krebs von den Lungen 6 [KL-6], related to inflammation and pneumocyte repair, respectively) would provide a biologic insight into the disease timeline allowing 1) to differentiate H, I and L phenotypes, 2) to predict outcome and 3) to reflect some of CT findings. METHODS AND FINDINGS This was a retrospective analysis of prospectively acquired data (COVID HUS cohort). Inclusion concerned any patient with severe COVID-19 pneumonia admitted to two intensive care units between March 1st and May 1st, 2020, in a high-density cluster of the first epidemic wave (Strasbourg University Hospital, France). Demographic, clinical, biological (standard, IL-6 [new generation ELISA], KL-6 [CLEIA technique]), spirometric (driving pressure, respiratory system compliance) and CT data were collected longitudinally. CT analysis included semi-automatic and automatic lung measurements and allowed segmentation of lung volumes into 4 (poorly aerated, non-aerated, overinflated and normally aerated) and 3 (ground-glass, restricted normally aerated, and overinflated) zones, respectively. The primary outcome was to challenge the IL-6/KL-6 ratio capacity to decipher the three COVID-19 ARDS phenotypes (H, I and L) defined on clinical, spirometric and radiologic grounds. Secondary outcomes were the analysis of the prognostic value of the IL-6/KL-6 ratio and its correlates with CT-acquired data. Multivariate analysis was based on principal component analysis. One hundred and forty-eight ventilated COVID-19 ICU patients from the COVID HUS cohort were assessed for eligibility and 77 were included in the full analysis. Most were male, all were under invasive mechanical ventilation and vasopressor therapy and displayed high severity scores (SAPSII: 48 [42-56]; SOFA: 8 [7-10]). The L, I and H COVID ARDS phenotypes were identified in 11, 15 and 48 patients, respectively. In three patients, the phenotype could not be defined precisely. Thirty patients (39%) died in the ICU and the number of ventilator-free days was 2 [0-2] days. The IL-6/KL-6 ratio was not significantly different between the L, I and H phenotypes and evolved according to similar patterns over time. Surviving and deceased patients displayed an inverse kinetic of KL-6. IL-6 and the IL-6/KL-6 ratio were linearly associated with ground-glass volume on semi-automatic and automatic CT lung measurements. CONCLUSIONS In our population of severe ventilated COVID ARDS patients, the IL-6/KL-6 ratio was not clue to differentiate the H, I and L phenotypes and tailor a personalized ventilatory approach. There was an interesting correlation between IL-6/KL-6 ratio and ground-glass volume as determined by automated lung CT analysis. Such correlation deserves more in-depth pathophysiological study, at best gathered from a prospective cohort with a larger sample size and histological analysis. TRIAL REGISTRATION COVID HUS Trial registration number: NCT04405726.
Collapse
Affiliation(s)
- Nicolas Partouche
- Service d'Anesthésie-Réanimation & Médecine Péri-Opératoire, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, UR3072, FMTS, FHU Omicare, Faculté de Médecine, Maïeutique et Sciences de la Santé, Université de Strasbourg, Strasbourg, France
| | - Myriam Maumy
- LIST3N, University of Technology of Troyes, Troyes, France
| | - Thien-Nga Chamaraux-Tran
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 1 Rue Laurent Fries, Illkirch-Graffenstaden, France
| | | | - Francis Schneider
- Service de Médecine Intensive-Réanimation, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Nicolas Meyer
- Service de santé Publique, GMRC, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Morgane Solis
- Faculté de Médecine, Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France - INSERM, UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Samira Fafi-Kremer
- Faculté de Médecine, Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France - INSERM, UMR_S1109, LabEx TRANSPLANTEX, Centre de Recherche d'Immunologie et d'Hématologie, Fédération Hospitalo-Universitaire (FHU) OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
| | - Eric Noll
- Service d'Anesthésie-Réanimation & Médecine Péri-Opératoire, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, UR3072, FMTS, FHU Omicare, Faculté de Médecine, Maïeutique et Sciences de la Santé, Université de Strasbourg, Strasbourg, France
| | - Julien Pottecher
- Service d'Anesthésie-Réanimation & Médecine Péri-Opératoire, Hôpital de Hautepierre, Hôpitaux Universitaires de Strasbourg, UR3072, FMTS, FHU Omicare, Faculté de Médecine, Maïeutique et Sciences de la Santé, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
4
|
Keskitalo S, Seppänen MRJ, Del Sol A, Varjosalo M. From rare to more common: The emerging role of omics in improving understanding and treatment of severe inflammatory and hyperinflammatory conditions. J Allergy Clin Immunol 2025; 155:1435-1450. [PMID: 39978687 DOI: 10.1016/j.jaci.2025.02.011] [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: 10/07/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
Inflammation is a pathogenic driver of many diseases, including atherosclerosis and rheumatoid arthritis. Hyperinflammation can be seen as any inflammatory response that is deleterious to the host, regardless of cause. In medicine, hyperinflammation is defined as severe, deleterious, and fluctuating systemic or local inflammation with presence of a cytokine storm. It has been associated with rare autoinflammatory disorders. However, advances in omics technologies, including genomics, proteomics, and metabolomics, have revealed it to be more common, occurring in sepsis and severe coronavirus disease 2019. With a focus on proteomics, this review highlights the key role of omics in this shift. Through an exploration of research, we present how omics technologies have contributed to improved diagnostics, prognostics, and targeted therapeutics in the field of hyperinflammation. We also discuss the integration of advanced technologies, multiomics approaches, and artificial intelligence in analyzing complex datasets to develop targeted therapies, and we address their potential for revolutionizing the clinical aspects of hyperinflammation. We emphasize personalized medicine approaches for effective treatments and outline challenges, including the need for standardized methodologies, robust bioinformatics tools, and ethical considerations regarding data privacy. This review aims to provide a comprehensive overview of the molecular mechanisms underpinning hyperinflammation and underscores the potential of omics technologies in enabling successful clinical management.
Collapse
Affiliation(s)
- Salla Keskitalo
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Mikko R J Seppänen
- Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland; Translational Immunology Research Program, University of Helsinki, Helsinki, Finland; European Reference Network Rare Immunodeficiency Autoinflammatory and Autoimmune Diseases Network (ERN RITA) Core Center, Helsinki, The Netherlands
| | - Antonio Del Sol
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Computational Biology Group, Basque Research and Technology Alliance (CIC bioGUNE-BRTA), Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Markku Varjosalo
- Institute of Biotechnology, Helsinki Institute of Life Science HiLIFE, University of Helsinki, Helsinki, Finland; Department of Biochemistry and Developmental Biology and Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
5
|
Qin J, Wang G, Han D. Benefits of melatonin on mortality in severe-to-critical COVID-19 patients: A systematic review and meta-analysis of randomized controlled trials. Clinics (Sao Paulo) 2025; 80:100638. [PMID: 40187234 PMCID: PMC12002743 DOI: 10.1016/j.clinsp.2025.100638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 03/05/2025] [Accepted: 03/23/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE This meta-analysis aimed to determine the efficacy of melatonin on mortality in patients with severe-to-critical illness COVID-19. METHODS A systematic search was made of PubMed, Embase, Cochrane Library, and clinicaltrials.gov, without language restrictions. Randomized Controlled Trials (RCTs) on the treatment of severe-to-critical COVID-19 with melatonin, compared with placebo or blank, were reviewed. Studies were pooled to Odds Ratios (ORs), with 95 % Confidence Intervals (95 % CIs). RESULTS Three RCTs (enrolling 451 participants) met the inclusion criteria. Melatonin showed a significant effect on in-hospital mortality (OR = 0.19, 95 % CI 0.05 to 0.74; p = 0.02). CONCLUSIONS Melatonin significantly reduced in-hospital mortality in patients with severe-to-critical COVID-19. Melatonin should be considered for severe-to-critical COVID-19 patients.
Collapse
Affiliation(s)
- Jinlv Qin
- Radioimmunoassay Center, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Guizuo Wang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, China
| | - Dong Han
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, China.
| |
Collapse
|
6
|
Ciurariu E, Balteanu MA, Georgescu M, Drăghici GA, Vlăsceanu SG, Șerb AF, Cioboată R. Left vs. Right Bundle Branch Block in COVID-19 Patients: Distinct Clinical Presentations and Prognostic Implications. J Clin Med 2025; 14:2310. [PMID: 40217761 PMCID: PMC11989766 DOI: 10.3390/jcm14072310] [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: 03/03/2025] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: COVID-19 is associated with multiple systemic effects, including cardiovascular complications. However, its interplay with cardiac conduction abnormalities remains underexplored. We compared the clinical profile of COVID-19 patients with pre-existing left bundle branch block (LBBB) or right bundle branch block (RBBB) at hospital admission. Methods: This study included 100 COVID-19 patients with antecedent BBB (50 LBBB, 50 RBBB). Critical cardiometabolic, renal, hematological, and inflammatory markers were measured. Logistic regression was used to identify key predictors differentiating COVID-19 patients with LBBB and RBBB. Spearman's correlations were applied to assess intra-strata associations for these variables. Results: COVID-19 patients with LBBB patients were significantly more likely to display lower systolic blood pressure (p = 0.012) but greater left atrial size (p = 0.008), left ventricular diameter (p = 0.001), and interventricular septal thickness (p = 0.023). Hematological and inflammatory markers differed, with LBBB patients being prone to exhibit higher red cell distribution width (p = 0.005), lymphocyte count (p < 0.001), neutrophil count (p = 0.045), and C-reactive protein (p < 0.001). This group also tended to show lower erythrocyte sedimentation rate (p = 0.013) and glycated hemoglobin (p = 0.045) but higher random glucose (p = 0.014). Absolute lymphocyte count, C-reactive protein, and left ventricular diameter were the most robust predictors distinguishing LBBB from RBBB. Significant associations were found exclusively for LBBB, all of them being weak. These predominantly negative relationships indicated an inflammatory origin, and most of them occurred for lymphocyte count. Conclusions: COVID-19 patients with LBBB and RBBB present distinct clinical profiles at hospital admission. The former group demonstrates a more adverse baseline clinical profile, particularly in terms of cardiac and inflammatory markers. These findings suggest that pre-existing BBB type may influence disease progression, potentially helping in risk stratification for COVID-19 patients.
Collapse
Affiliation(s)
- Elena Ciurariu
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania; (E.C.); (M.G.)
| | - Mara Amalia Balteanu
- Department of Pulmonology, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania;
| | - Marius Georgescu
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania; (E.C.); (M.G.)
- Center of Immuno-Physiology and Biotechnologies (CIFBIOTEH), “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania
| | - George Andrei Drăghici
- Department of Toxicology, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania
| | - Silviu Gabriel Vlăsceanu
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Alina-Florina Șerb
- Department of Biochemistry and Pharmacology, Biochemistry Discipline, “Victor Babeș” University of Medicine and Pharmacy Timișoara, Eftimie Murgu Square No. 2, 300041 Timișoara, Romania;
| | - Ramona Cioboată
- Pneumology Department, University of Medicine and Pharmacy, 200349 Craiova, Romania;
| |
Collapse
|
7
|
Lee EP, Lin JJ, Chen SH, Chan OW, Su YT, Hsiao MR, Hsia SH, Wu HP. Clinical Value of Tocilizumab in Reducing Mortality in Refractory Septic Shock in Children with Hematologic and Non-Hematologic Diseases. Cells 2025; 14:441. [PMID: 40136690 PMCID: PMC11941062 DOI: 10.3390/cells14060441] [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: 01/14/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Pediatric sepsis remains the main cause of morbidity and mortality among children. Interleukin (IL)-6 is usually produced after infection, and elevated IL-6 levels may cause multisystemic damage. This study aimed to evaluate the effect of tocilizumab, an IL-6 receptor antibody, on children with septic shock. METHODS We conducted a retrospective cohort study of children diagnosed with septic shock and admitted to the pediatric intensive care unit (PICU) between 2018 and 2024. Tocilizumab was administered within 24 h to patients with high IL-6 levels who developed refractory septic shock. Outcomes, including 28-day mortality, morbidity, length of PICU stay, and shock duration, were analyzed between septic children with different etiologies and differed treatments. RESULTS Fifty-four children with refractory septic shock were included. Patients treated with tocilizumab (n = 21) showed improved outcomes compared to those without tocilizumab (n = 33), including shorter PICU stays and lower mortality rates (14.2% vs. 54.5%, p = 0.03). Subgroup analysis revealed that in the non-hematologic group, tocilizumab-treated patients had a 0% mortality rate compared to 50% in untreated patients (p = 0.006). In the hematologic group, tocilizumab-treated patients exhibited a 27.2% mortality rate compared to 61.5% in untreated patients (p = 0.09). Trends in IL-6 levels (D1 to D7) were significantly higher in non-survivors compared to survivors and in patients with hematological malignancies compared to those without. No adverse events, including secondary infections or long-term liver impairment, were observed. CONCLUSIONS Tocilizumab appears to mitigate systemic inflammation and improve outcomes in children with refractory septic shock and elevated IL-6 levels. Further prospective studies are warranted to confirm these findings and establish treatment guidelines.
Collapse
Affiliation(s)
- En-Pei Lee
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Chang Gung Memorial Hospital at Linko, Kweishan, Taoyuan 33305, Taiwan; (E.-P.L.); (J.-J.L.); (O.-W.C.)
- College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan; (S.-H.C.); (Y.-T.S.); (M.-R.H.)
| | - Jainn-Jim Lin
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Chang Gung Memorial Hospital at Linko, Kweishan, Taoyuan 33305, Taiwan; (E.-P.L.); (J.-J.L.); (O.-W.C.)
- College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan; (S.-H.C.); (Y.-T.S.); (M.-R.H.)
| | - Shih-Hsiang Chen
- College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan; (S.-H.C.); (Y.-T.S.); (M.-R.H.)
- Divisions of Hematology and Oncology, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33315, Taiwan
| | - Oi-Wa Chan
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Chang Gung Memorial Hospital at Linko, Kweishan, Taoyuan 33305, Taiwan; (E.-P.L.); (J.-J.L.); (O.-W.C.)
- College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan; (S.-H.C.); (Y.-T.S.); (M.-R.H.)
| | - Ya-Ting Su
- College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan; (S.-H.C.); (Y.-T.S.); (M.-R.H.)
- Division of Pediatric Endocrinology and Genetics, Department of Pediatrics, Chang Gung Memorial Hospital, Taipei 10507, Taiwan
| | - Man-Ru Hsiao
- College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan; (S.-H.C.); (Y.-T.S.); (M.-R.H.)
- Department of Pharmacy, Chang Gung Memorial Hospital, Taoyuan 33315, Taiwan
| | - Shao-Hsuan Hsia
- Physician Assistant Department, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA
| | - Han-Ping Wu
- College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan; (S.-H.C.); (Y.-T.S.); (M.-R.H.)
- Department of Pediatrics, Chiayi Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| |
Collapse
|
8
|
Passos CR, Moreira AA, Reis RF, dos Santos RW, Lobosco M, Rocha BM. A Coupled Model of the Cardiovascular and Immune Systems to Analyze the Effects of COVID-19 Infection. BIOTECH 2025; 14:19. [PMID: 40227362 PMCID: PMC11940274 DOI: 10.3390/biotech14010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 04/15/2025] Open
Abstract
The COVID-19 pandemic has underscored the importance of understanding the interplay between the cardiovascular and immune systems during viral infections. SARS-CoV-2 enters human cells via the ACE-2 enzyme, initiating a cascade of immune responses. This study presents a coupled mathematical model that integrates the cardiovascular system (CVS) and immune system (IS), capturing their complex interactions during infection. The CVS model, based on ordinary differential equations, describes heart dynamics and pulmonary and systemic circulation, while the IS model simulates immune responses to SARS-CoV-2, including immune cell interactions and cytokine production. A coupling strategy transfers information from the IS to the CVS at specific intervals, enabling the exploration of immune-driven cardiovascular effects. Numerical simulations examined how these interactions influence infection severity and recovery. The coupled model accurately replicated the evolution of cardiac function in survivors and non-survivors of COVID-19. Survivors exhibited a left ventricular ejection fraction (LVEF) reduction of up to 25% while remaining within normal limits, whereas non-survivors showed a severe 4-fold decline, indicative of myocardial dysfunction. Similarly, the right ventricular ejection fraction (RV EF) decreased by approximately 50% in survivors but underwent a drastic 5-fold reduction in non-survivors. These findings highlight the model's capacity to distinguish differential cardiac dysfunction across clinical outcomes and its potential to enhance our understanding of COVID-19 pathophysiology.
Collapse
Affiliation(s)
- Camila Ribeiro Passos
- Computational Engineering, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil;
| | | | - Ruy Freitas Reis
- Graduate Program in Computational Modeling, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (R.F.R.); (M.L.)
- Computer Science Department, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Rodrigo Weber dos Santos
- Graduate Program in Computational Modeling, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (R.F.R.); (M.L.)
- Computer Science Department, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Marcelo Lobosco
- Graduate Program in Computational Modeling, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (R.F.R.); (M.L.)
- Computer Science Department, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Bernardo Martins Rocha
- Graduate Program in Computational Modeling, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil; (R.F.R.); (M.L.)
- Computer Science Department, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| |
Collapse
|
9
|
Kingstad-Bakke B, Lee W, Yount BL, Cleven T, Park H, Sullivan JA, Baric RC, Suresh M. Effector CD8 T cell differentiation in primary and breakthrough SARS-CoV-2 infection in mice. Commun Biol 2025; 8:392. [PMID: 40057586 PMCID: PMC11890755 DOI: 10.1038/s42003-025-07820-7] [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: 07/15/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
The nature of the effector and memory T cell response in the lungs following acute SARS-CoV-2 infections remains largely unknown. To define the pulmonary T-cell response to COVID-19, we compared effector and memory T-cell responses to SARS-CoV-2 and influenza A virus (IAV) in mice. Both viruses elicited potent effector T cell responses in lungs, but memory T cells showed exaggerated contraction in SARS-CoV-2-infected mice. Specifically, unlike the T-bet/EOMES-driven effector transcription program in IAV lungs, SARS-CoV-2-specific CD8 T cells embarked on a STAT-3-centric transcriptional program, a defining characteristic of a pro-fibro-inflammatory program: limited cytotoxicity, diminished expression of tissue-protective inhibitory receptors (PD-1, LAG-3, and TIGIT), and augmented mucosal imprinting (CD103). Circulating CD45RO+HLA-DR+ CD8 T cells in hospitalized COVID-19 patients expressed elevated levels of STAT-3 and low levels of TIGIT. IL-6 blockade experiments implicated IL-6 in STAT-3 induction and downregulation of PD-1 expression on SARS-CoV-2-specific primary effector CD8 T cells. Memory CD8 T cells specific to a single epitope, induced by mucosal vaccination, differentiated into cytotoxic effectors and expressed high levels of CD103, effectively reducing viral burden in lungs following a breakthrough SARS-CoV-2 infection. Our findings have implications for developing targeted immunotherapies to mitigate immunopathology and promote protective T cell immunity to SARS-CoV-2.
Collapse
Affiliation(s)
- Brock Kingstad-Bakke
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Woojong Lee
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Boyd L Yount
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Thomas Cleven
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Hongtae Park
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeremy A Sullivan
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Ralph C Baric
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - M Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
10
|
Wen Y, Zhao J, Zhang Z. Heterogeneity and longitudinal transcriptomic characteristics of Tregs in COVID-19 patients. Front Immunol 2025; 16:1548173. [PMID: 40114921 PMCID: PMC11922936 DOI: 10.3389/fimmu.2025.1548173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Introduction Regulatory T cells (Tregs) play a crucial role in maintaining immune tolerance by suppressing immune responses against pathogens. The fluctuation of Treg proportions in COVID-19 remains a topic of debate, and the mechanisms triggering Treg activation in COVID-19 are still unclear. Understanding these issues is essential for better managing immune responses in COVID-19 patients. Methods We collected a cohort of COVID-19 patients with varying disease severity and stage to explore the transcriptomic and functional traits of Tregs in these individuals. Using transcriptomic analysis, we evaluated the proportion and functionality of different Treg subsets, specifically HLA_DR+ Tregs, across different stages of COVID-19 patients. Results Our analysis revealed that the proportion of CCR7 + Tregs decreased as the disease advanced, while the cell proportion of HLA_DR+ regs escalated with the severity of the disease. Moreover, the transcription actor CARHSP1 exhibited apositive correlation with the proportion of HLA_DR+ Tregs. Notably, the heightened suppressive function of HLA_DR+ Tregs in severe COVID-19 patients, with interactions between PF4 and CXCR3, contributed to the homeostasis of HLA_DR+ Tregs in severe COVID-19 patients. Furthermore, we observed that Tregs in COVID-19 patients exhibited weakened TCR clonotype expansion, and the suppression of HLA_DR+ Tregs with expanded TCR clonotypes in severe COVID-19 cases did not show a significant increase compared to asymptomatic and mild COVID-19 groups. The findings indicate that Tregs may be activated through the bystander effect, as evidenced by the analysis of TCR clonotype characteristics. Discussion Our research delineates the diversity of dynamic alterations in Tregs and sheds light on potential mechanisms underlying Treg activation, providing a theoretical foundation and offering treatment strategies for managing COVID-19 patients.
Collapse
Affiliation(s)
| | - Juanjuan Zhao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
11
|
Abiri E, Mirzaii M, Moghbeli M, Atashi A, Harati AA. Investigating DNA damage caused by COVID-19 and influenza in post COVID-19. Mamm Genome 2025; 36:200-212. [PMID: 39537997 DOI: 10.1007/s00335-024-10082-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
The SARS-CoV-2 virus (termed COVID-19) was responsible for over 34 million global deaths. Although the COVID-19 pandemic has subsided, infection by emerging mutant variants of SARS-CoV-2 poses a continuing threat to public health. COVID-19 infection has been associated with the development of cytokine storm syndrome, hypercoagulability, immunological dysregulation and direct viral invasion of organs, and the long-term consequences for the health of COVID-19 survivors are currently unknown. Our research focuses on the possible mutagenic aspects of infection by COVID-19 and measures their harmful effects on DNA composition. DNA damage was investigated, using the comet assay method, during two periods: in the epidemic peak of COVID-19 and during the post-COVID-19 period, both in patients infected with COVID-19 and in those with influenza. During the epidemic peak, the levels of DNA damage ranged from the highest to the lowest levels in the following groups, respectively: intubated-ICU, non-intubated-ICU, non-ICU, and influenza, with a discernible increase in DNA damage in ICU-treated patients. The levels of DNA damage in the post-COVID-19 period were significantly lower compared to those in the epidemic peak period but there was still a discernible increase in DNA damage in the ICU group. Our results indicate that levels of DNA damage may be an effective indicator in prognostic decision-making and may therefore help to reduce mortality. Given that DNA damage and impaired repair processes can contribute to chronic diseases like diabetes, cancer, and neurodegenerative conditions, it will be crucial to investigate potential similar effects in patients with COVID-19.
Collapse
Affiliation(s)
- Elaheh Abiri
- Department of Biology, Islamic Azad University Damghan, Damghan Branch, Damghan, Iran
| | - Mehdi Mirzaii
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
| | - Majid Moghbeli
- Department of Biology, Islamic Azad University Damghan, Damghan Branch, Damghan, Iran
| | - Amir Atashi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Ahad Ali Harati
- Department of Biology, Islamic Azad University Damghan, Damghan Branch, Damghan, Iran
| |
Collapse
|
12
|
Russkikh IV, Popov OS, Klochkova TG, Sushentseva NN, Apalko SV, Asinovskaya AY, Mosenko SV, Sarana AM, Shcherbak SG. Comparative metabolomic analysis reveals shared and unique features of COVID-19 cytokine storm and surgical sepsis. Sci Rep 2025; 15:6622. [PMID: 39994234 PMCID: PMC11850835 DOI: 10.1038/s41598-025-90426-0] [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: 10/26/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
The clinical manifestations of the cytokine storm (CS) associated with COVID-19 resemble the acute phase of sepsis. Metabolomics may contribute to understanding the specific pathobiology of these two syndromes. The aim of this study was to compare serum metabolomic profiles in CS associated with COVID-19 vs. septic surgery patients. In a retrospective cross-sectional study, serum samples from patients with CS associated with COVID-19, with and without comorbidity, as well as serum samples from patients with surgical sepsis were investigated. Targeted metabolomic analysis was performed on all samples using LC-MS/MS. Analysis revealed that similar alterations in the serum metabolome of patients with COVID-19 and surgical septic patients were associated with amino acid metabolism, nitrogen metabolism, inflammatory status, methionine cycle and glycolysis. The most significant difference was found for serum levels of metabolites of kynurenine synthesis, tricarboxylic acid cycle, gamma-aminobutyric acid and niacinamide. The metabolic pathway of cysteine and methionine metabolism was significantly disturbed in COVID-19 and surgical septic patients. For the first time, the similarities and differences between the serum metabolomic profiles of patients with CS associated with COVID-19 and patients with surgical sepsis were investigated for patients from the Northwest of the Russian Federation.
Collapse
Affiliation(s)
- Iana V Russkikh
- Saint Petersburg State Health Care Establishment the City Hospital No. 40 of Health Department of the Saint Petersburg Kurortniy District Administration, St. Borisova, 9, 197706, Sestroretsk, Russian Federation
| | - Oleg S Popov
- Saint Petersburg State Health Care Establishment the City Hospital No. 40 of Health Department of the Saint Petersburg Kurortniy District Administration, St. Borisova, 9, 197706, Sestroretsk, Russian Federation
- Saint Petersburg State University, Government of the Russian Federation, 199034, Saint-Petersburg, Russian Federation
| | - Tatiana G Klochkova
- Saint Petersburg State Health Care Establishment the City Hospital No. 40 of Health Department of the Saint Petersburg Kurortniy District Administration, St. Borisova, 9, 197706, Sestroretsk, Russian Federation.
| | - Natalia N Sushentseva
- Saint Petersburg State Health Care Establishment the City Hospital No. 40 of Health Department of the Saint Petersburg Kurortniy District Administration, St. Borisova, 9, 197706, Sestroretsk, Russian Federation
| | - Svetlana V Apalko
- Saint Petersburg State Health Care Establishment the City Hospital No. 40 of Health Department of the Saint Petersburg Kurortniy District Administration, St. Borisova, 9, 197706, Sestroretsk, Russian Federation
- Saint Petersburg State University, Government of the Russian Federation, 199034, Saint-Petersburg, Russian Federation
| | - Anna Yu Asinovskaya
- Saint Petersburg State Health Care Establishment the City Hospital No. 40 of Health Department of the Saint Petersburg Kurortniy District Administration, St. Borisova, 9, 197706, Sestroretsk, Russian Federation
- Saint Petersburg State University, Government of the Russian Federation, 199034, Saint-Petersburg, Russian Federation
| | - Sergey V Mosenko
- Saint Petersburg State Health Care Establishment the City Hospital No. 40 of Health Department of the Saint Petersburg Kurortniy District Administration, St. Borisova, 9, 197706, Sestroretsk, Russian Federation
- Saint Petersburg State University, Government of the Russian Federation, 199034, Saint-Petersburg, Russian Federation
| | - Andrey M Sarana
- Saint Petersburg State University, Government of the Russian Federation, 199034, Saint-Petersburg, Russian Federation
| | - Sergey G Shcherbak
- Saint Petersburg State Health Care Establishment the City Hospital No. 40 of Health Department of the Saint Petersburg Kurortniy District Administration, St. Borisova, 9, 197706, Sestroretsk, Russian Federation
- Saint Petersburg State University, Government of the Russian Federation, 199034, Saint-Petersburg, Russian Federation
| |
Collapse
|
13
|
Lee C, Khan R, Mantsounga CS, Sharma S, Pierce J, Amelotte E, Butler CA, Farinha A, Parry C, Caballero O, Morrison JA, Uppuluri S, Whyte JJ, Kennedy JL, Zhang X, Choudhary G, Olson RM, Morrison AR. IL-1β-driven NF-κB transcription of ACE2 as a Mechanism of Macrophage Infection by SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.24.630260. [PMID: 39763770 PMCID: PMC11703209 DOI: 10.1101/2024.12.24.630260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Coronavirus disease 2019 (COVID-19), caused by infection with the enveloped RNA betacoronavirus, SARS-CoV-2, led to a global pandemic involving over 7 million deaths. Macrophage inflammatory responses impact COVID-19 severity; however, it is unclear whether macrophages are infected by SARS-CoV-2. We sought to identify mechanisms regulating macrophage expression of ACE2, the primary receptor for SARS-CoV-2, and to determine if macrophages are susceptible to productive infection. We developed a humanized ACE2 (hACE2) mouse whereby hACE2 cDNA was cloned into the mouse ACE2 locus under control of the native promoter. We validated the susceptibility of hACE2 mice to SARS-CoV-2 infection relative to wild-type mice and an established K18-hACE2 model of acute fulminating disease. Intranasal exposure to SARS-CoV-2 led to pulmonary consolidations with cellular infiltrate, edema, and hemorrhage, consistent with pneumonia, yet unlike the K18-hACE2 model, hACE2 mice survived and maintained stable weight. Infected hACE2 mice also exhibited a unique plasma chemokine, cytokine, and growth factor inflammatory signature relative to K18-hACE2 mice. Infected hACE2 mice demonstrated evidence of viral replication in infiltrating lung macrophages, and infection of macrophages in vitro revealed a transcriptional profile indicative of altered RNA and ribosomal processing machinery as well as activated cellular antiviral defense. Macrophage IL-1β-driven NF-κB transcription of ACE2 was an important mechanism of dynamic ACE2 upregulation, promoting macrophage susceptibility to infection. Experimental models of COVID-19 that make use of native hACE2 expression will allow for mechanistic insight into factors that can either promote host resilience or increase susceptibility to worsening severity of infection.
Collapse
Affiliation(s)
- Cadence Lee
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Rachel Khan
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Chris S. Mantsounga
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Sheila Sharma
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Julia Pierce
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Elizabeth Amelotte
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Celia A. Butler
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Andrew Farinha
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Crystal Parry
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Olivya Caballero
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Jeremi A. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Saketh Uppuluri
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Jeffrey J. Whyte
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri Division of Research, Innovation and Impact, Columbia, Missouri, USA
| | - Joshua L. Kennedy
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Arkansas Children’s Research Institute, Little Rock, Arkansas, USA
| | - Xuming Zhang
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
- Cardiovascular Research Center, Lifespan Cardiovascular Research Institute, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Rachel M. Olson
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri Division of Research, Innovation and Impact, Columbia, Missouri, USA
| | - Alan R. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
- Lead contact and corresponding author
| |
Collapse
|
14
|
Niimi N, Pankiv E, Adam RI, Hayes J, Maynes JT, Aoyama K. Roles of evidence synthesis studies and evidence-based clinical practice guidelines in pediatric perioperative outcomes research. J Anesth 2025; 39:1-4. [PMID: 39692878 DOI: 10.1007/s00540-024-03437-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 11/20/2024] [Indexed: 12/19/2024]
Affiliation(s)
- Naoko Niimi
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, 555 University Ave, #2211, Toronto, ON, M5G 1X8, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada
| | - Evelina Pankiv
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, 555 University Ave, #2211, Toronto, ON, M5G 1X8, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada
| | - Ruxandra-Ioana Adam
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, 555 University Ave, #2211, Toronto, ON, M5G 1X8, Canada
- Department of Psychology, Queen's University, Kingston, ON, Canada
| | - Jason Hayes
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, 555 University Ave, #2211, Toronto, ON, M5G 1X8, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada
| | - Jason T Maynes
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, 555 University Ave, #2211, Toronto, ON, M5G 1X8, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada
- Program in Molecular Medicine, SickKids Research Institute, Toronto, ON, Canada
| | - Kazuyoshi Aoyama
- Department of Anesthesia and Pain Medicine, The Hospital for Sick Children, 555 University Ave, #2211, Toronto, ON, M5G 1X8, Canada.
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, ON, Canada.
- Program in Child Health Evaluative Sciences, SickKids Research Institute, Toronto, ON, Canada.
| |
Collapse
|
15
|
Gouveia G, Saateh A, Swietlikowska A, Scarpellini C, Tsang E, Altug H, Merkx M, Dillen A, Leirs K, Spasic D, Lammertyn J, Gothelf KV, Bonedeau E, Porzberg N, Smeets RL, Koenen HJPM, Prins MWJ, de Jonge MI. Continuous Biosensing to Monitor Acute Systemic Inflammation, a Diagnostic Need for Therapeutic Guidance. ACS Sens 2025; 10:4-14. [PMID: 39692622 PMCID: PMC11773571 DOI: 10.1021/acssensors.4c02569] [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] [Revised: 11/14/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024]
Abstract
Continuous monitoring of acute inflammation can become a very important next step for guiding therapeutic interventions in severely ill patients. This Perspective discusses the current medical need for patients with acute inflammatory diseases and the potential of continuous biosensing technologies. First, we discuss biomarkers that could help to monitor the state of a patient with acute systemic inflammation based on theoretical studies and empirical data. Then, based on the state of the art, we describe sensing strategies that could be applied for the continuous monitoring of acute inflammatory biomarkers, followed by challenges that must be overcome. Nanoswitch-based continuous biosensors enable suitable measurement frequencies but still lack sensitivity, while regeneration risks lower sensor reliability. Developments are still needed in bioreceptors and molecular architectures, regeneration techniques, combined with suitable sampling and sample pretreatment methods, for bringing continuous biosensing of inflammation closer to reality. Furthermore, collaborations between healthcare professionals and scientists, regulatory bodies, and biosensor engineers are needed for a successful translation of sensing technologies from the laboratory to clinical practice.
Collapse
Affiliation(s)
- Guilherme Gouveia
- Department
of Laboratory Medicine, Laboratory of Medical Immunology, Radboud
Community for Infectious Diseases, Radboud
University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Abtin Saateh
- Institute
of Bioengineering, École Polytechnique
Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Anna Swietlikowska
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven 5600MB, The Netherlands
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, Eindhoven 5600MB, The Netherlands
| | - Claudia Scarpellini
- Department
of Biosystems - Biosensors Group, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Emily Tsang
- Department
of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus 8000 C, Denmark
| | - Hatice Altug
- Institute
of Bioengineering, École Polytechnique
Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Maarten Merkx
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven 5600MB, The Netherlands
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, Eindhoven 5600MB, The Netherlands
| | - Annelies Dillen
- Department
of Biosystems - Biosensors Group, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Karen Leirs
- Department
of Biosystems - Biosensors Group, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Dragana Spasic
- Department
of Biosystems - Biosensors Group, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Jeroen Lammertyn
- Department
of Biosystems - Biosensors Group, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Kurt V. Gothelf
- Department
of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus 8000 C, Denmark
| | - Estelle Bonedeau
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Nicola Porzberg
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Ruben L. Smeets
- Department
of Laboratory Medicine, Laboratory of Medical Immunology, Radboud
Community for Infectious Diseases, Radboud
University Medical Center, Nijmegen 6500 HB, The Netherlands
- Department
of Laboratory Medicine, Radboudumc Laboratory for Diagnostics, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Hans J. P. M. Koenen
- Department
of Laboratory Medicine, Laboratory of Medical Immunology, Radboud
Community for Infectious Diseases, Radboud
University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Menno W. J. Prins
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, Eindhoven 5600MB, The Netherlands
- Department
of Biomedical Engineering, Eindhoven University
of Technology, Eindhoven 5600MB, The Netherlands
- Department
of Applied Physics, Eindhoven University
of Technology, Eindhoven 5600MB, The Netherlands
- Helia Biomonitoring, De Lismortel 31, 5612 AR Eindhoven, The Netherlands
| | - Marien I. de Jonge
- Department
of Laboratory Medicine, Laboratory of Medical Immunology, Radboud
Community for Infectious Diseases, Radboud
University Medical Center, Nijmegen 6500 HB, The Netherlands
| |
Collapse
|
16
|
Margiotta RG, Sozio E, Del Ben F, Beltrami AP, Cesselli D, Comar M, Devito A, Fabris M, Curcio F, Tascini C, Sanguinetti G. Investigating the relationship between the immune response and the severity of COVID-19: a large-cohort retrospective study. Front Immunol 2025; 15:1452638. [PMID: 39845955 PMCID: PMC11750771 DOI: 10.3389/fimmu.2024.1452638] [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: 06/21/2024] [Accepted: 11/11/2024] [Indexed: 01/24/2025] Open
Abstract
The COVID-19 pandemic has left an indelible mark globally, presenting numerous challenges to public health. This crisis, while disruptive and impactful, has provided a unique opportunity to gather precious clinical data extensively. In this observational, case-control study, we utilized data collected at the Azienda Sanitaria Universitaria Friuli Centrale, Italy, to comprehensively characterize the immuno-inflammatory features in COVID-19 patients. Specifically, we employed multicolor flow cytometry, cytokine assays, and inflammatory biomarkers to elucidate the interplay between the infectious agent and the host's immune status. We characterized immuno-inflammatory profiles within the first 72 hours of hospital admission, stratified by age, disease severity, and time elapsed since symptom onset. Our findings indicate that patients admitted to the hospital shortly after symptom onset exhibit a distinct pattern compared to those who arrive later, characterized by a more active immune response and heightened cytokine activity, but lower markers of tissue damage. We used univariate and multivariate logistic regression models to identify informative markers for outcome severity. Predictors incorporating the immuno-inflammatory features significantly outperformed standard baselines, identifying up to 59% of patients with positive outcomes while maintaining a false omission rate as low as 4%. Overall, our study sheds light on the immuno-inflammatory aspects observed in COVID-19 patients prior to vaccination, providing insights for guiding the clinical management of first-time infections by a novel virus.
Collapse
Affiliation(s)
| | - Emanuela Sozio
- Infectious Disease Unit, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
| | - Fabio Del Ben
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Department of Laboratory Medicine, ASU FC, Udine, Italy
| | - Antonio Paolo Beltrami
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Department of Laboratory Medicine, ASU FC, Udine, Italy
| | - Daniela Cesselli
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Department of Laboratory Medicine, ASU FC, Udine, Italy
| | - Marco Comar
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | | | - Martina Fabris
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Department of Laboratory Medicine, ASU FC, Udine, Italy
| | - Francesco Curcio
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Department of Laboratory Medicine, ASU FC, Udine, Italy
| | - Carlo Tascini
- Infectious Disease Unit, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Guido Sanguinetti
- Physics Department, International School for Advanced Studies (SISSA), Trieste, Italy
| |
Collapse
|
17
|
Zhao YS, Shi YK, Li KF, Ma B, Lin SH, Xing Y, Xu F. Dexmedetomidine Regulates Macrophage Phenotype Remodeling Through AMPK/SIRT1 to Alleviate Inflammatory Mediators and Lung Injury. J Biochem Mol Toxicol 2025; 39:e70108. [PMID: 39692360 DOI: 10.1002/jbt.70108] [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: 06/01/2024] [Revised: 11/22/2024] [Accepted: 12/07/2024] [Indexed: 12/19/2024]
Abstract
Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is associated with high morbidity and mortality in the intensive care unit (ICU) and can cause excessive inflammation. Dexmedetomidine (DEX) is a drug that exerts anti-inflammatory effects. Identifying the anti-inflammatory mechanism of DEX in the context of ALI/ARDS possesses potential significance for the prevention and treatment of ARDS. In this study, DEX was used to treat mouse models of cecal ligation and puncture (CLP) and lipopolysaccharide (LPS)-stimulated cells. Immunofluorescence, western blot analysis, and flow cytometry were used to detect macrophage phenotypic markers in mice, and western blot analysis, real-time qPCR (RT-qPCR), ELISA, and immunofluorescence were used to detect macrophage phenotype markers in RAW264.7 cells. Flow cytometry was used to detect phenotypic markers of bone marrow-derived macrophages (BMDM). Culture medium collected from macrophages was used to cultivate human non-small cell adenocarcinoma epithelial cells (A549) to detect their aquaporins 1 (AQP1) expression and apoptosis status. Western blot analysis was used to detect the activation of the AMP-activated protein kinase (AMPK)/sirtuin 1(SIRT1) signaling pathway both in vivo and in vitro. The regulatory effect of DEX on macrophage phenotype remodeling was detected by knocking down AMPK expression in cells using AMPK shRNA. The results showed that in both in vivo and in vitro experiments, DEX downregulated the expression of M1 markers (tumor necrosis factor-α [TNF-α], nitric oxide synthase [iNOS], and cluster of differentiation [CD]-86) and upregulated the expression of M2 markers (arginase-1 [ARG-1], interleukin [IL]-10, and CD206) in macrophages. The culture medium of macrophages treated with DEX alleviated the edema and apoptosis of A549 cells. DEX activates the AMPK/SIRT1 signaling pathway in macrophages. After AMPK knockdown, the ability of DEX to regulate macrophage phenotype remodeling decreased. Together, this study suggests that DEX regulates macrophage phenotype remodeling by activating the AMPK/SIRT1 pathway, thereby reducing ALI/ARDS.
Collapse
Affiliation(s)
- Yi-Si Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ya-Kang Shi
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Forensic Medicine and Biomedical Informatics Research Room, Chongqing Medical University, Chongqing, China
| | - Ke-Feng Li
- Department of Critical Care Medicine, Fengjie Hospital, A Branch of The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bei Ma
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Critical Care Medicine, The First People's Hospital of Chongqing Liang Jiang New Area, Chongqing, China
| | - Shi-Hui Lin
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Xing
- Forensic Medicine and Biomedical Informatics Research Room, Chongqing Medical University, Chongqing, China
| | - Fang Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
18
|
Behzadi P, Chandran D, Chakraborty C, Bhattacharya M, Saikumar G, Dhama K, Chakraborty A, Mukherjee S, Sarshar M. The dual role of toll-like receptors in COVID-19: Balancing protective immunity and immunopathogenesis. Int J Biol Macromol 2025; 284:137836. [PMID: 39613064 DOI: 10.1016/j.ijbiomac.2024.137836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 11/01/2024] [Accepted: 11/17/2024] [Indexed: 12/01/2024]
Abstract
Toll-like receptors (TLRs) of human are considered as the most critical immunological mediators of inflammatory pathogenesis of COVID-19. These immunoregulatory glycoproteins are located on the surface and/or intracellular compartment act as innate immune sensors. Upon binding with distinct SARS-CoV-2 ligand(s), TLRs signal activation of different transcription factors that induce expression of the proinflammatory mediators that collectively induce 'cytokine storm'. Similarly, TLR activation is also pivotal in conferring protection to infection and invasion as well as upregulating the tissue repair pathways. This dual role of the human TLRs in deciding the fate of SARS-CoV-2 has made these receptor proteins as the critical mediators of immunoprotective and immunopathogenic consequences associated with COVID-19. Herein, pathbreaking discoveries exploring the immunobiological importance of the TLRs in COVID-19 and developing TLR-directed therapeutic intervention have been reviewed by accessing the up-to-date literatures available in the public domain/databases. In accordance with our knowledge in association with the importance of TLRs' role against viruses and identification of viral particles, they have been recognized as suitable candidates with high potential as vaccine adjuvants. In this regard, the agonists of TLR4 and TLR9 have effective potential in vaccine technology while the others need further investigations. This comprehensive review suggests that basal level expression of TLRs can act as friends to keep our body safe from strangers but act as a foe via overexpression. Therefore, selective inhibition of the overexpressed TLRs appears to be a solution to counteract the cytokine storm while TLR-agonists as vaccine adjuvants could lessen the risk of infection in the naïve population.
Collapse
Affiliation(s)
- Payam Behzadi
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran, 37541-374, Iran.
| | | | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, 700126, West Bengal, India
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, VyasaVihar, Balasore, 756020, Odisha, India
| | - Guttula Saikumar
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh, 243122, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, Izatnagar, Uttar Pradesh, 243122, India.
| | - Ankita Chakraborty
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, 713340, West Bengal, India
| | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, 713340, West Bengal, India.
| | - Meysam Sarshar
- Research Laboratories, Bambino Gesù Children's Hospital-IRCCS, 00146, Rome, Italy
| |
Collapse
|
19
|
Kudliński B, Zawadzki J, Kulińska W, Kania J, Murkos M, Stolińska M, Zgoła D, Noga A, Nowak P. Tocilizumab in COVID-19: A Double-Edged Sword? Biomedicines 2024; 12:2924. [PMID: 39767830 PMCID: PMC11672960 DOI: 10.3390/biomedicines12122924] [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: 11/05/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: SARS-CoV-2 was responsible for the global pandemic. Approximately 10-15% of patients with COVID-19 developed respiratory failure with adult acute respiratory distress syndrome (ARDS), which required treatment in the Intensive Care Unit (ICU). The cytokine storm observed in severe COVID-19 was frequently handled with steroids. Synergically, tocilizumab, an anti-interleukin-6 receptor monoclonal antibody, gained popularity as a cytokine storm-suppressing agent. However, immunosuppression was proven to increase the predisposition to infections with resistant bacteria. Our study aimed to assess the relationship between positive tests for secondary infections and the survival of patients with severe COVID-19-attributed ARDS treated with immunosuppressive agents. Methods: This study included 342 patients qualified for the ICU and mechanical ventilation (MV). The patients were divided based on the type of immunomodulating therapy and the culture tests results. Results: The results showed the highest survival rate among patients <61 years, favoring the combined treatment (tocilizumab + steroids). Atrial fibrillation (AF) and coronary heart disease (CHD) correlated with a lower survival rate than other comorbidities. Tocilizumab was associated with an increased risk of positive pathogen cultures, which could potentially cause secondary infections; however, the survival rate among these patients was higher. Conclusions: MV and ICU procedures as well as the application of tocilizumab significantly decreased the mortality rate in patients with severe COVID-19-related ARDS. The suppression of cytokine storms played a crucial role in survival. Tocilizumab was found to be both efficient and safe despite the 'side effect' of the increased risk of positive results for secondary infections.
Collapse
Affiliation(s)
- Bartosz Kudliński
- Department of Anesthesia, Critical Care and Rescue Medicine, Collegium Medicum, University in Zielona Góra, 65-046 Zielona Góra, Poland; (J.Z.); (W.K.); (J.K.); (M.M.); (M.S.); (D.Z.); (A.N.); (P.N.)
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Gutowski M, Klimkiewicz J, Rustecki B, Michałowski A, Skalec T, Lubas A. Peripheral and Organ Perfusion's Role in Prognosis of Disease Severity and Mortality in Severe COVID-19 Patients: Prospective Cohort Study. J Clin Med 2024; 13:7520. [PMID: 39768443 PMCID: PMC11728247 DOI: 10.3390/jcm13247520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/01/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025] Open
Abstract
Severe COVID-19 is associated with a generalized inflammatory response leading to peripheral and organ perfusion disorders. Objectives: This study aimed to evaluate the usefulness of peripheral and organ perfusion assessments in the prediction of prognosis and mortality in patients with severe COVID-19. Patients and Methods: In the first 48 h of hospitalization, peripheral perfusion (saturation, Finger Infrared Thermography-FIT; Capillary Refill Time-CRT), and the color Doppler renal cortex perfusion (RCP) were estimated in a group of 102 severe COVID-19 patients. Results: In total, 40 patients experienced deterioration and required intensification of oxygen treatment, and 24 finally died. In comparison with a stable course of the disease, patients with deterioration had initially higher WBC, CRP, AST, LDH, and CRT, but a lower oxygenation ratio and RCP. Deceased patients were older, had higher CRP, LDH, and CRT, but lower hemoglobin, oxygenation ratio, and RCP compared to survivors. In the multivariable regression analysis from perfusion parameters, only RCP and CRT were independently linked with deterioration (OR 0.002, p < 0.001; OR 1.825, p = 0.003, respectively) and death (OR 0.001, p = 0.004; OR 1.910, p = 0.003, respectively). Conclusions: Initial assessment of peripheral and organ perfusion can be helpful in identifying hospitalized severe COVID-19 patients with a higher risk of deterioration and death. Capillary Refill Time and Renal Cortical Perfusion were prognostic markers of deterioration or death. On the other hand, Finger Infrared Thermography and saturation were not statistically significant in predicting patient outcome. An RCP cut-off value below 0.127 and 0.112 [cm/s] and a Capillary Refill Time longer than 3.25 and 4.25 [s] indicate the risk of deterioration or death, respectively.
Collapse
Affiliation(s)
- Mateusz Gutowski
- Department of Anesthesiology and Intensive Care, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; (J.K.); (B.R.); (A.M.); (T.S.)
| | - Jakub Klimkiewicz
- Department of Anesthesiology and Intensive Care, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; (J.K.); (B.R.); (A.M.); (T.S.)
| | - Bartosz Rustecki
- Department of Anesthesiology and Intensive Care, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; (J.K.); (B.R.); (A.M.); (T.S.)
| | - Andrzej Michałowski
- Department of Anesthesiology and Intensive Care, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; (J.K.); (B.R.); (A.M.); (T.S.)
| | - Tomasz Skalec
- Department of Anesthesiology and Intensive Care, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland; (J.K.); (B.R.); (A.M.); (T.S.)
| | - Arkadiusz Lubas
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine-National Research Institute, 04-141 Warsaw, Poland
| |
Collapse
|
21
|
Nguyen CV, Luong CQ, Dao CX, Nguyen MH, Pham DT, Khuat NH, Pham QT, Hoang DT, Nguyen AD, Nguyen PM, Cao DD, Pham DT, Nguyen TQ, Nong VM, Dang DT, Nguyen DT, Nguyen VD, Le TQ, Nguyen VK, Ngo HD, Nguyen DV, Pham TT, Nguyen DT, Nguyen NT, Do TD, Huynh NT, Phan NT, Nguyen CD, Vo KH, Vu TT, Do CD, Dang TQ, Vu GV, Nguyen TC, Do SN. Predictive validity of interleukin 6 (IL-6) for the mortality in critically ill COVID-19 patients with the B.1.617.2 (Delta) variant in Vietnam: a single-centre, cross-sectional study. BMJ Open 2024; 14:e085971. [PMID: 39653572 PMCID: PMC11628983 DOI: 10.1136/bmjopen-2024-085971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024] Open
Abstract
OBJECTIVES To investigate the serum IL-6 levels and their rate of change in predicting the mortality of critically ill patients with COVID-19 in Vietnam. DESIGN A single-centre, cross-sectional study. SETTING An Intensive Care Centre for the Treatment of Critically Ill Patients with COVID-19 in Ho Chi Minh City, Vietnam. PARTICIPANTS We included patients aged 18 years or older who were critically ill with COVID-19 and presented to the study centre from 30 July 2021 to 15 October 2021. We excluded patients who did not have serum IL-6 measurements between admission and the end of the first day. PRIMARY OUTCOME MEASURES The primary outcome was hospital all-cause mortality. RESULTS Of 90 patients, 41.1% were men, the median age was 60.5 years (Q1-Q3: 52.0-71.0), and 76.7% of patients died in the hospital. Elevated IL-6 levels were observed on admission (41.79 pg/mL; Q1-Q3: 20.68-106.27) and on the third day after admission (72.00 pg/mL; Q1-Q3: 26.98-186.50), along with a significant rate of change in IL-6 during that period (839.5%; SD: 2753.2). While admission IL-6 level (areas under the receiver operator characteristic curve (AUROC): 0.610 (95% CI: 0.459 to 0.761); cut-off value ≥15.8 pg/mL) and rate of change in IL-6 on the third day of admission (AUROC: 0.586 (95% CI: 0.420 to 0.751); cut-off value ≥-58.7%) demonstrated poor discriminatory ability in predicting hospital mortality, the third day IL-6 rate of change from admission ≥-58.7% (adjusted OR: 12.812; 95% CI: 2.104 to 78.005) emerged as an independent predictor of hospital mortality. CONCLUSIONS This study focused on a highly selected cohort of critically ill COVID-19 patients with a high IL-6 level and mortality rate. Despite the poor discriminatory value of admission IL-6 levels, the rate of change in IL-6 proved valuable in predicting mortality. To identify critically ill COVID-19 patients with the highest risk for mortality, monitoring the serial serum IL-6 measurements and observing the rate of change in serum IL-6 levels over time are needed.
Collapse
Affiliation(s)
- Chi Van Nguyen
- Center for Emergency Medicine, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
| | - Chinh Quoc Luong
- Center for Emergency Medicine, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Co Xuan Dao
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| | - My Ha Nguyen
- Department of Health Organization and Management, Faculty of Public Health, Thai Binh University of Medicine and Pharmacy, Thai Binh, Viet Nam
| | - Dung Thi Pham
- Department of Nutrition and Food Safety, Faculty of Public Health, Thai Binh University of Medicine and Pharmacy, Thai Binh, Viet Nam
| | - Nhung Hong Khuat
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Intensive Care and Poison Control, Duc Giang General Hospital, Hanoi, Viet Nam
| | - Quynh Thi Pham
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Intensive Care Unit, University Medical Center Ho Chi Minh City, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Dat Tien Hoang
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| | - Anh Diep Nguyen
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Intensive Care Unit, Hanoi Heart Hospital, Hanoi, Viet Nam
| | - Phuong Minh Nguyen
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Intensive Care Unit, Thanh Nhan General Hospital, Hanoi, Viet Nam
| | - Duong Dai Cao
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Intensive Care and Poison Control, Ha Dong General Hospital, Hanoi, Viet Nam
| | - Dung Thuy Pham
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Stroke Center, Bach Mai Hospital, Hanoi, Viet Nam
| | - Thai Quoc Nguyen
- Center for Tropical Diseases, Bach Mai Hospital, Hanoi, Viet Nam
| | - Vuong Minh Nong
- Center for Tropical Diseases, Bach Mai Hospital, Hanoi, Viet Nam
| | - Dung Tuan Dang
- Center for Emergency Medicine, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Dat Tuan Nguyen
- Center for Emergency Medicine, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Vinh Duc Nguyen
- Center for Emergency Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| | - Thuan Quang Le
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Poison Control Center, Bach Mai Hospital, Hanoi, Viet Nam
| | - Viet Khoi Nguyen
- Radiology Centre, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Radiology, Hanoi Medical University, Hanoi, Viet Nam
| | - Hung Duc Ngo
- Center for Emergency Medicine, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
| | - Dung Van Nguyen
- Center for Tropical Diseases, Bach Mai Hospital, Hanoi, Viet Nam
| | - Thach The Pham
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| | - Dung Tien Nguyen
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Poison Control Center, Bach Mai Hospital, Hanoi, Viet Nam
| | - Nguyen Trung Nguyen
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
- Poison Control Center, Bach Mai Hospital, Hanoi, Viet Nam
| | - Tan Dang Do
- Radiology Centre, Bach Mai Hospital, Hanoi, Viet Nam
| | - Nhung Thi Huynh
- Department of Internal Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Nga Thu Phan
- Department of Health Organization and Management, Faculty of Public Health, Thai Binh University of Medicine and Pharmacy, Thai Binh, Viet Nam
| | - Cuong Duy Nguyen
- Department of Emergency and Critical Care Medicine, Thai Binh University of Medicine and Pharmacy, Thai Binh, Viet Nam
| | - Khoi Hong Vo
- Department of Neuro Intensive Care and Emergency Neurology, Neurology Center, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Neurology, Hanoi Medical University, Hanoi, Viet Nam
- Department of Neurology, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Thom Thi Vu
- Department of Basic Medical Sciences, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Cuong Duy Do
- Center for Tropical Diseases, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Infectious Diseases, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Tuan Quoc Dang
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| | - Giap Van Vu
- Department of Internal Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Respiratory Center, Bach Mai Hospital, Hanoi, Viet Nam
| | - Tan Cong Nguyen
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| | - Son Ngoc Do
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| |
Collapse
|
22
|
Gupta P, Gupta A, Bansal S, Sharma M, Saluja S, Balakrishnan I, Gupta K. Role of interleukin-6, serum ferritin, and d-dimer in hospitalized COVID-19 patients. Cytokine 2024; 184:156776. [PMID: 39383646 DOI: 10.1016/j.cyto.2024.156776] [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: 04/10/2024] [Revised: 07/24/2024] [Accepted: 09/30/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Various studies have observed an association between interleukin-6 (IL-6), serum ferritin, d-dimer, and in-hospital mortality in COVID-19 patients. However, multivariate regression analysis was not done in the majority of the studies, Also, the role of interleukin-6 (IL-6), serum ferritin, and d-dimer in hospitalized COVID-19 patients was not adequately studied and reported from our region. METHOD It was a retrospective cohort study in which the serum IL-6, serum ferritin, and d-dimer of 305 hospitalized COVID-19 patients were analyzed, and their association with mortality was determined. RESULTS In COVID-19 patients, the levels of IL-6 (P = 0.007), serum ferritin (P = 0.011), and d-dimer (P = 0.004) were significantly elevated in patients with severe SARS-CoV-2 illness (SpO2 < 90 % at admission). IL-6 levels were significantly elevated (186 pg/ml vs. 215 pg/ml, P = 0.003) in non-survivors compared to survivors. However, d-dimer (mg/ml) (P = 0.129) and serum ferritin (mg/ml) (P = 0.051) levels were similar between the two groups. The ROC curve (receiver operating characteristic curve) analysis showed a significant but poor area under the curve (AUC) between elevated IL-6 (>208 pg/ml) and in-hospital mortality (P < 0.008, AUC = 0.61). Kaplan-Meir survival analysis showed poor survival in patients with elevated IL-6 (>208 pg/ml) (Pby log-rank: 0.010) and elevated d-dimer (>1780 mg/ml) (P by log-rank: 0.036). The multivariate cox-regression analysis did not show any association between IL-6, serum ferritin, d-dimer, and in-hospital mortality (P > 0.05). Also, no association was found between serum levels of IL-6, serum ferritin, d-dimer, and the use of a ventilator (P > 0.05) and the severity of SARS-CoV-2 illness (P > 0.05) on multivariate binary logistic regression analysis. CONCLUSION In this study, the serum levels of IL-6, serum ferritin, and d-dimer were not associated with in-hospital mortality in hospitalized COVID-19 patients on multivariate cox-regression analysis, and were the markers of severe SARS-CoV-2 illness.
Collapse
Affiliation(s)
- Praveen Gupta
- Department of Cardiology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India.
| | - Anunay Gupta
- Department of Cardiology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Sandeep Bansal
- Department of Cardiology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Monica Sharma
- Department of Hematology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Sumita Saluja
- Department of Hematology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Ira Balakrishnan
- Department of Anesthesia and Critical Care, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Kapil Gupta
- Department of Anesthesia and Critical Care, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| |
Collapse
|
23
|
Leonard J, Sinha P. Precision Medicine in Acute Respiratory Distress Syndrome: Progress, Challenges, and the Road ahead. Clin Chest Med 2024; 45:835-848. [PMID: 39443001 PMCID: PMC11507056 DOI: 10.1016/j.ccm.2024.08.005] [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] [Indexed: 10/25/2024]
Abstract
Several novel high-dimensional biologic measurements are increasingly being applied to biomedical sciences. Acute respiratory distress syndrome (ARDS) is a theoretically fertile ground for such approaches. Not only are these biologic and analytic tools available to better understand ARDS but also arguably, simpler approaches such as respiratory physiology has been vastly underutilized as a means of delivering precision-based care in the field. Here we review the progress made in ARDS toward discovering biologically homogeneous phenotypes, treatment responsive subgroups, the challenges to implement these discoveries at the bedside, and the road ahead that will enable precision medicine in ARDS.
Collapse
Affiliation(s)
- Jennifer Leonard
- Department of Trauma and Acute Care Surgery, Washington University, 660 South Euclid Avenue, St Louis, MO 63110, USA
| | - Pratik Sinha
- Division of Clinical and Translational Research, Department of Anesthesia, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8054, St Louis, MO 63110, USA; Division of Critical Care, Department of Anesthesia, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8054, St Louis, MO 63110, USA.
| |
Collapse
|
24
|
Raza ML, Imam MH, Zehra W, Jamil S. Neuro-inflammatory pathways in COVID-19-induced central nervous system injury: Implications for prevention and treatment strategies. Exp Neurol 2024; 382:114984. [PMID: 39368535 DOI: 10.1016/j.expneurol.2024.114984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/15/2024] [Accepted: 09/29/2024] [Indexed: 10/07/2024]
Abstract
This review explores the neuroinflammatory pathways underlying COVID-19-induced central nervous system (CNS) injury, with a focus on mechanisms of brain damage and strategies for prevention. A comprehensive literature review was conducted to summarize current knowledge on the pathways by which SARS-CoV-2 reaches the brain, the neuroinflammatory responses triggered by viral infection, neurological symptoms and long COVID. Results: We discuss the mechanisms of neuroinflammation in COVID-19, including blood-brain barrier disruption, cytokine storm, microglial activation, and peripheral immune cell infiltration. Additionally, we highlight potential strategies for preventing CNS injury, including pharmacological interventions, immunomodulatory therapies, and lifestyle modifications. Conclusively, Understanding the neuroinflammatory pathways in COVID-19-induced CNS injury is crucial for developing effective prevention and treatment strategies to protect brain health during and after viral infection.
Collapse
Affiliation(s)
- Muhammad Liaquat Raza
- Department of Infection Prevention & Control, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia; King Abdullah International Medical Research Center, Riyadh, Saudi Arabia; King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| | | | | | - Subia Jamil
- Faculty of Pharmacy, Jinnah University for Women, University, Karachi, Pakistan
| |
Collapse
|
25
|
Liang P, Li Y, Meng L, Li Y, Mai H, Li T, Ma J, Ma J, Wang J, Zhuan B, Zhou W. Prognostic significance of serum interleukin-6 in severe/critical COVID-19 patients treated with tocilizumab: a detailed observational study analysis. Sci Rep 2024; 14:29634. [PMID: 39609511 PMCID: PMC11605089 DOI: 10.1038/s41598-024-81028-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024] Open
Abstract
Baseline IL-6 levels have been found to be non-predictive of subsequent outcomes following tocilizumab treatment, highlighting the need for more reliable predictive markers. To address this, a retrospective analysis was conducted on the clinical profiles, diagnostic tests, and follow-up prognoses of 60 patients with severe or critical COVID-19, all of whom were identified as experiencing a cytokine storm and subsequently received tocilizumab treatment. Among the patients, the overall survival rate during follow-up was 80%, with further analysis revealing that advanced age was an independent risk factor for adverse outcomes. Following tocilizumab administration, a statistically significant increase in IL-6 and D-dimer levels was observed, while markers such as C-reactive protein (CRP), procalcitonin (PCT), and fibrinogen demonstrated reductions compared to pre-treatment values. Specifically, IL-6 levels initially surged briefly after tocilizumab intervention before gradually diminishing. To assess the prognostic utility of IL-6, the Receiver Operating Characteristic (ROC) curve was employed, which yielded an area under the curve (AUC) of 0.812, indicating strong predictive capability, with a sensitivity of 100% and a specificity of 53.49%. The optimal cut-off value for IL-6 was identified at 147.79 pg/mL. In conclusion, IL-6 levels tend to rise transiently following tocilizumab therapy, before gradually declining. These post-treatment IL-6 measurements may serve as a valuable biomarker for assessing prognosis in patients undergoing this treatment.
Collapse
Affiliation(s)
- Panpan Liang
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yan Li
- Department of Anesthesiology, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Li'e Meng
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yuting Li
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Hailing Mai
- Department of Critical Care Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Tao Li
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Jiarui Ma
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Junhui Ma
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Jing Wang
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Bing Zhuan
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China.
| | - Wei Zhou
- Department of Respiratory Medicine, People's Hospital of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China.
| |
Collapse
|
26
|
Wick KD, Siegel L, Oldmixon C, Lundgren JD, Thompson BT, Jones C, Leroux C, Matthay MA. Longitudinal importance of the soluble receptor for advanced glycation end-products in nonintubated hospitalized patients with COVID-19 pneumonia. Am J Physiol Lung Cell Mol Physiol 2024; 327:L607-L614. [PMID: 39076084 PMCID: PMC11563646 DOI: 10.1152/ajplung.00350.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/31/2024] Open
Abstract
The soluble receptor for advanced glycation end-products (sRAGE) is a marker of alveolar type I cell injury associated with outcomes in COVID-19 pneumonia. How plasma sRAGE changes over time and whether it remains associated with long-term clinical outcomes beyond a single measurement in COVID-19 have not been well studied. We studied two cohorts in randomized clinical trials of monoclonal antibody treatment for COVID-19 (bamlanivimab and tixagevimab/cilgavimab). We first studied the association between baseline plasma sRAGE and 90-day clinical outcomes, which had been previously demonstrated in the bamlanivimab cohort, among hospitalized patients with COVID-19 supported with high-flow nasal oxygen (HFNO) or noninvasive ventilation (NIV) in the tixagevimab/cilgavimab study. Next, we investigated the relationship between day 3 sRAGE and 90-day outcomes and how plasma sRAGE changes over the first 3 days of hospitalization in both clinical trial cohorts. We found that plasma sRAGE in the highest quartile in the HFNO/NIV participants in the tixagevimab/cilgavimab trial was associated with a significantly lower rate of 90-day sustained recovery [recovery rate ratio = 0.31, 95% confidence interval (CI) = 0.14-0.71, P = 0.005] and with a significantly higher rate of 90-day mortality (hazard ratio = 2.49, 95% CI = 1.15-5.43, P = 0.021) compared with the lower three quartiles. Day 3 plasma sRAGE in both clinical trial cohorts remained associated with 90-day clinical outcomes. The trajectory of sRAGE was not influenced by treatment assignment. Our results indicate that plasma sRAGE is a valuable prognostic marker in COVID-19 up to 3 days after initial hospital presentation.NEW & NOTEWORTHY The soluble receptor for advanced glycation end-products (sRAGE) is a marker of alveolar type I epithelial cell injury associated with clinical outcomes in acute respiratory distress syndrome and, more recently, in hospitalized subjects with COVID-19. How plasma sRAGE changes over time and whether plasma sRAGE remains associated with long-term clinical outcomes beyond a single baseline measurement in patients with COVID-19 have not been well studied.
Collapse
Affiliation(s)
- Katherine D Wick
- Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Lianne Siegel
- Division of Biostatistics and Health Data Science, University of Minnesota, Minneapolis, Minnesota, United States
| | - Cathryn Oldmixon
- Biostatistics Center, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Jens D Lundgren
- CHIP (Centre of Excellence for Health, Immunity and Infections), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - B Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, Massachusetts, United States
| | - Chayse Jones
- Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Carolyn Leroux
- Cardiovascular Research Institute, University of California, San Francisco, California, United States
| | - Michael A Matthay
- Cardiovascular Research Institute, University of California, San Francisco, California, United States
- Department of Medicine and Anesthesia, University of California, San Francisco, California, United States
| |
Collapse
|
27
|
Tian R, Jiang J, Ding J, Zhao J, Zhou X. To study the impact of COVID-19 on the epidemiological characteristics of allergic rhinitis based on local big data in China. Sci Rep 2024; 14:25101. [PMID: 39443644 PMCID: PMC11500020 DOI: 10.1038/s41598-024-76252-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
To investigate the big data characteristics and trend changes of patients with allergic rhinitis (AR) who sought medical attention at our hospital before (from 2018 to 2019) and after (from 2020 to 2023) COVID-19, and provide reference basis for the treatment of AR. This study used a descriptive epidemiological method to analyze the big data and trend changes of AR patients. A total of 62,196 AR patients were collected, of whom 32,874 were male and 29,322 were female, with an age range of 1-89 years. The monthly change trend of AR patients showed a marked seasonality. The number of AR patients increased year by year. There was no significant difference in the number of patients between different genders. There was a significant difference in the number of patients between different age groups. The number of AR patients increased markedly from 2018 to 2023, and COVID-19 seems to have accelerated this process. There is a clear seasonal pattern. The number of boy patients is significantly higher than that of girl patients, and the change in female hormones may affect the incidence of AR. Therefore, it is necessary to update management measures and formulate relevant policies in response to the changing trend of AR since the COVID-19 epidemic.
Collapse
Affiliation(s)
- Ruru Tian
- Affiliated Hospital of Hangzhou Normal University, Hangzhou, China.
| | - Jianhua Jiang
- Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Jinv Ding
- Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Jieling Zhao
- Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Xinru Zhou
- Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
28
|
Xu R, Xia C, He X, Hu C, Li Y, Zhang Y, Chen Z. siRNA Nanoparticle Dry Powder Formulation with High Transfection Efficiency and Pulmonary Deposition for Acute Lung Injury Treatment. ACS APPLIED MATERIALS & INTERFACES 2024; 16:54344-54358. [PMID: 39325628 DOI: 10.1021/acsami.4c04241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Acute lung injury (ALI) is a severe inflammatory syndrome, which was caused by diverse factors. The COVID-19 pandemic has resulted in a higher mortality rate of these conditions. Currently, effective treatments are lacking. Although siRNA nucleotide-based drugs are promising therapeutic approaches, their poor stability and inability to efficiently reach target cells limit their clinical translation. This study identified a peptide from known cell-penetrating peptides that can form an efficient anti-inflammatory complex with TNF-α siRNA, termed SAR6EW/TNF-α siRNA. This complex can effectively transport TNF-α siRNA into the cytoplasm and achieve potent gene silencing in vitro as well as in vivo. By using lactose and triarginine as coexcipients and optimizing the spray-drying process, a powder was produced with micrometer-scale spherical and porous structures, enhancing aerosol release and lung delivery efficiency. The dry powder formulation and process preserve the stability and integrity of the siRNA. When administered intratracheally to ALI model mice, the complex powder demonstrated specific pulmonary gene silencing activity and significantly reduced inflammation symptoms caused by ALI, suggesting a potential strategy for the clinical therapeutic approach of respiratory diseases.
Collapse
Affiliation(s)
- Rui Xu
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chenjie Xia
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiongxiong He
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Changhui Hu
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yinjia Li
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yufeng Zhang
- Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin 214400, China
| | - Zhipeng Chen
- College of pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
29
|
Thys K, Loza MJ, Lynn L, Callewaert K, Varma L, Crabbe M, Van Wesenbeeck L, Van Landuyt E, De Meyer S, Aerssens J, Verbrugge I. Pharmacodynamic, prognostic, and predictive biomarkers in severe and critical COVID-19 patients treated with sirukumab. Sci Rep 2024; 14:22981. [PMID: 39362933 PMCID: PMC11452205 DOI: 10.1038/s41598-024-74196-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024] Open
Abstract
We examined candidate biomarkers for efficacy outcomes in hospitalized COVID-19 patients who were treated with sirukumab, an IL-6 neutralizing antibody, in a randomized, double-blind, placebo-controlled, phase 2 trial. Between May 2020 and March 2021, 209 patients were randomized (sirukumab, n = 139; placebo, n = 70); 112 had critical COVID-19. Serum biomarkers were evaluated for the pharmacodynamic effect of sirukumab and for their potential prognostic and predictive effect on time to sustained clinical improvement up to Day 28, clinical improvement at Day 28, and mortality at Day 28. The absence of detectable IL-4 increase and smaller increases in CCL13 post-baseline were most significantly associated with better response to sirukumab (versus placebo) treatment for all clinical efficacy outcomes tested, especially in patients with critical COVID-19. These data suggest that patients with critical COVID-19 without detectable sirukumab-induced IL-4 levels are more likely to benefit from sirukumab treatment. ClinicalTrials.gov Identifier: NCT04380961.
Collapse
Affiliation(s)
- Kim Thys
- Janssen Pharmaceutica NV, Beerse, Belgium
| | - Matthew J Loza
- Janssen Research & Development, LLC, PA, Spring House, USA.
| | | | | | - Lisa Varma
- Janssen Research & Development, LLC, PA, Spring House, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Vassiliou AG, Roumpaki A, Keskinidou C, Athanasiou N, Tsipilis S, Jahaj E, Vrettou CS, Giannopoulou V, Halioti A, Ferentinos G, Dimopoulou I, Kotanidou A, Langleben D, Orfanos SE. Transpulmonary Plasma Endothelin-1 Arterial:Venous Ratio Differentiates Survivors from Non-Survivors in Critically Ill Patients with COVID-19-Induced Acute Respiratory Distress Syndrome. Int J Mol Sci 2024; 25:10640. [PMID: 39408968 PMCID: PMC11476705 DOI: 10.3390/ijms251910640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Endothelin-1 (ET-1) is a potent vasoconstrictor produced by endothelial cells and cleared from circulating blood mainly in the pulmonary vasculature. In a healthy pulmonary circulation, the rate of local production of ET-1 is less than its rate of clearance. In the present study, we aimed to investigate whether the abnormal pulmonary circulatory handling of ET-1 relates to poor clinical outcomes in patients with coronavirus disease 2019 (COVID-19)-induced acute respiratory distress syndrome (ARDS). To this end, central venous and systemic arterial ET-1 plasma levels were simultaneously measured on Days 1 and 3 following ICU admission in mechanically ventilated COVID-19 patients with ARDS (COVID-19 ARDS, N = 18). Central venous and systemic arterial ET-1 plasma levels were also measured in two distinct SARS-CoV-2-negative mechanically ventilated critically ill patient groups, matched for age, sex, and critical illness severity, with ARDS (non-COVID-19 ARDS, N = 14) or without ARDS (non-COVID-19 non-ARDS, N = 20). Upon ICU admission, COVID-19-induced ARDS patients had higher systemic arterial and central venous ET-1 levels compared to the non-COVID-19 ARDS and non-COVID-19 non-ARDS patients (p < 0.05), yet a normal systemic arterial:central venous (A:V) ET-1 ratio [0.63 (0.49-1.02)], suggesting that pulmonary ET-1 clearance is intact in these patients. On the other hand, the non-COVID-19 ARDS patients demonstrated abnormal ET-1 handling [A:V ET-1 ratio 1.06 (0.93-1.20)], while the non-COVID-19 non-ARDS group showed normal ET-1 handling [0.79 (0.52-1.11)]. On Day 3, the A:V ratio in all three groups was <1. When the COVID-19 ARDS patients were divided based on 28-day ICU mortality, while their systemic arterial and central venous levels did not differ, the A:V ET-1 ratio was statistically significantly higher upon ICU admission in the non-survivors [0.95 (0.78-1.34)] compared to the survivors [0.57 (0.48-0.92), p = 0.027]. Our results highlight the potential importance of ET-1 as both a biomarker and a therapeutic target in critically ill COVID-19 patients. The elevated A:V ET-1 ratio in non-survivors suggests that the early disruption of pulmonary ET-1 handling may be a key marker of poor prognosis.
Collapse
Affiliation(s)
- Alice G. Vassiliou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, “Evangelismos” Hospital, 10676 Athens, Greece; (A.R.); (C.K.); (N.A.); (S.T.); (E.J.); (C.S.V.); (V.G.); (A.H.); (G.F.); (I.D.); (A.K.)
| | - Anastasia Roumpaki
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, “Evangelismos” Hospital, 10676 Athens, Greece; (A.R.); (C.K.); (N.A.); (S.T.); (E.J.); (C.S.V.); (V.G.); (A.H.); (G.F.); (I.D.); (A.K.)
| | - Chrysi Keskinidou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, “Evangelismos” Hospital, 10676 Athens, Greece; (A.R.); (C.K.); (N.A.); (S.T.); (E.J.); (C.S.V.); (V.G.); (A.H.); (G.F.); (I.D.); (A.K.)
| | - Nikolaos Athanasiou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, “Evangelismos” Hospital, 10676 Athens, Greece; (A.R.); (C.K.); (N.A.); (S.T.); (E.J.); (C.S.V.); (V.G.); (A.H.); (G.F.); (I.D.); (A.K.)
| | - Stamatios Tsipilis
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, “Evangelismos” Hospital, 10676 Athens, Greece; (A.R.); (C.K.); (N.A.); (S.T.); (E.J.); (C.S.V.); (V.G.); (A.H.); (G.F.); (I.D.); (A.K.)
| | - Edison Jahaj
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, “Evangelismos” Hospital, 10676 Athens, Greece; (A.R.); (C.K.); (N.A.); (S.T.); (E.J.); (C.S.V.); (V.G.); (A.H.); (G.F.); (I.D.); (A.K.)
| | - Charikleia S. Vrettou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, “Evangelismos” Hospital, 10676 Athens, Greece; (A.R.); (C.K.); (N.A.); (S.T.); (E.J.); (C.S.V.); (V.G.); (A.H.); (G.F.); (I.D.); (A.K.)
| | - Vassiliki Giannopoulou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, “Evangelismos” Hospital, 10676 Athens, Greece; (A.R.); (C.K.); (N.A.); (S.T.); (E.J.); (C.S.V.); (V.G.); (A.H.); (G.F.); (I.D.); (A.K.)
| | - Asimenia Halioti
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, “Evangelismos” Hospital, 10676 Athens, Greece; (A.R.); (C.K.); (N.A.); (S.T.); (E.J.); (C.S.V.); (V.G.); (A.H.); (G.F.); (I.D.); (A.K.)
| | - Georgios Ferentinos
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, “Evangelismos” Hospital, 10676 Athens, Greece; (A.R.); (C.K.); (N.A.); (S.T.); (E.J.); (C.S.V.); (V.G.); (A.H.); (G.F.); (I.D.); (A.K.)
| | - Ioanna Dimopoulou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, “Evangelismos” Hospital, 10676 Athens, Greece; (A.R.); (C.K.); (N.A.); (S.T.); (E.J.); (C.S.V.); (V.G.); (A.H.); (G.F.); (I.D.); (A.K.)
| | - Anastasia Kotanidou
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, “Evangelismos” Hospital, 10676 Athens, Greece; (A.R.); (C.K.); (N.A.); (S.T.); (E.J.); (C.S.V.); (V.G.); (A.H.); (G.F.); (I.D.); (A.K.)
| | - David Langleben
- Center for Pulmonary Vascular Disease, Division of Cardiology, Azrieli Heart Center and Lady Davis Institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada;
| | - Stylianos E. Orfanos
- First Department of Critical Care Medicine & Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, “Evangelismos” Hospital, 10676 Athens, Greece; (A.R.); (C.K.); (N.A.); (S.T.); (E.J.); (C.S.V.); (V.G.); (A.H.); (G.F.); (I.D.); (A.K.)
| |
Collapse
|
31
|
Deus MDC, Gadotti AC, Dias ES, Monte Alegre JB, Van Spitzenbergen BAK, Andrade GB, Tozoni SS, Stocco RB, Olandoski M, Tuon FFB, Pinho RA, de Noronha L, Baena CP, Moreno-Amaral AN. Prospective Variation of Cytokine Trends during COVID-19: A Progressive Approach from Disease Onset until Outcome. Int J Mol Sci 2024; 25:10578. [PMID: 39408907 PMCID: PMC11477561 DOI: 10.3390/ijms251910578] [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: 08/21/2024] [Revised: 09/19/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
COVID-19 is characterized by pronounced hypercytokinemia. The cytokine switch, marked by an imbalance between pro-inflammatory and anti-inflammatory cytokines, emerged as a focal point of investigation throughout the COVID-19 pandemic. However, the kinetics and temporal dynamics of cytokine release remain contradictory, making the development of new therapeutics difficult, especially in severe cases. This study collected serum samples from SARS-CoV-2 infected patients at 72 h intervals and monitored them for various cytokines at each timepoint until hospital discharge or death. Cytokine levels were analyzed based on time since symptom onset and patient outcomes. All cytokines studied prospectively were strong predictors of mortality, particularly IL-4 (AUC = 0.98) and IL-1β (AUC = 0.96). First-timepoint evaluations showed elevated cytokine levels in the mortality group (p < 0.001). Interestingly, IFN-γ levels decreased over time in the death group but increased in the survival group. Patients who died exhibited sustained levels of IL-1β and IL-4 and increased IL-6 levels over time. These findings suggest cytokine elevation is crucial in predicting COVID-19 mortality. The dynamic interplay between IFN-γ and IL-4 highlights the balance between Th1/Th2 immune responses and underscores IFN-γ as a powerful indicator of immune dysregulation throughout the infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Andrea Novais Moreno-Amaral
- Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Escola de Medicina, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, PR, Brazil; (M.d.C.D.); (A.C.G.); (E.S.D.); (J.B.M.A.); (B.A.K.V.S.); (G.B.A.); (S.S.T.); (R.B.S.); (M.O.); (F.F.B.T.); (R.A.P.); (L.d.N.); (C.P.B.)
| |
Collapse
|
32
|
Jaishwal P, Jha K, Singh SP. Revisiting the dimensions of universal vaccine with special focus on COVID-19: Efficacy versus methods of designing. Int J Biol Macromol 2024; 277:134012. [PMID: 39048013 DOI: 10.1016/j.ijbiomac.2024.134012] [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: 10/28/2023] [Revised: 05/28/2024] [Accepted: 07/17/2024] [Indexed: 07/27/2024]
Abstract
Even though the use of SARS-CoV-2 vaccines during the COVID-19 pandemic showed unprecedented success in a short time, it also exposed a flaw in the current vaccine design strategy to offer broad protection against emerging variants of concern. However, developing broad-spectrum vaccines is still a challenge for immunologists. The development of universal vaccines against emerging pathogens and their variants appears to be a practical solution to mitigate the economic and physical effects of the pandemic on society. Very few reports are available to explain the basic concept of universal vaccine design and development. This review provides an overview of the innate and adaptive immune responses generated against vaccination and essential insight into immune mechanisms helpful in designing universal vaccines targeting influenza viruses and coronaviruses. In addition, the characteristics, safety, and factors affecting the efficacy of universal vaccines have been discussed. Furthermore, several advancements in methods worthy of designing universal vaccines are described, including chimeric immunogens, heterologous prime-boost vaccines, reverse vaccinology, structure-based antigen design, pan-reactive antibody vaccines, conserved neutralizing epitope-based vaccines, mosaic nanoparticle-based vaccines, etc. In addition to the several advantages, significant potential constraints, such as defocusing the immune response and subdominance, are also discussed.
Collapse
Affiliation(s)
- Puja Jaishwal
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | - Kisalay Jha
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| | | |
Collapse
|
33
|
Wang R, Lan C, Benlagha K, Camara NOS, Miller H, Kubo M, Heegaard S, Lee P, Yang L, Forsman H, Li X, Zhai Z, Liu C. The interaction of innate immune and adaptive immune system. MedComm (Beijing) 2024; 5:e714. [PMID: 39286776 PMCID: PMC11401974 DOI: 10.1002/mco2.714] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 08/11/2024] [Accepted: 08/11/2024] [Indexed: 09/19/2024] Open
Abstract
The innate immune system serves as the body's first line of defense, utilizing pattern recognition receptors like Toll-like receptors to detect pathogens and initiate rapid response mechanisms. Following this initial response, adaptive immunity provides highly specific and sustained killing of pathogens via B cells, T cells, and antibodies. Traditionally, it has been assumed that innate immunity activates adaptive immunity; however, recent studies have revealed more complex interactions. This review provides a detailed dissection of the composition and function of the innate and adaptive immune systems, emphasizing their synergistic roles in physiological and pathological contexts, providing new insights into the link between these two forms of immunity. Precise regulation of both immune systems at the same time is more beneficial in the fight against immune-related diseases, for example, the cGAS-STING pathway has been found to play an important role in infections and cancers. In addition, this paper summarizes the challenges and future directions in the field of immunity, including the latest single-cell sequencing technologies, CAR-T cell therapy, and immune checkpoint inhibitors. By summarizing these developments, this review aims to enhance our understanding of the complexity interactions between innate and adaptive immunity and provides new perspectives in understanding the immune system.
Collapse
Affiliation(s)
- Ruyuan Wang
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Caini Lan
- Cancer Center Union Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Kamel Benlagha
- Alloimmunity, Autoimmunity and Transplantation Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160 Paris France
| | - Niels Olsen Saraiva Camara
- Department of Immunology Institute of Biomedical Sciences University of São Paulo (USP) São Paulo São Paulo Brazil
| | - Heather Miller
- Coxiella Pathogenesis Section, Laboratory of Bacteriology Rocky Mountain Laboratories National Institute of Allergy and Infectious Diseases, National Institutes of Health Hamilton Montana USA
| | - Masato Kubo
- Division of Molecular Pathology Research Institute for Biomedical Sciences (RIBS) Tokyo University of Science Noda Chiba Japan
| | - Steffen Heegaard
- Department of Ophthalmology Rigshospitalet Hospital Copenhagen University Copenhagen Denmark
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine Li Ka Shing Faculty of Medicine The University of Hong Kong Hong Kong China
| | - Lu Yang
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| | - Huamei Forsman
- Department of Laboratory Medicine Institute of Biomedicine, University of Gothenburg Gothenburg Sweden
| | - Xingrui Li
- Department of Thyroid and Breast Surgery Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan China
| | - Zhimin Zhai
- Department of Hematology The Second Hospital of Anhui Medical University Hefei China
| | - Chaohong Liu
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology Wuhan Hubei China
| |
Collapse
|
34
|
Cidade JP, Souza-Dantas VC, Mamfrim RB, Miranda RC, Caroli HT, Oliveira NA, Thompson AEF, Oliveira GE, Póvoa P. Advancing insights in critical COVID-19: unraveling lymphopenia through propensity score matching - Findings from the Multicenter LYMPH-COVID Study. CRITICAL CARE SCIENCE 2024; 36:e20240236en. [PMID: 39356899 PMCID: PMC11463993 DOI: 10.62675/2965-2774.20240236-en] [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: 09/26/2023] [Accepted: 04/25/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE To elucidate the impact of lymphopenia on critical COVID-19 patient outcomes. METHODS We conducted a multicenter prospective cohort study across five hospitals in Portugal and Brazil from 2020 to 2021. The study included adult patients admitted to the intensive care unit with SARS-CoV-2 pneumonia. Patients were categorized into two groups based on their lymphocyte counts within 48 hours of intensive care unit admission: the Lymphopenia Group (lymphocyte serum count < 1 × 109/L) and the Nonlymphopenia Group. Multivariate logistic regression, propensity score matching, Kaplan‒Meier survival curve analysis and Cox proportional hazards regression analysis were used. RESULTS A total of 912 patients were enrolled, with 191 (20.9%) in the Nonlymphopenia Group and 721 (79.1%) in the Lymphopenia Group. Lymphopenia patients displayed significantly elevated disease severity indices, including Sequential Organ Failure Assessment and Simplified Acute Physiology Score 3 scores, at intensive care unit admission (p = 0.001 and p < 0.001, respectively). Additionally, they presented heightened requirements for vasopressor support (p = 0.045) and prolonged intensive care unit and in-hospital stays (both p < 0.001). Multivariate logistic regression analysis after propensity score matching revealed a significant contribution of lymphopenia to mortality, with an odds ratio of 1,621 (95%CI: 1,275 - 2,048; p < 0.001). Interaction models revealed an increase of 8% in mortality for each decade of longevity in patients with concomitant lymphopenia. In the subanalysis utilizing three-group stratification, the Severe Lymphopenia Group had the highest mortality rate, not only in direct comparisons but also in Kaplan‒Meier survival analysis (log-rank test p = 0.0048). CONCLUSION Lymphopenia in COVID-19 patients is associated with increased disease severity and an increased risk of mortality, underscoring the need for prompt support for critically ill high-risk patients. These findings offer important insights into improving patient care strategies for COVID-19 patients.
Collapse
Affiliation(s)
- José Pedro Cidade
- Department of Intensive CareHospital São Francisco XavierCentro Hospitalar Lisboa OcidentalLisbonPortugalIntensive Care Unit 4, Department of Intensive Care, Hospital São Francisco Xavier, Centro Hospitalar Lisboa Ocidental - Lisbon, Portugal.
| | - Vicente Cês Souza-Dantas
- Instituto D’Or de Pesquisa e EnsinoRio de JaneiroRJBrazilInstituto D’Or de Pesquisa e Ensino - Rio de Janeiro (RJ), Brazil.
| | - Rafaela Braga Mamfrim
- Hospital Copa D’OrRio de JaneiroRJBrazilHospital Copa D’Or - Rio de Janeiro (RJ), Brazil.
| | | | | | | | | | - Gabriela E. Oliveira
- Hospital Copa D’OrRio de JaneiroRJBrazilHospital Copa D’Or - Rio de Janeiro (RJ), Brazil.
| | - Pedro Póvoa
- Department of Intensive CareHospital São Francisco XavierCentro Hospitalar Lisboa OcidentalLisbonPortugalIntensive Care Unit 4, Department of Intensive Care, Hospital São Francisco Xavier, Centro Hospitalar Lisboa Ocidental - Lisbon, Portugal.
| |
Collapse
|
35
|
Jaiswal A, Shrivastav S, Kushwaha HR, Chaturvedi R, Singh RP. Oncogenic potential of SARS-CoV-2-targeting hallmarks of cancer pathways. Cell Commun Signal 2024; 22:447. [PMID: 39327555 PMCID: PMC11426004 DOI: 10.1186/s12964-024-01818-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/04/2024] [Indexed: 09/28/2024] Open
Abstract
The 2019 outbreak of SARS-CoV-2 has caused a major worldwide health crisis with high rates of morbidity and death. Interestingly, it has also been linked to cancer, which begs the issue of whether it plays a role in carcinogenesis. Recent studies have revealed various mechanisms by which SARS-CoV-2 can influence oncogenic pathways, potentially promoting cancer development. The virus encodes several proteins that alter key signaling pathways associated with cancer hallmarks. Unlike classical oncogenic viruses, which transform cells through viral oncogenes or by activating host oncogenes, SARS-CoV-2 appears to promote tumorigenesis by inhibiting tumor suppressor genes and pathways while activating survival, proliferation, and inflammation-associated signaling cascades. Bioinformatic analyses and experimental studies have identified numerous interactions between SARS-CoV-2 proteins and cellular components involved in cancer-related processes. This review explores the intricate relationship between SARS-CoV-2 infection and cancer, focusing on the regulation of key hallmarks driving initiation, promotion and progression of cancer by viral proteins. By elucidating the underlying mechanisms driving cellular transformation, the potential of SARS-CoV-2 as an oncovirus is highlighted. Comprehending these interplays is essential to enhance our understanding of COVID-19 and cancer biology and further formulating strategies to alleviate SARS-CoV-2 influence on cancer consequences.
Collapse
Affiliation(s)
- Aishwarya Jaiswal
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Sanah Shrivastav
- SRM Institute of Science and Technology, Delhi-NCR Campus, Ghaziabad, Uttar Pradesh, India
| | - Hemant R Kushwaha
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Rana P Singh
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi, India.
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
36
|
Singh K, Rubenstein K, Callier V, Shaw-Saliba K, Rupert A, Dewar R, Laverdure S, Highbarger H, Lallemand P, Huang ML, Jerome KR, Sampoleo R, Mills MG, Greninger AL, Juneja K, Porter D, Benson CA, Dempsey W, El Sahly HM, Focht C, Jilg N, Paules CI, Rapaka RR, Uyeki TM, Clifford Lane H, Beigel J, Dodd LE. SARS-CoV-2 RNA and Nucleocapsid Antigen Are Blood Biomarkers Associated With Severe Disease Outcomes That Improve in Response to Remdesivir. J Infect Dis 2024; 230:624-634. [PMID: 38657001 PMCID: PMC11420797 DOI: 10.1093/infdis/jiae198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/09/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Although antivirals remain important for the treatment COVID-19, methods to assess treatment efficacy are lacking. Here, we investigated the impact of remdesivir on viral dynamics and their contribution to understanding antiviral efficacy in the multicenter Adaptive COVID-19 Treatment Trial 1, which randomized patients to remdesivir or placebo. METHODS Longitudinal specimens collected during hospitalization from a substudy of 642 patients with COVID-19 were measured for viral RNA (upper respiratory tract and plasma), viral nucleocapsid antigen (serum), and host immunologic markers. Associations with clinical outcomes and response to therapy were assessed. RESULTS Higher baseline plasma viral loads were associated with poorer clinical outcomes, and decreases in viral RNA and antigen in blood but not the upper respiratory tract correlated with enhanced benefit from remdesivir. The treatment effect of remdesivir was most pronounced in patients with elevated baseline nucleocapsid antigen levels: the recovery rate ratio was 1.95 (95% CI, 1.40-2.71) for levels >245 pg/mL vs 1.04 (95% CI, .76-1.42) for levels <245 pg/mL. Remdesivir also accelerated the rate of viral RNA and antigen clearance in blood, and patients whose blood levels decreased were more likely to recover and survive. CONCLUSIONS Reductions in SARS-CoV-2 RNA and antigen levels in blood correlated with clinical benefit from antiviral therapy. CLINICAL TRIAL REGISTRATION NCT04280705 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Kanal Singh
- National Institute of Allergy and Infectious Diseases, Bethesda
| | - Kevin Rubenstein
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research
| | - Viviane Callier
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research
| | | | - Adam Rupert
- National Laboratory for Cancer Research, Frederick, Maryland
| | - Robin Dewar
- National Laboratory for Cancer Research, Frederick, Maryland
| | | | | | | | - Meei-Li Huang
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
| | - Keith R Jerome
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
- Fred Hutchinson Cancer Center, Seattle, Washington
| | - Reigran Sampoleo
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
| | - Margaret G Mills
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
| | - Alexander L Greninger
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington
| | | | | | | | - Walla Dempsey
- National Institute of Allergy and Infectious Diseases, Bethesda
| | - Hana M El Sahly
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | | | - Nikolaus Jilg
- Massachusetts General Hospital and Brigham and Women's Hospital, Harvard Medical School, Boston
| | - Catharine I Paules
- Division of Infectious Diseases, Milton S. Hershey Medical Center, Penn State Health, Hershey, Pennsylvania
| | - Rekha R Rapaka
- Center for Vaccine Development and Global Health, School of Medicine, University of Maryland, Baltimore
| | - Timothy M Uyeki
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - H Clifford Lane
- National Institute of Allergy and Infectious Diseases, Bethesda
| | - John Beigel
- National Institute of Allergy and Infectious Diseases, Bethesda
| | - Lori E Dodd
- National Institute of Allergy and Infectious Diseases, Bethesda
| |
Collapse
|
37
|
Farnesi-de-Assunção TS, Oliveira-Scussel ACDM, Rodrigues WF, Matos BS, da Silva DAA, de Andrade E Silva LE, Mundim FV, Helmo FR, Bernardes E Borges AV, Desidério CS, Trevisan RO, Obata MMS, Barbosa LM, Lemes MR, Costa-Madeira JC, Barbosa RM, Cunha ACCH, Pereira LQ, Tanaka SCSV, de Vito FB, Monteiro IB, Ferreira YM, Machado GH, Moraes-Souza H, Rodrigues DBR, de Oliveira CJF, da Silva MV, Júnior VR. COVID-19 Inflammatory Syndrome: Lessons from TNFRI and CRP about the Risk of Death in Severe Disease. Biomedicines 2024; 12:2138. [PMID: 39335653 PMCID: PMC11428742 DOI: 10.3390/biomedicines12092138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Background/Objectives: Cytokine storm in severe COVID-19 is responsible for irreversible tissue damage and death. Soluble mediators from the TNF superfamily, their correlation with clinical outcome, and the use of TNF receptors as a potent predictor for clinical outcome were evaluated. Methods: Severe COVID-19 patients had the levels of soluble mediators from the TNF superfamily quantified and categorized according to the clinical outcome (death versus survival). Statistical modeling was performed to predict clinical outcomes. Results: COVID-19 patients have elevated serum levels from the TNF superfamily. Regardless of sex and age, the sTNFRI levels were observed to be significantly higher in deceased patients from the first weeks following the onset of symptoms. We analyzed hematological parameters and inflammatory markers, and there was a difference between the groups for the following factors: erythrocytes, hemoglobin, hematocrit, leukocytes, neutrophils, band cells, lymphocytes, monocytes, CRP, IL-8, IFN-γ, IL-10, IL-6, IL-4, IL-2, leptin MIF sCD40L, and sTNFRI (p < 0.05). A post hoc analysis showed an inferential capacity over 70% for some hematological markers, CRP, and inflammatory mediators in deceased patients. sTNFRI was strongly associated with death, and the sTNFRI/sTNFRII ratio differed between outcomes (p < 0.001; power above 90%), highlighting the impact of these proteins on clinical results. The final logistic model, including sTNFRI/sTNFRII and CRP, indicated high sensitivity, specificity, accuracy, and an eight-fold higher odds ratio for an unfavorable outcome. Conclusions: The joint use of the sTNFRI/sTNFRII ratio with CRP proves to be a promising tool to assist in the clinical management of patients hospitalized for COVID-19.
Collapse
Affiliation(s)
| | | | - Wellington Francisco Rodrigues
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Beatriz Sodré Matos
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Djalma Alexandre Alves da Silva
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Leonardo Eurípedes de Andrade E Silva
- Clinical Analysis and Pathological Anatomy Laboratory, Empresa Brasileira de Serviços Hospitalares (EBSERH), Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Fabiano Vilela Mundim
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Fernanda Rodrigues Helmo
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | | | - Chamberttan Souza Desidério
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Rafael Obata Trevisan
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Malu Mateus Santos Obata
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Laís Milagres Barbosa
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Marcela Rezende Lemes
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Juliana Cristina Costa-Madeira
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Rafaela Miranda Barbosa
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | | | - Loren Queli Pereira
- Hematological Research Laboratory, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | | | | | - Ivan Borges Monteiro
- UNIMED São Domingos Hospital, Uberaba 38025-110, MG, Brazil
- Alencar Gomes da Silva Regional Hospital, Uberaba 38060-200, MG, Brazil
| | | | | | - Hélio Moraes-Souza
- Hematological Research Laboratory, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Denise Bertulucci Rocha Rodrigues
- Centro de Formação Especial em Saúde (CEFORES), Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
- Department of Immunology, Medical School, University of Uberaba, Uberaba 38010-200, MG, Brazil
| | - Carlo José Freire de Oliveira
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Marcos Vinicius da Silva
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Virmondes Rodrigues Júnior
- Department of Microbiology, Immunology and Parasitology, Federal University of Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| |
Collapse
|
38
|
Demeter F, Bihari G, Vadicsku D, Sinkovits G, Kajdácsi E, Horváth L, Réti M, Müller V, Iványi Z, Gál J, Gopcsa L, Reményi P, Szathmáry B, Lakatos B, Szlávik J, Bobek I, Prohászka ZZ, Förhécz Z, Masszi T, Vályi-Nagy I, Prohászka Z, Cervenak L. Anti-Inflammatory Cytokine Profiles in Thrombotic Thrombocytopenic Purpura-Differences Compared to COVID-19. Int J Mol Sci 2024; 25:10007. [PMID: 39337495 PMCID: PMC11432022 DOI: 10.3390/ijms251810007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Thromboinflammation/immunothrombosis plays a role in several diseases including thrombotic thrombocytopenic purpura (TTP) and COVID-19. Unlike the extensive research that has been conducted on COVID-19 cytokine storms, the baseline and acute phase cytokine profiles of TTP are poorly characterized. Moreover, we compared the cytokine profiles of TTP and COVID-19 to identify the disease-specific/general characteristics of thromboinflammation/immunothrombosis. Plasma concentrations of 33 soluble mediators (SMs: cytokines, chemokines, soluble receptors, and growth factors) were measured by multiplex bead-based LEGENDplex™ immunoassay from 32 COVID-19 patients (32 non-vaccinated patients in three severity groups), 32 TTP patients (remission/acute phase pairs of 16 patients), and 15 control samples. Mainly, the levels of innate immunity-related SMs changed in both diseases. In TTP, ten SMs decreased in both remission and acute phases compared to the control, one decreased, and two increased only in the acute phase compared to remission, indicating mostly anti-inflammatory changes. In COVID-19, ten pro-inflammatory SMs increased, whereas one decreased with increasing severity compared to the control. In severe COVID-19, sixteen SMs exceeded acute TTP levels, with only one higher in TTP. PCA identified CXCL10, IL-1RA, and VEGF as the main discriminators among their cytokine profiles. The innate immune response is altered in both diseases. The cytokine profile of TTP suggests a distinct pathomechanism from COVID-19 and supports referring to TTP as thromboinflammatory rather than immunothrombotic, emphasizing thrombosis over inflammation as the driving force of the acute phase.
Collapse
Affiliation(s)
- Flóra Demeter
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - György Bihari
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Dorina Vadicsku
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - György Sinkovits
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Erika Kajdácsi
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
- Research Group for Immunology and Hematology, Semmelweis University—HUN-REN-SU (Office for Supported Research Groups), 1085 Budapest, Hungary
| | - Laura Horváth
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Marienn Réti
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Veronika Müller
- Department of Pulmonology, Semmelweis University, 1083 Budapest, Hungary
| | - Zsolt Iványi
- Department of Anaesthesiology and Intensive Therapy, Semmelweis University, 1085 Budapest, Hungary
| | - János Gál
- Department of Anaesthesiology and Intensive Therapy, Semmelweis University, 1085 Budapest, Hungary
| | - László Gopcsa
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Péter Reményi
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Beáta Szathmáry
- Department of Infectology, Central Hospital of Southern Pest, National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Botond Lakatos
- Department of Infectology, Central Hospital of Southern Pest, National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - János Szlávik
- Department of Infectology, Central Hospital of Southern Pest, National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Ilona Bobek
- Department of Anaesthesiology and Intensive Therapy, Central Hospital of Southern Pest, National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Zita Z. Prohászka
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Zsolt Förhécz
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - Tamás Masszi
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| | - István Vályi-Nagy
- Department of Haematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Haematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Zoltán Prohászka
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
- Research Group for Immunology and Hematology, Semmelweis University—HUN-REN-SU (Office for Supported Research Groups), 1085 Budapest, Hungary
| | - László Cervenak
- Department of Internal Medicine and Haematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
39
|
Wang S, Li W, Wang Z, Yang W, Li E, Xia X, Yan F, Chiu S. Emerging and reemerging infectious diseases: global trends and new strategies for their prevention and control. Signal Transduct Target Ther 2024; 9:223. [PMID: 39256346 PMCID: PMC11412324 DOI: 10.1038/s41392-024-01917-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 09/12/2024] Open
Abstract
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhenshan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Wanying Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
- Department of Laboratory Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
40
|
Ling RR, Ramanathan K, Shen L, Barbaro RP, Shekar K, Brodie D, MacLaren G. Immunomodulators in patients receiving extracorporeal membrane oxygenation for COVID-19: a propensity-score adjusted analysis of the ELSO registry. Ann Intensive Care 2024; 14:128. [PMID: 39162921 PMCID: PMC11336150 DOI: 10.1186/s13613-024-01368-1] [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: 05/20/2024] [Accepted: 08/11/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Mortality for patients receiving extracorporeal membrane oxygenation (ECMO) for COVID-19 increased over the course of the pandemic. We investigated the association between immunomodulators and mortality for patients receiving ECMO for COVID-19. METHODS We retrospectively analysed the Extracorporeal Life Support Organisation registry from 1 January, 2020, through 31 December, 2021, to compare the outcomes of patients who received no immunomodulators, only corticosteroids, only other immunomodulators (selective interleukin blockers, janus-kinase inhibitors, convalescent plasma, and intravenous immunoglobulin), and a combination of corticosteroids and other immunomodulators administered either before or during ECMO. We used Cox regression models to estimate survival time until 90 days. We estimated the propensity score of receiving different immunomodulators using multinomial regression, and incorporated these scores into the regression models. RESULTS We included 7181 patients in the final analysis; 6169 patients received immunomodulators either before or during ECMO. The 90-day survival was 58.1% (95%-CI 55.1-61.2%) for patients receiving no immunomodulators, 50.7% (95%-CI 49.0-52.5%) for those receiving only corticosteroids, 62.2% (95%-CI 57.4-67.0%) for those receiving other immunomodulators, and 48.5% (95%-CI 46.7-50.4%) for those receiving corticosteroids and other immunomodulators. Compared to patients without immunomodulators, patients receiving either corticosteroids alone (HR: 1.13, 95%-CI 1.01-1.28) or with other immunomodulators (HR: 1.21, 95%-CI: 1.07-1.54) had significantly shorter survival time, while patients receiving only other immunomodulators had significantly longer survival time (HR: 0.79, 95%-CI: 0.66-0.96). The receipt of immunomodulators (across all three groups) was associated with an increase in secondary infections. CONCLUSIONS In this cohort study, we found that immunomodulators, in particular corticosteroids, were associated with significantly higher mortality amongst patients receiving ECMO for COVID-19, after adjusting for potential confounding variables and propensity score. In addition, patients receiving corticosteroids with or without other immunomodulators had longer ECMO runs, which has potential implications for resource allocation. While residual confounding likely remains, further studies are required to evaluate the timing of immunomodulators and better understand the possible mechanisms behind this association, including secondary infections.
Collapse
Affiliation(s)
- Ryan Ruiyang Ling
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Level 9, 1E Kent Ridge Road, Singapore, 119228, Singapore
| | - Kollengode Ramanathan
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Level 9, 1E Kent Ridge Road, Singapore, 119228, Singapore.
- Cardiothoracic Intensive Care Unit, National University Heart Centre, National University Hospital, National University Health System, Singapore, Singapore.
| | - Liang Shen
- Biostatistics Unit, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore, Singapore
| | - Ryan P Barbaro
- Division of Paediatrics Critical Care Medicine, University of Michigan, Ann Arbor, USA
- Child Health Evaluation and Research Center, University of Michigan, Ann Arbor, MI, USA
| | - Kiran Shekar
- Adult Intensive Care Services, Prince Charles Hospital, Brisbane, QLD, Australia
- Queensland University of Technology, Brisbane, Australia
- University of Queensland, Brisbane and Bond University, Gold Coast, QLD, Australia
| | - Daniel Brodie
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Graeme MacLaren
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Level 9, 1E Kent Ridge Road, Singapore, 119228, Singapore
- Cardiothoracic Intensive Care Unit, National University Heart Centre, National University Hospital, National University Health System, Singapore, Singapore
| |
Collapse
|
41
|
Hawryłkowicz V, Stasiewicz B, Maciejewska D, Sołek-Pastuszka J, Komorniak N, Skonieczna-Żydecka K, Martynova-Van Kley A, Stachowska E. The Link between Inflammation, Lipid Derivatives, and Microbiota Metabolites in COVID-19 Patients: Implications on Eating Behaviors and Nutritional Status. Int J Mol Sci 2024; 25:7899. [PMID: 39063142 PMCID: PMC11276903 DOI: 10.3390/ijms25147899] [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: 06/07/2024] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Extreme inflammation that continues even after infections can lead to a cytokine storm. In recent times, one of the most common causes of cytokine storm activation has been SARS-CoV-2 infection. A cytokine storm leads to dysregulation and excessive stimulation of the immune system, producing symptoms typical of post-COVID syndrome, including chronic fatigue, shortness of breath, joint pain, trouble concentrating (known as "brain fog"), and even direct organ damage in the heart, lungs, kidneys, and brain. This work summarizes the current knowledge regarding inflammation and the cytokine storm related to SARS-CoV-2 infection. Additionally, changes in lipid metabolism and microbiota composition under the influence of inflammation in COVID-19, along with the possible underlying mechanisms, are described. Finally, this text explores potential health implications related to changes in eating behaviors and nutritional status in COVID-19 patients. Although research on the cytokine storm is still ongoing, there is convincing evidence suggesting that severe immune and inflammatory responses during the acute phase of COVID-19 may lead to long-term health consequences. Understanding these links is key to developing treatment strategies and supporting patients after infection.
Collapse
Affiliation(s)
- Viktoria Hawryłkowicz
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (D.M.); (N.K.)
| | - Beata Stasiewicz
- Department of Human Nutrition, The Faculty of Food Science, University of Warmia and Mazury in Olsztyn, Sloneczna 45f, 10-718 Olsztyn, Poland
| | - Dominika Maciejewska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (D.M.); (N.K.)
| | - Joanna Sołek-Pastuszka
- Department of Anesthesiology and Intensive Care, Pomeranian Medical University, 71-242 Szczecin, Poland;
| | - Natalia Komorniak
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (D.M.); (N.K.)
| | | | | | - Ewa Stachowska
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, 71-460 Szczecin, Poland; (V.H.); (D.M.); (N.K.)
| |
Collapse
|
42
|
de Andres PJ, Ferreiro S, Flores A, Garcia A, Henriquez-Camacho C. Histological Assessment of Respiratory Tract and Liver of BALB/c Mice Nebulized with Tocilizumab. Pharmaceutics 2024; 16:862. [PMID: 39065559 PMCID: PMC11280351 DOI: 10.3390/pharmaceutics16070862] [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: 05/25/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Pulmonary drug delivery offers a minimally invasive and efficient method for treating lung conditions, leveraging the lungs' extensive surface area and blood flow for rapid drug absorption. Nebulized therapies aim to deliver drugs directly to the lung tissue. This study investigates the histological impact of nebulized tocilizumab-a monoclonal antibody targeting IL-6, traditionally administered intravenously for rheumatoid arthritis and severe COVID-19-on a murine model. Thirty BALB/c mice were nebulized with tocilizumab (10 mg, 5 mg, and 2.5 mg) and six controls were nebulized with saline solution. They were euthanized 48 h later, and their organs (lungs, nasal mucosa, and liver) were analyzed by a microscopic histological evaluation. The results indicate that all the mice survived the 48 h post-nebulization period without systemic compromise. The macroscopic examination showed no abnormalities, and the histopathological analysis revealed greater lung vascular changes in the control group than in the nebulized animals, which is attributable to the euthanasia with carbon dioxide. Additionally, increased alveolar macrophages were observed in the nebulized groups compared to controls. No significant histological changes were observed in the liver, indicating the safety of nebulized tocilizumab. In conclusion, these findings suggest the potential of nebulized tocilizumab for treating pulmonary inflammation, warranting further research to establish its efficacy and safety in clinical settings.
Collapse
Affiliation(s)
- Paloma Jimena de Andres
- Departamento de Medicina y Cirugía Animal, Facultad de Veterinaria de la, Universidad Complutense de Madrid, 28040 Madrid, Spain;
| | - Sergio Ferreiro
- Unidad de Veterinaria, Radiodiagnóstico y Cirugía Experimental del, Centro de Apoyo Tecnológico de la, Facultad de Ciencias de la Salud de la, Universidad Rey Juan Carlos, 28922 Alcorcon, Spain;
| | - Angela Flores
- Servicio de Farmacia del Hospital Universitario Rey Juan Carlos, 28993 Mostoles, Spain; (A.F.); (A.G.)
| | - Almudena Garcia
- Servicio de Farmacia del Hospital Universitario Rey Juan Carlos, 28993 Mostoles, Spain; (A.F.); (A.G.)
| | - Cesar Henriquez-Camacho
- Servicio de Medicina Interna del Hospital Universitario de Móstoles, 28935 Mostoles, Spain
- Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, 28922 Alcorcon, Spain
| |
Collapse
|
43
|
Visser MPJ, Dofferhoff ASM, van den Ouweland JMW, de Jong PA, Zanen P, van Daal H, Theeuwen EB, Kramers C, Janssen R, Walk J. Vitamin K2 Supplementation in Hospitalised COVID-19 Patients: A Randomised Controlled Trial. J Clin Med 2024; 13:3476. [PMID: 38930004 PMCID: PMC11205124 DOI: 10.3390/jcm13123476] [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: 05/02/2024] [Revised: 05/30/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Background: In observational studies, high levels of desphospho-uncarboxylated matrix gla protein (dp-ucMGP) that result from vitamin K deficiency were consistently associated with poor clinical outcomes during COVID-19. Vitamin K-activated matrix gla protein (MGP) is required to protect against elastic fibre degradation, and a deficiency may contribute to pathology. However, intervention trials assessing the effects of vitamin K supplementation in COVID-19 are lacking. Methods: This is a single-centre, phase 2, double-blind, randomised, placebo-controlled trial investigating the effects of vitamin K2 supplementation in 40 hospitalised COVID-19 patients requiring supplemental oxygen. Individuals were randomly assigned in a 1:1 ratio to receive 999 mcg of vitamin K2-menaquinone-7 (MK-7)-or a placebo daily until discharge or for a maximum of 14 days. Dp-ucMGP, the rate of elastic fibre degradation quantified by desmosine, and hepatic vitamin K status quantified by PIVKA-II were measured. Grade 3 and 4 adverse events were collected daily. As an exploratory objective, circulating vitamin K2 levels were measured. Results: Vitamin K2 was well tolerated and did not increase the number of adverse events. A linear mixed model analysis showed that dp-ucMGP and PIVKA-II decreased significantly in subjects that received supplementation compared to the controls (p = 0.008 and p = 0.0017, respectively), reflecting improved vitamin K status. The decrease in dp-ucMGP correlated with higher plasma MK-7 levels (p = 0.015). No significant effect on desmosine was found (p = 0.545). Conclusions: These results demonstrate that vitamin K2 supplementation during COVID-19 is safe and decreases dp-ucMGP. However, the current dose of vitamin K2 failed to show a protective effect against elastic fibre degradation.
Collapse
Affiliation(s)
- Margot P. J. Visser
- Department of Pulmonary Medicine, Canisius-Wilhelmina Hospital, 6532 SZ Nijmegen, The Netherlands
| | - Anton S. M. Dofferhoff
- Department of Internal Medicine, Canisius-Wilhelmina Hospital, 6532 SZ Nijmegen, The Netherlands
| | | | - Pim A. de Jong
- Department of Radiology, University Medical Centre Utrecht and Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Pieter Zanen
- Department of Pulmonary Diseases, St Antonius Hospital, 3435 CM Nieuwegein, The Netherlands
| | - Henny van Daal
- Department of Clinical Chemistry, Canisius-Wilhelmina Hospital, 6532 SZ Nijmegen, The Netherlands
| | - Eline B. Theeuwen
- Department of Pulmonary Medicine, Canisius-Wilhelmina Hospital, 6532 SZ Nijmegen, The Netherlands
| | - Cornelis Kramers
- Department of Pharmacology-Toxicology and Internal Medicine, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
| | - Rob Janssen
- Department of Pulmonary Medicine, Canisius-Wilhelmina Hospital, 6532 SZ Nijmegen, The Netherlands
| | - Jona Walk
- Department of Internal Medicine, Canisius-Wilhelmina Hospital, 6532 SZ Nijmegen, The Netherlands
| |
Collapse
|
44
|
Li X, Mi Z, Liu Z, Rong P. SARS-CoV-2: pathogenesis, therapeutics, variants, and vaccines. Front Microbiol 2024; 15:1334152. [PMID: 38939189 PMCID: PMC11208693 DOI: 10.3389/fmicb.2024.1334152] [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: 11/06/2023] [Accepted: 05/29/2024] [Indexed: 06/29/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), emerged in December 2019 with staggering economic fallout and human suffering. The unique structure of SARS-CoV-2 and its underlying pathogenic mechanism were responsible for the global pandemic. In addition to the direct damage caused by the virus, SARS-CoV-2 triggers an abnormal immune response leading to a cytokine storm, culminating in acute respiratory distress syndrome and other fatal diseases that pose a significant challenge to clinicians. Therefore, potential treatments should focus not only on eliminating the virus but also on alleviating or controlling acute immune/inflammatory responses. Current management strategies for COVID-19 include preventative measures and supportive care, while the role of the host immune/inflammatory response in disease progression has largely been overlooked. Understanding the interaction between SARS-CoV-2 and its receptors, as well as the underlying pathogenesis, has proven to be helpful for disease prevention, early recognition of disease progression, vaccine development, and interventions aimed at reducing immunopathology have been shown to reduce adverse clinical outcomes and improve prognosis. Moreover, several key mutations in the SARS-CoV-2 genome sequence result in an enhanced binding affinity to the host cell receptor, or produce immune escape, leading to either increased virus transmissibility or virulence of variants that carry these mutations. This review characterizes the structural features of SARS-CoV-2, its variants, and their interaction with the immune system, emphasizing the role of dysfunctional immune responses and cytokine storm in disease progression. Additionally, potential therapeutic options are reviewed, providing critical insights into disease management, exploring effective approaches to deal with the public health crises caused by SARS-CoV-2.
Collapse
Affiliation(s)
- Xi Li
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ze Mi
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhenguo Liu
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
45
|
Li Y, Gong Y, Xu G. New insights into kidney disease after COVID-19 infection and vaccination: histopathological and clinical findings. QJM 2024; 117:317-337. [PMID: 37402613 DOI: 10.1093/qjmed/hcad159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/23/2023] [Indexed: 07/06/2023] Open
Abstract
In addition to its pulmonary effects, coronavirus disease 2019 (COVID-19) has also been found to cause acute kidney injury (AKI), which has been linked to high mortality rates. In this review, we collected data from 20 clinical studies on post-COVID-19-related AKI and 97 cases of AKI associated with COVID-19 vaccination. Acute tubular injury was by far the most common finding in the kidneys of patients with COVID-19-related AKI. Among patients hospitalized for COVID-19, 34.0% developed AKI, of which 59.0%, 19.1% and 21.9% were Stages 1, 2 and 3, respectively. Though kidney disease and other adverse effects after COVID-19 vaccination overall appear rare, case reports have accumulated suggesting that COVID-19 vaccination may be associated with a risk of subsequent kidney disease. Among the patients with post-vaccination AKI, the most common pathologic findings include crescentic glomerulonephritis (29.9%), acute tubular injury (23.7%), IgA nephropathy (18.6%), antineutrophil cytoplasmic autoantibody-associated vasculitis (17.5%), minimal change disease (17.5%) and thrombotic microangiopathy (10.3%). It is important to note that crescentic glomerulonephritis appears to be more prevalent in patients who have newly diagnosed renal involvement. The proportions of patients with AKI Stages 1, 2 and 3 after COVID-19 vaccination in case reports were 30.9%, 22.7% and 46.4%, respectively. In general, clinical cases of new-onset and recurrent nephropathy with AKI after COVID-19 vaccination have a positive prognosis. In this article, we also explore the underlying pathophysiological mechanisms of AKI associated with COVID-19 infection and its vaccination by describing key renal morphological and clinical features and prognostic findings.
Collapse
Affiliation(s)
- Yebei Li
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang 330006, P.R. China
| | - Yan Gong
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang 330006, P.R. China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang 330006, P.R. China
| |
Collapse
|
46
|
Vazquez Guillamet MC, Vazquez Guillamet R, Rjob A, Reynolds D, Parikh B, Despotovic V, Byers DE, Ellebedy AH, Kollef MH, Mudd PA. Quantitative SARS-CoV-2 RT-PCR and Bronchoalveolar Cytokine Concentrations Redefine the COVID-19 Phenotypes in Critically Ill Patients. J Intensive Care Med 2024; 39:525-533. [PMID: 38629466 DOI: 10.1177/08850666231217707] [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: 05/14/2024]
Abstract
RATIONALE Recent studies suggest that both hypo- and hyperinflammatory acute respiratory distress syndrome (ARDS) phenotypes characterize severe COVID-19-related pneumonia. The role of lung Severe Acute Respiratory Syndrome - Coronavirus 2 (SARS-CoV-2) viral load in contributing to these phenotypes remains unknown. OBJECTIVES To redefine COVID-19 ARDS phenotypes when considering quantitative SARS-CoV-2 RT-PCR in the bronchoalveolar lavage of intubated patients. To compare the relevance of deep respiratory samples versus plasma in linking the immune response and the quantitative viral loads. METHODS Eligible subjects were adults diagnosed with COVID-19 ARDS who required mechanical ventilation and underwent bronchoscopy. We recorded the immune response in the bronchoalveolar lavage and plasma and the quantitative SARS-CoV-2 RT-PCR in the bronchoalveolar lavage. Hierarchical clustering on principal components was applied separately on the 2 compartments' datasets. Baseline characteristics were compared between clusters. MEASUREMENTS AND RESULTS Twenty subjects were enrolled between August 2020 and March 2021. Subjects underwent bronchoscopy on average 3.6 days after intubation. All subjects were treated with dexamethasone prior to bronchoscopy, 11 of 20 (55.6%) received remdesivir and 1 of 20 (5%) received tocilizumab. Adding viral load information to the classic 2-cluster model of ARDS revealed a new cluster characterized by hypoinflammatory responses and high viral load in 23.1% of the cohort. Hyperinflammatory ARDS was noted in 15.4% of subjects. Bronchoalveolar lavage clusters were more stable compared to plasma. CONCLUSIONS We identified a unique group of critically ill subjects with COVID-19 ARDS who exhibit hypoinflammatory responses but high viral loads in the lower airways. These clusters may warrant different treatment approaches to improve clinical outcomes.
Collapse
Affiliation(s)
- M Cristina Vazquez Guillamet
- Division of Infectious Diseases, Washington University in St. Louis, St. Louis, MO, USA
- Division of Pulmonary and Critical Care, Washington University in St. Louis, MO, USA
| | | | - Ashraf Rjob
- Department of Internal Medicine, Mountain View Regional Medical Center, Virginia, USA
| | - Daniel Reynolds
- Division of Pulmonary and Critical Care, Washington University in St. Louis, MO, USA
| | - Bijal Parikh
- Department of Pathology, Washington University in St. Louis, MO, USA
| | - Vladimir Despotovic
- Division of Pulmonary and Critical Care, Washington University in St. Louis, MO, USA
| | - Derek E Byers
- Division of Pulmonary and Critical Care, Washington University in St. Louis, MO, USA
| | - Ali H Ellebedy
- Department of Pathology, Washington University in St. Louis, MO, USA
| | - Marin H Kollef
- Division of Pulmonary and Critical Care, Washington University in St. Louis, MO, USA
| | - Philip A Mudd
- Department of Emergency Medicine, Washington University in St. Louis, MO, USA
| |
Collapse
|
47
|
Alladina JW, Giacona FL, Haring AM, Hibbert KA, Medoff BD, Schmidt EP, Thompson T, Maron BA, Alba GA. Circulating Biomarkers of Endothelial Dysfunction Associated With Ventilatory Ratio and Mortality in ARDS Resulting From SARS-CoV-2 Infection Treated With Antiinflammatory Therapies. CHEST CRITICAL CARE 2024; 2:100054. [PMID: 39035722 PMCID: PMC11259037 DOI: 10.1016/j.chstcc.2024.100054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
BACKGROUND The association of plasma biomarkers and clinical outcomes in ARDS resulting from SARS-CoV-2 infection predate the evidence-based use of immunomodulators. RESEARCH QUESTION Which plasma biomarkers are associated with clinical outcomes in patients with ARDS resulting from SARS-CoV-2 infection treated routinely with immunomodulators? STUDY DESIGN AND METHODS We collected plasma from patients with ARDS resulting from SARS-CoV-2 infection within 24 h of admission to the ICU between December 2020 and March 2021 (N = 69). We associated 16 total biomarkers of inflammation (eg, IL-6), coagulation (eg, D-dimer), epithelial injury (eg, surfactant protein D), and endothelial injury (eg, angiopoietin-2) with the primary outcome of in-hospital mortality and secondary outcome of ventilatory ratio (at baseline and day 3). RESULTS Thirty patients (43.5%) died within 60 days. All patients received corticosteroids and 6% also received tocilizumab. Compared with survivors, nonsurvivors demonstrated a higher baseline modified Sequential Organ Failure Assessment score (median, 8.5 [interquartile range (IQR), 7-9] vs 7 [IQR, 5-8]); P = .004), lower Pao2 to Fio2 ratio (median, 153 [IQR, 118-182] vs 184 [IQR, 142-247]; P = .04), and higher ventilatory ratio (median, 2.0 [IQR, 1.9-2.3] vs 1.5 [IQR, 1.4-1.9]; P < .001). No difference was found in inflammatory, coagulation, or epithelial biomarkers between groups. Nonsurvivors showed higher median neural precursor cell expressed, developmentally down-regulated 9 (NEDD9) levels (median, 8.4 ng/mL [IQR, 7.0-11.2 ng/mL] vs 6.9 ng/mL [IQR, 5.5-8.0 ng/mL]; P = .0025), von Willebrand factor domain A2 levels (8.7 ng/mL [IQR, 7.9-9.7 ng/mL] vs 6.5 ng/mL [IQR, 5.7-8.7 ng/mL]; P = .007), angiopoietin-2 levels (9.0 ng/mL [IQR, 7.9-14.1 ng/mL] vs 7.0 ng/mL [IQR, 5.6-10.6 ng/mL]; P = .01), and syndecan-1 levels (15.9 ng/mL [IQR, 14.5-17.5 ng/mL] vs 12.6 ng/mL [IQR, 10.5-16.1 ng/mL]; P = .01). Only NEDD9 level met the adjusted threshold for significance (P < .003). Plasma NEDD9 level was associated with 60-day mortality (adjusted OR, 9.7; 95% CI, 1.6-60.4; P = .015). Syndecan-1 level correlated with both baseline (ρ = 0.4; P = .001) and day 3 ventilatory ratio (ρ = 0.5; P < .001). INTERPRETATION Biomarkers of inflammation, coagulation, and epithelial injury were not associated with clinical outcomes in a small cohort of patients with ARDS uniformly treated with immunomodulators. However, endothelial biomarkers, including plasma NEDD9, were associated with 60-day mortality.
Collapse
Affiliation(s)
- Jehan W Alladina
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Francesca L Giacona
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Alexis M Haring
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Kathryn A Hibbert
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Benjamin D Medoff
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Eric P Schmidt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Taylor Thompson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Bradley A Maron
- Department of Medicine; University of Maryland School of Medicine, Baltimore, University of Maryland-Institute for Health Computing, Bethesda, MD
| | - George A Alba
- Division of Pulmonary and Critical Care Medicine, Bethesda, MD, Department of Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
48
|
Deng X, Tang K, Wang Z, He S, Luo Z. Impacts of Inflammatory Cytokines Variants on Systemic Inflammatory Profile and COVID-19 Severity. J Epidemiol Glob Health 2024; 14:363-378. [PMID: 38376765 PMCID: PMC11176143 DOI: 10.1007/s44197-024-00204-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/30/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Cytokine storm is known to impact the prognosis of coronavirus disease 2019 (COVID-19), since pro-inflammatory cytokine variants are associated with cytokine storm. It is tempting to speculate that pro-inflammatory cytokines variants may impact COVID-19 outcomes by modulating cytokine storm. Here, we verified this hypothesis via a comprehensive analysis. METHODS PubMed, Cochrane Library, Central, CINAHL, and ClinicalTrials.gov were searched until December 15, 2023. Case-control or cohort studies that investigated the impacts of rs1800795 or rs1800629 on COVID-19 susceptibility, severity, mortality, IL-6, TNF-α, or CRP levels were included after an anonymous review by two independent reviewers and consultations of disagreement by a third independent reviewer. RESULTS 47 studies (8305 COVID-19 individuals and 17,846 non-COVID-19 individuals) were analyzed. The rs1800629 A allele (adenine at the -308 position of the promoter was encoded by the A allele) was associated with higher levels of tumor necrosis factor-α (TNF-α) and C-reactive protein (CRP). In contrast, the rs1800795 C allele (cytosine at the -174 position of the promoter was encoded by the C allele) was linked to higher levels of interleukin-6 (IL-6) and CRP. In addition, the A allele of rs1800629 increased the severity and mortality of COVID-19. However, the C allele of rs1800795 only increased COVID-19 susceptibility. CONCLUSIONS rs1800629 and rs1800795 variants of pro-inflammatory cytokines have significant impacts on systemic inflammatory profile and COVID-19 clinical outcomes. rs1800629 may serve as a genetic marker for severe COVID-19.
Collapse
Affiliation(s)
- XueJun Deng
- Department of Cardiology, Suining Central Hospital, Suining, 629000, Sichuan, China
| | - Kai Tang
- Department of Cardiology, Suining Central Hospital, Suining, 629000, Sichuan, China
| | - Zhiqiang Wang
- Orthopedic Center 1 Department of Orthopedic Trauma, Suining Central Hospital, Suining, Sichuan, China.
| | - Suyu He
- The Fourth Department of Digestive Disease Center, Suining Central Hospital, Suining, 629000, Sichuan, China.
| | - Zhi Luo
- Department of Cardiology, Suining Central Hospital, Suining, 629000, Sichuan, China.
| |
Collapse
|
49
|
Katagiri D, Tsukada A, Izumi S, Shimizu Y, Terada-Hirashima J, Uemura Y, Kusaba Y, Takasaki J, Takoi H, Tamura-Nakano M, Hojo M, Takano H, Noiri E, Abe S, Azuma A, Sugiyama H. Blood perfusion with polymyxin B immobilized columns in patients with COVID-19 requiring oxygen therapy. Sci Rep 2024; 14:12550. [PMID: 38822071 PMCID: PMC11143350 DOI: 10.1038/s41598-024-63330-2] [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: 12/15/2023] [Accepted: 05/28/2024] [Indexed: 06/02/2024] Open
Abstract
Extracorporeal blood purification with polymyxin B immobilized fiber column direct hemoperfusion (PMX-DHP), is reported to be effective in treating COVID-19 pneumonitis with oxygen demand. This multicenter prospective study evaluated the efficacy and safety of PMX-DHP in oxygen-requiring patients with COVID-19 admitted between September 28, 2020, and March 31, 2022. The primary endpoint was the percentage of clinical improvement 15 days after treatment. The secondary endpoint was the percentage of worsened disease status. Data from the COVID-19 patient registry were used for the synthetic control group. The improvement rate on Day 15 did not differ between PMX-treated patients and controls; however, the deterioration rate was 0.38 times lower in the PMX-treated group, and the death rates on Day 29 were 0 and 11.1% in the PMX-treated and control groups, respectively. The PMX group showed a 0.73 times higher likelihood for reduced intensive care demand, as 16.7% of PMX-treated patients and 22.8% of controls worsened. After treatment blood oxygenation improved, urinary β2-microglobulin and liver-type fatty acid-binding protein showed significant decreases, and IL-6 decreased once during treatment but did not persist. In this study, PMX treatment effectively prevented the worsening of COVID-19 pathology, accompanied by improved oxygenation. PMX treatment to remove activated cells may effectively improve patient outcomes.
Collapse
Affiliation(s)
- Daisuke Katagiri
- Department of Nephrology, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Akinari Tsukada
- Department of Respiratory Medicine, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinyu Izumi
- Department of Respiratory Medicine, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan.
| | - Yosuke Shimizu
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Junko Terada-Hirashima
- Department of Respiratory Medicine, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yukari Uemura
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yusaku Kusaba
- Department of Respiratory Medicine, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Jin Takasaki
- Department of Respiratory Medicine, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Hiroyuki Takoi
- Department of Respiratory Medicine, Tokyo Medical University Hospital, Tokyo, Japan
| | - Miwa Tamura-Nakano
- Communal Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Masayuki Hojo
- Department of Respiratory Medicine, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Hideki Takano
- Department of Nephrology, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Eisei Noiri
- National Center Biobank Network, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinji Abe
- Department of Respiratory Medicine, Tokyo Medical University Hospital, Tokyo, Japan
| | - Arata Azuma
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
- Mihara General Hospital, Pulmonary Medicine and Clinical Research Center, Saitama, Japan
| | - Haruhito Sugiyama
- Department of Respiratory Medicine, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
50
|
Caixeta RAV, Batista AM, Caetano MW, Palmieri M, Schwab G, Zerbinati RM, Victor ASP, Gallo CDB, Tozetto-Mendoza TR, Junges R, Ortega KL, Costa ALF, Sarmento DJDS, Pallos D, Lindoso JAL, Giannecchini S, Braz-Silva PH. Investigation of Oral Shedding of Torquetenovirus (TTV) in Moderate-to-Severe COVID-19 Hospitalised Patients. Viruses 2024; 16:831. [PMID: 38932124 PMCID: PMC11209259 DOI: 10.3390/v16060831] [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: 03/09/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Torquetenovirus (TTV) is a small DNA virus constituting the human virome. High levels of TTV-DNA have been shown to be associated with immunosuppression and inflammatory chronic disorders. AIM To assess the possible association between the salivary viral load of TTV-DNA in patients hospitalised due to COVID-19 and disease severity. METHODS Saliva samples collected from 176 patients infected with SARS-CoV-2 were used to investigate the presence of SARS-CoV-2 and TTV-DNA by use of real-time RT-PCR. RESULTS The majority of patients were male with severe COVID-19. Presence of SARS-CoV-2 was observed in the saliva of 64.77% of patients, showing TTV-DNA in 55.68% of them. Patients with impaired clinical conditions (p < 0.001), which evolved to death (p = 0.003), showed a higher prevalence of TTV-DNA. The median viral load in patients with severe condition was 4.99 log10 copies/mL, in which those who were discharged and those evolving to death had values of 3.96 log10 copies/mL and 6.27 log10 copies/mL, respectively. A statistically significant association was found between the distribution of TTV-DNA viral load in saliva samples and severity of COVID-19 (p = 0.004) and disease outcomes (p < 0.001). CONCLUSIONS These results indicate that TTV-DNA in saliva could be a useful biomarker of COVID-19 severity and prognosis.
Collapse
Affiliation(s)
- Rafael Antônio Velôso Caixeta
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, Brazil; (R.A.V.C.); (M.W.C.); (M.P.); (C.d.B.G.); (K.L.O.)
| | - Alexandre Mendes Batista
- Laboratory of Virology (LIM-52-HCFMUSP), Institute of Tropical Medicine, University of São Paulo School of Medicine, São Paulo 05403-000, Brazil; (A.M.B.); (G.S.); (R.M.Z.); (A.S.P.V.); (T.R.T.-M.)
| | - Matheus Willian Caetano
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, Brazil; (R.A.V.C.); (M.W.C.); (M.P.); (C.d.B.G.); (K.L.O.)
- Laboratory of Virology (LIM-52-HCFMUSP), Institute of Tropical Medicine, University of São Paulo School of Medicine, São Paulo 05403-000, Brazil; (A.M.B.); (G.S.); (R.M.Z.); (A.S.P.V.); (T.R.T.-M.)
| | - Michelle Palmieri
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, Brazil; (R.A.V.C.); (M.W.C.); (M.P.); (C.d.B.G.); (K.L.O.)
| | - Gabriela Schwab
- Laboratory of Virology (LIM-52-HCFMUSP), Institute of Tropical Medicine, University of São Paulo School of Medicine, São Paulo 05403-000, Brazil; (A.M.B.); (G.S.); (R.M.Z.); (A.S.P.V.); (T.R.T.-M.)
| | - Rodrigo Melim Zerbinati
- Laboratory of Virology (LIM-52-HCFMUSP), Institute of Tropical Medicine, University of São Paulo School of Medicine, São Paulo 05403-000, Brazil; (A.M.B.); (G.S.); (R.M.Z.); (A.S.P.V.); (T.R.T.-M.)
| | - Andressa Silva Pereira Victor
- Laboratory of Virology (LIM-52-HCFMUSP), Institute of Tropical Medicine, University of São Paulo School of Medicine, São Paulo 05403-000, Brazil; (A.M.B.); (G.S.); (R.M.Z.); (A.S.P.V.); (T.R.T.-M.)
| | - Camila de Barros Gallo
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, Brazil; (R.A.V.C.); (M.W.C.); (M.P.); (C.d.B.G.); (K.L.O.)
| | - Tânia Regina Tozetto-Mendoza
- Laboratory of Virology (LIM-52-HCFMUSP), Institute of Tropical Medicine, University of São Paulo School of Medicine, São Paulo 05403-000, Brazil; (A.M.B.); (G.S.); (R.M.Z.); (A.S.P.V.); (T.R.T.-M.)
| | - Roger Junges
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, 0313 Oslo, Norway;
| | - Karem L. Ortega
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, Brazil; (R.A.V.C.); (M.W.C.); (M.P.); (C.d.B.G.); (K.L.O.)
- Oral Medicine, Oral Surgery and Implantology Unit (MedOralRes), Faculty of Medicine and Dentistry, University of Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | | | | | - Débora Pallos
- School of Dentistry, University of Santo Amaro, São Paulo 04743-030, Brazil;
| | - José Angelo Lauletta Lindoso
- Emílio Ribas Institute of Infectious Diseases, São Paulo 01246-900, Brazil;
- Laboratory of Protozoology (LIM-49-HC-FMUSP), Institute of Tropical Medicine, University of São Paulo School of Medicine, São Paulo 01246-903, Brazil
- Department of Infectious Diseases, University of São Paulo School of Medicine, São Paulo 01246-903, Brazil
| | - Simone Giannecchini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Paulo Henrique Braz-Silva
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, Brazil; (R.A.V.C.); (M.W.C.); (M.P.); (C.d.B.G.); (K.L.O.)
- Laboratory of Virology (LIM-52-HCFMUSP), Institute of Tropical Medicine, University of São Paulo School of Medicine, São Paulo 05403-000, Brazil; (A.M.B.); (G.S.); (R.M.Z.); (A.S.P.V.); (T.R.T.-M.)
| |
Collapse
|