1
|
Pius-Sadowska E, Kulig P, Niedźwiedź A, Baumert B, Rogińska D, Łuczkowska K, Sobuś A, Parczewski M, Kawa M, Paczkowska E, Machaliński B. The micro-RNA expression profile predicts the severity of SARS-CoV-2 infection. Sci Rep 2025; 15:17139. [PMID: 40382351 DOI: 10.1038/s41598-025-01229-2] [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: 10/23/2024] [Accepted: 05/05/2025] [Indexed: 05/20/2025] Open
Abstract
Although much is known about the pathophysiology of severe COVID-19, there are still areas that remain to be determined. It is well established that the pivotal molecular event is a hyperinflammatory response also referred to as a cytokine storm. The aim of this retrospective cohort study was to determine miRNAs which might be predictive for the admission to the intensive care unit (ICU). We analyzed blood samples from 210 COVID-19 patients and the control group consisted of 80 healthy individuals. Results revealed the miRNA expression pattern has the potential to predict the severity of COVID-19, reflecting the clinical symptoms of the infection, such as the need for oxygen therapy and concomitant pneumonia. In particular, low expression of miRNAs miR106a-5p, miR17-5p, miR181a-5p, miR191-5p, miR20a-5p and miR451a, especially in the initial phase of the disease, is associated with an unfavorable clinical course of SARS-CoV-2 infection (admission to the ICU).
Collapse
Affiliation(s)
- Ewa Pius-Sadowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Piotr Kulig
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Anna Niedźwiedź
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Bartłomiej Baumert
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Dorota Rogińska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Anna Sobuś
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Miłosz Parczewski
- Department of Infectious, Tropical Diseases and Immune Deficiency, Pomeranian Medical University in Szczecin, Arkońska 4 Street, 71-455, Szczecin, Poland
| | - Miłosz Kawa
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Edyta Paczkowska
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University in Szczecin, Al. Powstańców Wielkopolskich 72, 70-111, Szczecin, Poland.
| |
Collapse
|
2
|
Cui J, Cai W, Zhang L, Wu Y, Huang Y, Zhao W. Decreased monocytic HLA-DR in patients with sepsis: Prediction of diagnosis, severity and prognosis. Clin Biochem 2025; 135:110851. [PMID: 39550023 DOI: 10.1016/j.clinbiochem.2024.110851] [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: 06/26/2024] [Revised: 11/10/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
OBJECTIVE Sepsis is characterized by high incidence and mortality rates, making early recognition and risk stratification critical for preventing delayed treatment and overtreatment. This study investigated the potential of monocytic (m) HLA-DR as a diagnostic and prognostic biomarker of sepsis. METHODS In this prospective study, we collected blood in EDTA-anticoagulated tubes within 48 h from patients diagnosed with sepsis or infection and analyzed the percentage of mHLA-DR in peripheral blood mononuclear cells, C-reactive protein, and procalcitonin within 2 h of collection. We gathered clinical and laboratory data, including sex, age, and comorbidities, calculated the number of dysfunctional organs and sequential organ failure assessment (SOFA) score, and recorded the survival status of patients with sepsis on the 30th day after admission. RESULTS mHLA-DR levels were lower in patients with sepsis (median 46.60 [interquartile range 23.86-66.51]%) than infection (75.44 [52.13-91.50]%). mHLA-DR could distinguish sepsis from infection with an area under the curve (AUC) of 0.724 (95 %CI 0.624-0.824). Decreased mHLA-DR levels have been found in septic patients with shock or secondary infections. mHLA-DR expression decreased with an increasing number of dysfunctional organs and higher SOFA score. In 30-day non-survivors, mHLA-DR levels were 26.94 (12.06-44.45)%, significantly lower than in survivors (55.20 [24.83-72.37]%). mHLA-DR predicted sepsis prognosis with an AUC of 0.750 (95 %CI 0.623-0.877). When the cut-off value was <52.29 %, the sensitivity and specificity of mHLA-DR for prognosis were 100 % and 52.83 %, respectively. The 30-day survival rate of septic patients with mHLA-DR ≥ 52.29 % was 6.798 (95 %CI 2.075-22.27) times higher than that of patients with mHLA-DR < 52.29 %. CONCLUSION mHLA-DR negatively correlates with the severity of sepsis and could be used as a diagnostic and prognostic biomarker for sepsis.
Collapse
Affiliation(s)
- Juanjuan Cui
- Department of Infectious Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Wen Cai
- Department of Infectious Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Li Zhang
- Center of Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Yueyuan Wu
- Center of Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Yan Huang
- Department of Infectious Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Weifeng Zhao
- Department of Infectious Diseases, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
3
|
Mascle O, Dupuis C, Brailova M, Bonnet B, Mirand A, De Beauchene RC, Philipponnet C, Adda M, Calvet L, Cassagnes L, Henquell C, Sapin V, Evrard B, Souweine B. Clustering based on renal and inflammatory admission parameters in critically ill patients admitted to the ICU. PLoS One 2024; 19:e0307938. [PMID: 39485788 PMCID: PMC11530013 DOI: 10.1371/journal.pone.0307938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/15/2024] [Indexed: 11/03/2024] Open
Abstract
INTRODUCTION The COVID-19 pandemic has been associated with significant variability in acute kidney injury (AKI) incidence, leading to concerns regarding patient heterogeneity. The study's primary objective was a cluster analysis, to identify homogeneous subgroups of patients (clusters) using baseline characteristics, including inflammatory biomarkers. The secondary objectives were the comparisons of MAKE-90 and mortality between the different clusters at three months. METHODS This retrospective single-center study was conducted in the Medical Intensive Care Unit of the University Hospital of Clermont-Ferrand, France. Baseline data, clinical and biological characteristics on ICU admission, and outcomes at day 90 were recorded. The primary outcome was the risk of major adverse kidney events at 90 days (MAKE-90). Clusters were determined using hierarchical clustering on principal components approach based on admission characteristics, biomarkers and serum values of immune dysfunction and kidney function. RESULTS It included consecutive adult patients admitted between March 20, 2020 and February 28, 2021 for severe COVID-19. A total of 149 patients were included in the study. Three clusters were identified of which two were fully described (cluster 3 comprising 2 patients). Cluster 1 comprised 122 patients with fewer organ dysfunctions, moderate immune dysfunction, and was associated with reduced mortality and a lower incidence of MAKE-90. Cluster 2 comprised 25 patients with greater disease severity, immune dysfunction, higher levels of suPAR and L-FABP/U Creat, and greater organ support requirement, incidence of AKI, day-90 mortality and MAKE-90. CONCLUSIONS This study identified two clusters of severe COVID-19 patients with distinct biological characteristics and renal event risks. Such clusters may help facilitate the identification of targeted populations for future clinical trials. Also, it may help to understand the significant variability in AKI incidence observed in COVID-19 patients.
Collapse
Affiliation(s)
- Olivier Mascle
- CHU de Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | - Claire Dupuis
- CHU de Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
- Unité de Nutrition Humaine, INRAe, CRNH Auvergne, Université Clermont Auvergne, Clermont Ferrand, France
| | - Marina Brailova
- CHU de Clermont-Ferrand, Service de Biochimie Médicale, Clermont-Ferrand, France
| | - Benjamin Bonnet
- CHU de Clermont-Ferrand, Service d’Immunologie, Clermont-Ferrand, France
- Laboratoire d’Immunologie, ECREIN, UMR1019 UNH, UFR Médecine de Clermont-Ferrand, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Audrey Mirand
- CHU de Clermont-Ferrand, 3IHP, Service de Virologie, Clermont-Ferrand, France
- UMR CNRS 6023 LMGE, Université Clermont Auvergne, Clermont-Ferrand, France
| | | | | | - Mireille Adda
- CHU de Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | - Laure Calvet
- CHU de Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | - Lucie Cassagnes
- CHU de Clermont-Ferrand, Service de Radiologie, Clermont-Ferrand, France
| | - Cécile Henquell
- CHU de Clermont-Ferrand, 3IHP, Service de Virologie, Clermont-Ferrand, France
- UMR CNRS 6023 LMGE, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Vincent Sapin
- CHU de Clermont-Ferrand, Service de Biochimie Médicale, Clermont-Ferrand, France
| | - Bertrand Evrard
- CHU de Clermont-Ferrand, Service d’Immunologie, Clermont-Ferrand, France
- Laboratoire d’Immunologie, ECREIN, UMR1019 UNH, UFR Médecine de Clermont-Ferrand, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Bertrand Souweine
- CHU de Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
- UMR CNRS 6023 LMGE, Université Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
4
|
Krisht AAH, Grapin K, de Beauchene RC, Bonnet B, Cassagnes L, Evrard B, Adda M, Souweine B, Dupuis C. SARS-CoV2 pneumonia patients admitted to the ICU: Analysis according to clinical and biological parameters and the extent of lung parenchymal lesions on chest CT scan, a monocentric observational study. PLoS One 2024; 19:e0308014. [PMID: 39298399 PMCID: PMC11412649 DOI: 10.1371/journal.pone.0308014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/16/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND CT-scan and inflammatory and coagulation biomarkers could help in prognostication of COVID-19 in patients on ICU admission. OBJECTIVE The objectives of this study were to measure the prognostic value of the extent of lung parenchymal lesions on computed tomography (CT) and of several coagulation and inflammatory biomarkers, and to explore the characteristics of the patients depending on the extent of lung parenchymal lesions. DESIGN Retrospective monocentric observational study achieved on a dataset collected prospectively. SETTING Medical ICU of the university hospital of Clermont-Ferrand, France. PATIENTS All consecutive adult patients aged ≥18 years admitted between 20 March, 2020 and 31 August, 2021 for COVID-19 pneumonia. INTERVENTIONS Characteristics at baseline and during ICU stay, and outcomes at day 60 were recorded. The extent of lung parenchyma lesions observed on the chest CT performed on admission was established by artificial intelligence software. MEASUREMENTS Several clinical characteristics and laboratory features were collected on admission including plasma interleukin-6, HLA-DR monocytic-expression rate (mHLA-DR), and the extent of lung parenchymal lesions. Factors associated with day-60 mortality were investigated by uni- and multivariate survival analyses. RESULTS 270 patients were included. Inflammation biomarkers including the levels of neutrophils, CRP, ferritin and Il10 were the indices the most associated with the severity of the extent of the lung lesions. Patients with more extensive lung parenchymal lesions (≥ 75%) on admission had higher CRP serum levels. The extent of lung parenchymal lesions was associated with a decrease in the PaO2/FiO2 ratio(p<0.01), fewer ventilatory-free days (p = 0.03), and a higher death rate at day 60(p = 0.01). Extent of the lesion of more than 75% was independently associated with day-60 mortality (aHR = 1.72[1.06; 2.78], p = 0.03). The prediction of death at day 60 was improved when considering simultaneously biological and radiological markers obtained on ICU admission (AUC = 0.78). CONCLUSIONS The extent of lung parenchyma lesions on CT was associated with inflammation, and the combination of coagulation and inflammatory biomarkers and the extent of the lesions predicted the poorest outcomes.
Collapse
Affiliation(s)
- Abed al Hadi Krisht
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | - Kévin Grapin
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | | | - Benjamin Bonnet
- CHU Clermont-Ferrand, Service d’Immunologie, Clermont-Ferrand, France
- Université Clermont Auvergne, Laboratoire d’Immunologie, ECREIN, UMR1019 UNH, UFR Médecine de Clermont-Ferrand, Clermont-Ferrand, France
| | - Lucie Cassagnes
- CHU Clermont-Ferrand, Service de Radiologie, Clermont-Ferrand, France
- Université Clermont Auvergne, Unité de Nutrition Humaine, INRAe, CRNH Auvergne, Clermont Ferrand, France
| | - Bertrand Evrard
- CHU Clermont-Ferrand, Service d’Immunologie, Clermont-Ferrand, France
- Université Clermont Auvergne, Laboratoire d’Immunologie, ECREIN, UMR1019 UNH, UFR Médecine de Clermont-Ferrand, Clermont-Ferrand, France
| | - Mireille Adda
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
| | - Bertrand Souweine
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
- Université Clermont Auvergne, CNRS, LMGE, Clermont-Ferrand, France
| | - Claire Dupuis
- CHU Clermont-Ferrand, Service de Médecine Intensive et Réanimation, Clermont-Ferrand, France
- Université Clermont Auvergne, Unité de Nutrition Humaine, INRAe, CRNH Auvergne, Clermont Ferrand, France
| |
Collapse
|
5
|
Hawwari I, Rossnagel L, Rosero N, Maasewerd S, Vasconcelos MB, Jentzsch M, Demczuk A, Teichmann LL, Meffert L, Bertheloot D, Ribeiro LS, Kallabis S, Meissner F, Arditi M, Atici AE, Noval Rivas M, Franklin BS. Platelet transcription factors license the pro-inflammatory cytokine response of human monocytes. EMBO Mol Med 2024; 16:1901-1929. [PMID: 38977927 PMCID: PMC11319489 DOI: 10.1038/s44321-024-00093-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/10/2024] Open
Abstract
In humans, blood Classical CD14+ monocytes contribute to host defense by secreting large amounts of pro-inflammatory cytokines. Their aberrant activity causes hyper-inflammation and life-threatening cytokine storms, while dysfunctional monocytes are associated with 'immunoparalysis', a state of immune hypo responsiveness and reduced pro-inflammatory gene expression, predisposing individuals to opportunistic infections. Understanding how monocyte functions are regulated is critical to prevent these harmful outcomes. We reveal platelets' vital role in the pro-inflammatory cytokine responses of human monocytes. Naturally low platelet counts in patients with immune thrombocytopenia or removal of platelets from healthy monocytes result in monocyte immunoparalysis, marked by impaired cytokine response to immune challenge and weakened host defense transcriptional programs. Remarkably, supplementing monocytes with fresh platelets reverses these conditions. We discovered that platelets serve as reservoirs of key cytokine transcription regulators, such as NF-κB and MAPK p38, and pinpointed the enrichment of platelet NF-κB2 in human monocytes by proteomics. Platelets proportionally restore impaired cytokine production in human monocytes lacking MAPK p38α, NF-κB p65, and NF-κB2. We uncovered a vesicle-mediated platelet-monocyte-propagation of inflammatory transcription regulators, positioning platelets as central checkpoints in monocyte inflammation.
Collapse
Affiliation(s)
- Ibrahim Hawwari
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany.
| | - Lukas Rossnagel
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Nathalia Rosero
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Salie Maasewerd
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | | | - Marius Jentzsch
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Agnieszka Demczuk
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lino L Teichmann
- Department of Medicine III, University Hospital Bonn, Bonn, Germany
| | - Lisa Meffert
- Department of Medicine III, University Hospital Bonn, Bonn, Germany
| | - Damien Bertheloot
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Lucas S Ribeiro
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Sebastian Kallabis
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Felix Meissner
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Moshe Arditi
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Asli E Atici
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children's, Cedars Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Bernardo S Franklin
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany.
| |
Collapse
|
6
|
Monneret G, Voirin N, Richard JC, Cour M, Rimmelé T, Garnier L, Yonis H, Coudereau R, Gossez M, Malcus C, Wallet F, Delignette MC, Dailler F, Buisson M, Argaud L, Lukaszewicz AC, Venet F. Monitoring monocyte HLA-DR expression and CD4 + T lymphocyte count in dexamethasone-treated severe COVID-19 patients. Ann Intensive Care 2024; 14:76. [PMID: 38762684 PMCID: PMC11102415 DOI: 10.1186/s13613-024-01310-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/12/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND A 10-day dexamethasone regimen has emerged as the internationally adopted standard-of-care for severe COVID-19 patients. However, the immune response triggered by SARS-CoV-2 infection remains a complex and dynamic phenomenon, leading to various immune profiles and trajectories. The immune status of severe COVID-19 patients following complete dexamethasone treatment has yet to be thoroughly documented. RESULTS To analyze monocyte HLA-DR expression (mHLA-DR) and CD4 + T lymphocyte count (CD4) in critically ill COVID-19 patients after a dexamethasone course and evaluate their association with 28-day ICU mortality, adult COVID-19 patients (n = 176) with an ICU length of stay of at least 10 days and under dexamethasone treatment were included. Associations between each biomarker value (or in combination) measured at day 10 after ICU admission and 28-day mortality in ICU were evaluated. At day 10, the majority of patients presented decreased values of both parameters. A significant association between low mHLA-DR and 28-day mortality was observed. This association remained significant in a multivariate analysis including age, comorbidities or pre-existing immunosuppression (adjusted Hazard ratio (aHR) = 2.86 [1.30-6.32], p = 0.009). Similar results were obtained with decreased CD4 + T cell count (aHR = 2.10 [1.09-4.04], p = 0.027). When combining these biomarkers, patients with both decreased mHLA-DR and low CD4 presented with an independent and significant elevated risk of 28-day mortality (i.e., 60%, aHR = 4.83 (1.72-13.57), p = 0.001). CONCLUSIONS By using standardized immunomonitoring tools available in clinical practice, it is possible to identify a subgroup of patients at high risk of mortality at the end of a 10-day dexamethasone treatment. This emphasizes the significance of integrating immune monitoring into the surveillance of intensive care patients in order to guide further immumodulation approaches.
Collapse
Affiliation(s)
- Guillaume Monneret
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France.
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France.
| | | | - Jean-Christophe Richard
- Medical Intensive Care Department, Hospices Civils de Lyon, Croix-Rousse University Hospital, 69004, Lyon, France
| | - Martin Cour
- Medical Intensive Care Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437, Lyon, France
| | - Thomas Rimmelé
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France
- Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437, Lyon, France
| | - Lorna Garnier
- Immunology Laboratory, Hospices Civils de Lyon, Lyon-Sud University Hospital, 69495, Pierre Bénite, France
| | - Hodane Yonis
- Medical Intensive Care Department, Hospices Civils de Lyon, Croix-Rousse University Hospital, 69004, Lyon, France
| | - Remy Coudereau
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France
| | - Morgane Gossez
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude, Bernard-Lyon 1, Lyon, France
| | - Christophe Malcus
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France
| | - Florent Wallet
- Intensive Care Department, Hospices Civils de Lyon, Lyon-Sud University Hospital, 69495, Pierre-Bénite, France
| | - Marie-Charlotte Delignette
- Anesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Croix-Rousse University Hospital, 69004, Lyon, France
| | - Frederic Dailler
- Neurological Anesthesiology and Intensive Care Department, Hospices Civils de Lyon, Pierre Wertheimer Hospital, Lyon, France
| | - Marielle Buisson
- Centre d'Investigation Clinique de Lyon (CIC 1407 Inserm), Hospices Civils de Lyon, Lyon, France
| | - Laurent Argaud
- Medical Intensive Care Department, Hospices Civils de Lyon, Croix-Rousse University Hospital, 69004, Lyon, France
| | - Anne-Claire Lukaszewicz
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression", Joint Research Unit HCL-bioMérieux, (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), 69003, Lyon, France
- Medical Intensive Care Department, Hospices Civils de Lyon, Edouard Herriot Hospital, 69437, Lyon, France
| | - Fabienne Venet
- Immunology Laboratory, Hospices Civils de Lyon, Edouard Herriot Hospital, 5 Place d'Arsonval, 69437, Lyon, France
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude, Bernard-Lyon 1, Lyon, France
| |
Collapse
|
7
|
Zhang Y, Bharathi V, Dokoshi T, de Anda J, Ursery LT, Kulkarni NN, Nakamura Y, Chen J, Luo EWC, Wang L, Xu H, Coady A, Zurich R, Lee MW, Matsui T, Lee H, Chan LC, Schepmoes AA, Lipton MS, Zhao R, Adkins JN, Clair GC, Thurlow LR, Schisler JC, Wolfgang MC, Hagan RS, Yeaman MR, Weiss TM, Chen X, Li MMH, Nizet V, Antoniak S, Mackman N, Gallo RL, Wong GCL. Viral afterlife: SARS-CoV-2 as a reservoir of immunomimetic peptides that reassemble into proinflammatory supramolecular complexes. Proc Natl Acad Sci U S A 2024; 121:e2300644120. [PMID: 38306481 PMCID: PMC10861912 DOI: 10.1073/pnas.2300644120] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 10/28/2023] [Indexed: 02/04/2024] Open
Abstract
It is unclear how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leads to the strong but ineffective inflammatory response that characterizes severe Coronavirus disease 2019 (COVID-19), with amplified immune activation in diverse cell types, including cells without angiotensin-converting enzyme 2 receptors necessary for infection. Proteolytic degradation of SARS-CoV-2 virions is a milestone in host viral clearance, but the impact of remnant viral peptide fragments from high viral loads is not known. Here, we examine the inflammatory capacity of fragmented viral components from the perspective of supramolecular self-organization in the infected host environment. Interestingly, a machine learning analysis to SARS-CoV-2 proteome reveals sequence motifs that mimic host antimicrobial peptides (xenoAMPs), especially highly cationic human cathelicidin LL-37 capable of augmenting inflammation. Such xenoAMPs are strongly enriched in SARS-CoV-2 relative to low-pathogenicity coronaviruses. Moreover, xenoAMPs from SARS-CoV-2 but not low-pathogenicity homologs assemble double-stranded RNA (dsRNA) into nanocrystalline complexes with lattice constants commensurate with the steric size of Toll-like receptor (TLR)-3 and therefore capable of multivalent binding. Such complexes amplify cytokine secretion in diverse uninfected cell types in culture (epithelial cells, endothelial cells, keratinocytes, monocytes, and macrophages), similar to cathelicidin's role in rheumatoid arthritis and lupus. The induced transcriptome matches well with the global gene expression pattern in COVID-19, despite using <0.3% of the viral proteome. Delivery of these complexes to uninfected mice boosts plasma interleukin-6 and CXCL1 levels as observed in COVID-19 patients.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA9009
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
- Biomedical Engineering, School of Engineering, Westlake University, Hangzhou, Zhejiang310012, China
| | - Vanthana Bharathi
- University of North Carolina Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Tatsuya Dokoshi
- Department of Dermatology, University of California San Diego, La Jolla, CA92093
| | - Jaime de Anda
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA9009
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
| | - Lauryn Tumey Ursery
- University of North Carolina Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Nikhil N. Kulkarni
- Department of Dermatology, University of California San Diego, La Jolla, CA92093
| | - Yoshiyuki Nakamura
- Department of Dermatology, University of California San Diego, La Jolla, CA92093
| | - Jonathan Chen
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA9009
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
| | - Elizabeth W. C. Luo
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA9009
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
| | - Lamei Wang
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Hua Xu
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Alison Coady
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA92093
| | - Raymond Zurich
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA92093
| | - Michelle W. Lee
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA9009
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
| | - Tsutomu Matsui
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA94025
| | - HongKyu Lee
- Division of Molecular Medicine, Harbor-University of California Los Angeles Medical Center, Los Angeles County, Torrance, CA90502
| | - Liana C. Chan
- Division of Molecular Medicine, Harbor-University of California Los Angeles Medical Center, Los Angeles County, Torrance, CA90502
- Division of Infectious Diseases, Harbor-University of California Los Angeles Medical Center, Los Angeles County, Torrance, CA90502
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Institute for Infection & Immunity, Lundquist Institute for Biomedical Innovation, Harbor-University of California Los Angeles Medical Center, Torrance, CA90502
| | - Athena A. Schepmoes
- Environmental Molecular Science Division, Pacific Northwest National Laboratory, Richland, WA99354
| | - Mary S. Lipton
- Environmental Molecular Science Division, Pacific Northwest National Laboratory, Richland, WA99354
| | - Rui Zhao
- Environmental Molecular Science Division, Pacific Northwest National Laboratory, Richland, WA99354
| | - Joshua N. Adkins
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA99354
| | - Geremy C. Clair
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA99354
| | - Lance R. Thurlow
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Jonathan C. Schisler
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Computational Medicine Program, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Matthew C. Wolfgang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Robert S. Hagan
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Michael R. Yeaman
- Division of Molecular Medicine, Harbor-University of California Los Angeles Medical Center, Los Angeles County, Torrance, CA90502
- Division of Infectious Diseases, Harbor-University of California Los Angeles Medical Center, Los Angeles County, Torrance, CA90502
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Institute for Infection & Immunity, Lundquist Institute for Biomedical Innovation, Harbor-University of California Los Angeles Medical Center, Torrance, CA90502
| | - Thomas M. Weiss
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA94025
| | - Xinhua Chen
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Melody M. H. Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
| | - Victor Nizet
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA92093
| | - Silvio Antoniak
- Department of Pathology and Laboratory Medicine, University of North Carolina Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Nigel Mackman
- University of North Carolina Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Richard L. Gallo
- Department of Dermatology, University of California San Diego, La Jolla, CA92093
| | - Gerard C. L. Wong
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA9009
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
| |
Collapse
|
8
|
Grapin K, De Bauchene R, Bonnet B, Mirand A, Cassagnes L, Calvet L, Thouy F, Bouzgarrou R, Henquell C, Evrard B, Adda M, Souweine B, Dupuis C. Severe Acute Respiratory Syndrome Coronavirus 2 Pneumonia in Critically Ill Patients: A Cluster Analysis According to Baseline Characteristics, Biological Features, and Chest CT Scan on Admission. Crit Care Med 2024; 52:e38-e46. [PMID: 37889095 DOI: 10.1097/ccm.0000000000006105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
BACKGROUND Inconsistent results from COVID-19 studies raise the issue of patient heterogeneity. OBJECTIVE The objective of this study was to identify homogeneous subgroups of patients (clusters) using baseline characteristics including inflammatory biomarkers and the extent of lung parenchymal lesions on CT, and to compare their outcomes. DESIGN Retrospective single-center study. SETTING Medical ICU of the University Hospital of Clermont-Ferrand, France. PATIENTS All consecutive adult patients aged greater than or equal to 18 years, admitted between March 20, 2020, and August 31, 2021, for COVID-19 pneumonia. INTERVENTIONS Characteristics at baseline, during ICU stay, and outcomes at day 60 were recorded. On the chest CT performed at admission the extent of lung parenchyma lesions was established by artificial intelligence software. MEASUREMENTS AND MAIN RESULTS Clusters were determined by hierarchical clustering on principal components using principal component analysis of admission characteristics including plasma interleukin-6, human histocompatibility leukocyte antigen-DR expression rate on blood monocytes (HLA-DR) monocytic-expression rate (mHLA-DR), and the extent of lung parenchymal lesions. Factors associated with day 60 mortality were investigated by univariate survival analysis. Two hundred seventy patients were included. Four clusters were identified and three were fully described. Cluster 1 (obese patients, with moderate hypoxemia, moderate extent of lung parenchymal lesions, no inflammation, and no down-regulation of mHLA-DR) had a better prognosis at day 60 (hazard ratio [HR] = 0.27 [0.15-0.46], p < 0.01), whereas cluster 2 (older patients with comorbidities, moderate extent of lung parenchyma lesions but significant hypoxemia, inflammation, and down-regulation of mHLA-DR) and cluster 3 (patients with severe parenchymal disease, hypoxemia, inflammatory reaction, and down-regulation of mHLA-DR) had an increased risk of mortality (HR = 2.07 [1.37-3.13], p < 0.01 and HR = 1.52 [1-2.32], p = 0.05, respectively). In multivariate analysis, only clusters 1 and 2 were independently associated with day 60 death. CONCLUSIONS Three clusters with distinct characteristics and outcomes were identified. Such clusters could facilitate the identification of targeted populations for the next trials.
Collapse
Affiliation(s)
- Kévin Grapin
- CHU Clermont-Ferrand, Service de Médecine intensive et réanimation, Clermont-Ferrand, France
| | | | - Benjamin Bonnet
- CHU Clermont-Ferrand, Service d'Immunologie, Clermont-Ferrand, France
- Université Clermont Auvergne, Laboratoire d'Immunologie, ECREIN, UMR1019, UNH, UFR Médecine de Clermont-Ferrand, Clermont-Ferrand, France
| | - Audrey Mirand
- CHU Clermont-Ferrand, 3IHP, Service de virologie, Clermont-Ferrand, France
- Université Clermont Auvergne, UMR CNRS 6023, LMGE, Clermont-Ferrand, France
| | - Lucie Cassagnes
- CHU Clermont-Ferrand, Service de Radiologie, Clermont-Ferrand, France
- Université Clermont Auvergne, ASMS, UMR 1019, UNH, INRAe, CRNH Auvergne, Clermont-Ferrand, France
| | - Laure Calvet
- CHU Clermont-Ferrand, Service de Médecine intensive et réanimation, Clermont-Ferrand, France
| | - François Thouy
- CHU Clermont-Ferrand, Service de Médecine intensive et réanimation, Clermont-Ferrand, France
| | - Radhia Bouzgarrou
- CHU Clermont-Ferrand, Service de Médecine intensive et réanimation, Clermont-Ferrand, France
| | - Cécile Henquell
- CHU Clermont-Ferrand, 3IHP, Service de virologie, Clermont-Ferrand, France
- Université Clermont Auvergne, UMR CNRS 6023, LMGE, Clermont-Ferrand, France
| | - Bertrand Evrard
- CHU Clermont-Ferrand, Service d'Immunologie, Clermont-Ferrand, France
- Université Clermont Auvergne, Laboratoire d'Immunologie, ECREIN, UMR1019, UNH, UFR Médecine de Clermont-Ferrand, Clermont-Ferrand, France
| | - Mireille Adda
- CHU Clermont-Ferrand, Service de Médecine intensive et réanimation, Clermont-Ferrand, France
| | - Bertrand Souweine
- CHU Clermont-Ferrand, Service de Médecine intensive et réanimation, Clermont-Ferrand, France
- Université Clermont Auvergne, UMR CNRS 6023, LMGE, Clermont-Ferrand, France
| | - Claire Dupuis
- CHU Clermont-Ferrand, Service de Médecine intensive et réanimation, Clermont-Ferrand, France
- Université Clermont Auvergne, ASMS, UMR 1019, UNH, INRAe, CRNH Auvergne, Clermont-Ferrand, France
| |
Collapse
|
9
|
Ling-Hu T, Simons LM, Dean TJ, Rios-Guzman E, Caputo MT, Alisoltani A, Qi C, Malczynski M, Blanke T, Jennings LJ, Ison MG, Achenbach CJ, Larkin PM, Kaul KL, Lorenzo-Redondo R, Ozer EA, Hultquist JF. Integration of individualized and population-level molecular epidemiology data to model COVID-19 outcomes. Cell Rep Med 2024; 5:101361. [PMID: 38232695 PMCID: PMC10829796 DOI: 10.1016/j.xcrm.2023.101361] [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: 12/02/2022] [Revised: 08/07/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with enhanced transmissibility and immune escape have emerged periodically throughout the coronavirus disease 2019 (COVID-19) pandemic, but the impact of these variants on disease severity has remained unclear. In this single-center, retrospective cohort study, we examined the association between SARS-CoV-2 clade and patient outcome over a two-year period in Chicago, Illinois. Between March 2020 and March 2022, 14,252 residual diagnostic specimens were collected from SARS-CoV-2-positive inpatients and outpatients alongside linked clinical and demographic metadata, of which 2,114 were processed for viral whole-genome sequencing. When controlling for patient demographics and vaccination status, several viral clades were associated with risk for hospitalization, but this association was negated by the inclusion of population-level confounders, including case count, sampling bias, and shifting standards of care. These data highlight the importance of integrating non-virological factors into disease severity and outcome models for the accurate assessment of patient risk.
Collapse
Affiliation(s)
- Ted Ling-Hu
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Lacy M Simons
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Taylor J Dean
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Estefany Rios-Guzman
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Matthew T Caputo
- Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Arghavan Alisoltani
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Chao Qi
- Clinical Microbiology Laboratory, Department of Pathology, Northwestern Memorial Hospital, Chicago, IL 60611, USA
| | - Michael Malczynski
- Clinical Microbiology Laboratory, Department of Pathology, Northwestern Memorial Hospital, Chicago, IL 60611, USA
| | - Timothy Blanke
- Diagnostic Molecular Biology Laboratory, Northwestern Memorial Hospital, Chicago, IL 60611, USA
| | - Lawrence J Jennings
- Clinical Microbiology Laboratory, Department of Pathology, Northwestern Memorial Hospital, Chicago, IL 60611, USA
| | - Michael G Ison
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Chad J Achenbach
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Havey Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Paige M Larkin
- Department of Molecular Microbiology, Northshore University HealthSystem, Evanston, IL 60201, USA
| | - Karen L Kaul
- Department of Pathology, Northshore University HealthSystem, Evanston, IL 60201, USA
| | - Ramon Lorenzo-Redondo
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Egon A Ozer
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA
| | - Judd F Hultquist
- Department of Medicine, Division of Infectious Diseases, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pathogen Genomics and Microbial Evolution, Northwestern University Havey Institute for Global Health, Chicago, IL 60611, USA.
| |
Collapse
|
10
|
de Nooijer AH, Pickkers P, Netea MG, Kox M. Inflammatory biomarkers to predict the prognosis of acute bacterial and viral infections. J Crit Care 2023; 78:154360. [PMID: 37343422 DOI: 10.1016/j.jcrc.2023.154360] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/07/2023] [Indexed: 06/23/2023]
Abstract
Mortality in acute infections is mostly associated with sepsis, defined as 'life-threatening organ dysfunction caused by a dysregulated host response to infection'. It remains challenging to identify the patients with increased mortality risk due to the high heterogeneity in the dysregulated host immune response and disease progression. Biomarkers reflecting different pathways involved in the inflammatory response might improve prediction of mortality risk (prognostic enrichment) among patients with acute infections by reducing heterogeneity of the host response, as well as suggest novel strategies for patient stratification and treatment (predictive enrichment) through precision medicine approaches. The predictive value of inflammatory biomarkers has been extensively investigated in bacterial infections and the recent COVID-19 pandemic caused an increased interest in inflammatory biomarkers in this viral infection. However, limited research investigated whether the prognostic potential of these biomarkers differs between bacterial and viral infections. In this narrative review, we provide an overview of the value of various inflammatory biomarkers for the prediction of mortality in bacterial and viral infections.
Collapse
Affiliation(s)
- Aline H de Nooijer
- Department of Internal Medicine, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands; Department of Intensive Care Medicine, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands; Radboud University Medical Center for Infectious Diseases, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands; Radboud University Medical Center for Infectious Diseases, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands; Radboud University Medical Center for Infectious Diseases, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands; Department of Immunology and Metabolism, Life & Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands; Radboud University Medical Center for Infectious Diseases, Radboud University Medical Center, 6500 HB Nijmegen, the Netherlands.
| |
Collapse
|
11
|
Andrejkovits ÁV, Huțanu A, Susányi EJ, Negrea V, Văsieșiu AM. The Prognostic Utility of Cytokines in Hospitalized COVID-19 Patients. J Crit Care Med (Targu Mures) 2023; 9:208-217. [PMID: 37969879 PMCID: PMC10644278 DOI: 10.2478/jccm-2023-0025] [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: 07/08/2023] [Accepted: 09/14/2023] [Indexed: 11/17/2023] Open
Abstract
Introduction The severity of COVID-19 relies on several factors, but the overproduction of pro-inflammatory cytokines remains a central mechanism. The aim of this study was to investigate the predictive utility of interleukin (IL)-6, IL-8, IL-10, IL-12, tumor necrosis factor alpha (TNF-α), and interferon gamma (IFN-γ) measurement in patients with COVID-19. Material and Methods We prospectively enrolled 181 adult patients with COVID-19 admitted to the 1st Infectious Disease County Hospital Târgu Mureș from December 2020 to September 2021. Serum cytokine levels were measured and correlated with disease severity, need for oxygen therapy, intensive care unit (ICU) transfer, and outcome. Results We found significantly higher serum levels of IL-6, IL-8, and IL-10 in patients with severe COVID-19 and in those with a fatal outcome. The logistic regression analysis showed a significant predictive value for IL-8 regarding disease severity, and for IL6 and IL-10 regarding ICU transfer and fatal outcome. Conclusions Serum levels of IL-6, IL-8, and IL-10 were significantly increased in patients with COVID-19, but their predictive value regarding disease severity and the need for oxygen therapy was poor. We found IL-6 and IL-10 to have a good predictive performance regarding ICU transfer and fatal outcome.
Collapse
Affiliation(s)
- Ákos Vince Andrejkovits
- Doctoral School of Medicine and Pharmacy, I.O.S.U.D., George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Romania
| | - Adina Huțanu
- Department of Laboratory Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Romania
- Center for Advanced Medical and Pharmaceutical Research, Targu Mures, Romania
| | - Ervin József Susányi
- First Infectious Disease Clinic of Targu Mureș, Mureș County Clinical Hospital, Romania
| | - Valentina Negrea
- Doctoral School of Medicine and Pharmacy, I.O.S.U.D., George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Romania
- Department of Infectious Disease, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Romania
| | - Anca Meda Văsieșiu
- First Infectious Disease Clinic of Targu Mureș, Mureș County Clinical Hospital, Romania
- Department of Infectious Disease, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Romania
| |
Collapse
|
12
|
Gaudet A, Kreitmann L, Nseir S. ICU-Acquired Colonization and Infection Related to Multidrug-Resistant Bacteria in COVID-19 Patients: A Narrative Review. Antibiotics (Basel) 2023; 12:1464. [PMID: 37760760 PMCID: PMC10525572 DOI: 10.3390/antibiotics12091464] [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: 08/04/2023] [Revised: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
A large proportion of ICU-acquired infections are related to multidrug-resistant bacteria (MDR). Infections caused by these bacteria are associated with increased mortality, and prolonged duration of mechanical ventilation and ICU stay. The aim of this narrative review is to report on the association between COVID-19 and ICU-acquired colonization or infection related to MDR bacteria. Although a huge amount of literature is available on COVID-19 and MDR bacteria, only a few clinical trials have properly evaluated the association between them using a non-COVID-19 control group and accurate design and statistical methods. The results of these studies suggest that COVID-19 patients are at a similar risk of ICU-acquired MDR colonization compared to non-COVID-19 controls. However, a higher risk of ICU-acquired infection related to MDR bacteria has been reported in several studies, mainly ventilator-associated pneumonia and bloodstream infection. Several potential explanations could be provided for the high incidence of ICU-acquired infections related to MDR. Immunomodulatory treatments, such as corticosteroids, JAK2 inhibitors, and IL-6 receptor antagonist, might play a role in the pathogenesis of these infections. Additionally, a longer stay in the ICU was reported in COVID-19 patients, resulting in higher exposure to well-known risk factors for ICU-acquired MDR infections, such as invasive procedures and antimicrobial treatment. Another possible explanation is the surge during successive COVID-19 waves, with excessive workload and low compliance with preventive measures. Further studies should evaluate the evolution of the incidence of ICU-acquired infections related to MDR bacteria, given the change in COVID-19 patient profiles. A better understanding of the immune status of critically ill COVID-19 patients is required to move to personalized treatment and reduce the risk of ICU-acquired infections. The role of specific preventive measures, such as targeted immunomodulation, should be investigated.
Collapse
Affiliation(s)
- Alexandre Gaudet
- Médecine Intensive Réanimation, CHU de Lille, F-59000 Lille, France;
- CNRS, Inserm U1019-UMR9017-CIIL-Centre d’Infection et d’Immunité de Lille, Institut Pasteur de Lille, CHU Lille, Université de Lille, F-59000 Lille, France
| | - Louis Kreitmann
- Centre for Antimicrobial Optimisation, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London W12 0HS, UK;
- Department of Intensive Care Medicine, Imperial College Healthcare NHS Trust, London NW1 5QH, UK
| | - Saad Nseir
- Médecine Intensive Réanimation, CHU de Lille, F-59000 Lille, France;
- Inserm U1285, Université de Lille, CNRS, UMR 8576-UGSF, F-59000 Lille, France
| |
Collapse
|
13
|
Gałuszka-Bulaga A, Tkacz K, Węglarczyk K, Siedlar M, Baran J. Air pollution induces pyroptosis of human monocytes through activation of inflammasomes and Caspase-3-dependent pathways. J Inflamm (Lond) 2023; 20:26. [PMID: 37563611 PMCID: PMC10416410 DOI: 10.1186/s12950-023-00353-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/17/2023] [Indexed: 08/12/2023] Open
Abstract
According to the World Health Organization (WHO), air pollution is one of the most serious threats for our planet. Despite a growing public awareness of the harmful effects of air pollution on human health, the specific influence of particulate matter (PM) on human immune cells remains poorly understood. In this study, we investigated the effect of PM on peripheral blood monocytes in vitro. Monocytes from healthy donors (HD) were exposed to two types of PM: NIST (SRM 1648a, standard urban particulate matter from the US National Institute for Standards and Technology) and LAP (SRM 1648a with the organic fraction removed). The exposure to PM-induced mitochondrial ROS production followed by the decrease of mitochondrial membrane potential and activation of apoptotic protease activating factor 1 (Apaf-1), Caspase-9, and Caspase-3, leading to the cleavage of Gasdermin E (GSDME), and initiation of pyroptosis. Further analysis showed a simultaneous PM-dependent activation of inflammasomes, including NLRP3 (nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3) and Caspase-1, followed by cleavage of Gasdermin D (GSDMD) and secretion of IL-1β. These observations suggest that PM-treated monocytes die by pyroptosis activated by two parallel signaling pathways, related to the inorganic and organic PM components. The release of IL-1β and expression of danger-associated molecular patterns (DAMPs) by pyroptotic cells further activated the remnant viable monocytes to produce inflammatory cytokines (TNF-α, IL-6, IL-8) and protected them from death induced by the second challenge with PM.In summary, our report shows that PM exposure significantly impacts monocyte function and induces their death by pyroptosis. Our observations indicate that the composition of PM plays a crucial role in this process-the inorganic fraction of PM is responsible for the induction of the Caspase-3-dependent pyroptotic pathway. At the same time, the canonical inflammasome path is activated by the organic components of PM, including LPS (Lipopolysaccharide/endotoxin). PM-induced pyroptosis of human monocytes. Particulate matter (PM) treatment affects monocytes viability already after 15 min of their exposure to NIST or LAP in vitro. The remnant viable monocytes in response to danger-associated molecular patterns (DAMPs) release pro-inflammatory cytokines and activate Th1 and Th17 cells. The mechanism of PM-induced cell death includes the increase of reactive oxygen species (ROS) production followed by collapse of mitochondrial membrane potential (ΔΨm), activation of Apaf-1, Caspase-9 and Caspase-3, leading to activation of Caspase-3-dependent pyroptotic pathway, where Caspase-3 cleaves Gasdermin E (GSDME) to produce a N-terminal fragment responsible for the switch from apoptosis to pyroptosis. At the same time, PM activates the canonical inflammasome pathway, where activated Caspase-1 cleaves the cytosolic Gasdermin D (GSDMD) to produce N-terminal domain allowing IL-1β secretion. As a result, PM-treated monocytes die by pyroptosis activated by two parallel pathways-Caspase-3-dependent pathway related to the inorganic fraction of PM and the canonical inflammasome pathway dependent on the organic components of PM.
Collapse
Affiliation(s)
- Adrianna Gałuszka-Bulaga
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka Street 265, 30-663 Krakow, Poland
| | - Karolina Tkacz
- Department of Clinical Immunology, University Children’s Hospital, Krakow, Poland
| | - Kazimierz Węglarczyk
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka Street 265, 30-663 Krakow, Poland
- Department of Clinical Immunology, University Children’s Hospital, Krakow, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka Street 265, 30-663 Krakow, Poland
- Department of Clinical Immunology, University Children’s Hospital, Krakow, Poland
| | - Jarek Baran
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Wielicka Street 265, 30-663 Krakow, Poland
- Department of Clinical Immunology, University Children’s Hospital, Krakow, Poland
| |
Collapse
|
14
|
Tsilingiris D, Vallianou NG, Karampela I, Christodoulatos GS, Papavasileiou G, Petropoulou D, Magkos F, Dalamaga M. Laboratory Findings and Biomarkers in Long COVID: What Do We Know So Far? Insights into Epidemiology, Pathogenesis, Therapeutic Perspectives and Challenges. Int J Mol Sci 2023; 24:10458. [PMID: 37445634 PMCID: PMC10341908 DOI: 10.3390/ijms241310458] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Long COVID (LC) encompasses a constellation of long-term symptoms experienced by at least 10% of people after the initial SARS-CoV-2 infection, and so far it has affected about 65 million people. The etiology of LC remains unclear; however, many pathophysiological pathways may be involved, including viral persistence; a chronic, low-grade inflammatory response; immune dysregulation and a defective immune response; the reactivation of latent viruses; autoimmunity; persistent endothelial dysfunction and coagulopathy; gut dysbiosis; hormonal and metabolic dysregulation; mitochondrial dysfunction; and autonomic nervous system dysfunction. There are no specific tests for the diagnosis of LC, and clinical features including laboratory findings and biomarkers may not specifically relate to LC. Therefore, it is of paramount importance to develop and validate biomarkers that can be employed for the prediction, diagnosis and prognosis of LC and its therapeutic response, although this effort may be hampered by challenges pertaining to the non-specific nature of the majority of clinical manifestations in the LC spectrum, small sample sizes of relevant studies and other methodological issues. Promising candidate biomarkers that are found in some patients are markers of systemic inflammation, including acute phase proteins, cytokines and chemokines; biomarkers reflecting SARS-CoV-2 persistence, the reactivation of herpesviruses and immune dysregulation; biomarkers of endotheliopathy, coagulation and fibrinolysis; microbiota alterations; diverse proteins and metabolites; hormonal and metabolic biomarkers; and cerebrospinal fluid biomarkers. At present, there are only two reviews summarizing relevant biomarkers; however, they do not cover the entire umbrella of current biomarkers, their link to etiopathogenetic mechanisms or the diagnostic work-up in a comprehensive manner. Herein, we aim to appraise and synopsize the available evidence on the typical laboratory manifestations and candidate biomarkers of LC, their classification based on pathogenetic mechanisms and the main LC symptomatology in the frame of the epidemiological and clinical aspects of the syndrome and furthermore assess limitations and challenges as well as potential implications in candidate therapeutic interventions.
Collapse
Affiliation(s)
- Dimitrios Tsilingiris
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece;
| | - Natalia G. Vallianou
- Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou Street, 10676 Athens, Greece;
| | - Irene Karampela
- 2nd Department of Critical Care, Medical School, University of Athens, Attikon General University Hospital, 1 Rimini Street, 12462 Athens, Greece;
| | | | - Georgios Papavasileiou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (G.P.); (D.P.)
| | - Dimitra Petropoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (G.P.); (D.P.)
| | - Faidon Magkos
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, DK-2200 Frederiksberg, Denmark;
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527 Athens, Greece; (G.P.); (D.P.)
| |
Collapse
|
15
|
De Waard A, Lefebvre L, Textoris J, Payen D. Case Report: Intercurrent infections in COVID-19-induced sustained immunodepression: is interferon gamma a suitable drug? Front Immunol 2023; 14:1183665. [PMID: 37359519 PMCID: PMC10285411 DOI: 10.3389/fimmu.2023.1183665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/15/2023] [Indexed: 06/28/2023] Open
Abstract
Acute immuno-depression syndrome (AIDs) had been observed in many life-threatening conditions leading to the Intensive Care Unit. and is associated with recurrent secondary infections. We report one COVID-19 patient with a severe ARDS, demonstrating acute immunodepression syndrome lasting for several weeks. The occurrence of secondary infections despite long treatment by antibiotics led to combined interferon γ (IFNγ) as reported previously. The response to IFNγ was evaluated by the flowcytometry HLA-DR expression on circulating monocytes, which was repeated from time to time. The severe COVID-19 patients responded well to IFNγ without adverse events.
Collapse
Affiliation(s)
- Aurianne De Waard
- Intensive Care Unit, Centre Hospitalier Intercommunal Aix-Pertuis, Aix en Provence, France
| | - Laurent Lefebvre
- Intensive Care Unit, Centre Hospitalier Intercommunal Aix-Pertuis, Aix en Provence, France
| | - Julien Textoris
- EA7426 “Pathophysiology of Injury-Induced Immunosuppression (PI3)”, Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux, Lyon, France
| | - Didier Payen
- Université Paris 7 Denis Diderot, Unité de Formation et de Recherche (UFR) de Médecine, Paris, France
| |
Collapse
|
16
|
Hawerkamp HC, Dyer AH, Patil ND, McElheron M, O’Dowd N, O’Doherty L, Mhaonaigh AU, George AM, O’Halloran AM, Reddy C, Kenny RA, Little MA, Martin-Loeches I, Bergin C, Kennelly SP, Donnelly SC, Bourke NM, Long A, Sui J, Doherty DG, Conlon N, Cheallaigh CN, Fallon PG. Characterisation of the pro-inflammatory cytokine signature in severe COVID-19. Front Immunol 2023; 14:1170012. [PMID: 37063871 PMCID: PMC10101230 DOI: 10.3389/fimmu.2023.1170012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
Clinical outcomes from infection with SARS-CoV-2, the cause of the COVID-19 pandemic, are remarkably variable ranging from asymptomatic infection to severe pneumonia and death. One of the key drivers of this variability is differing trajectories in the immune response to SARS-CoV-2 infection. Many studies have noted markedly elevated cytokine levels in severe COVID-19, although results vary by cohort, cytokine studied and sensitivity of assay used. We assessed the immune response in acute COVID-19 by measuring 20 inflammatory markers in 118 unvaccinated patients with acute COVID-19 (median age: 70, IQR: 58-79 years; 48.3% female) recruited during the first year of the pandemic and 44 SARS-CoV-2 naïve healthy controls. Acute COVID-19 was associated with marked elevations in nearly all pro-inflammatory markers, whilst eleven markers (namely IL-1β, IL-2, IL-6, IL-10, IL-18, IL-23, IL-33, TNF-α, IP-10, G-CSF and YKL-40) were associated with disease severity. We observed significant correlations between nearly all markers elevated in those infected with SARS-CoV-2 consistent with widespread immune dysregulation. Principal component analysis highlighted a pro-inflammatory cytokine signature (with strongest contributions from IL-1β, IL-2, IL-6, IL-10, IL-33, G-CSF, TNF-α and IP-10) which was independently associated with severe COVID-19 (aOR: 1.40, 1.11-1.76, p=0.005), invasive mechanical ventilation (aOR: 1.61, 1.19-2.20, p=0.001) and mortality (aOR 1.57, 1.06-2.32, p = 0.02). Our findings demonstrate elevated cytokines and widespread immune dysregulation in severe COVID-19, adding further evidence for the role of a pro-inflammatory cytokine signature in severe and critical COVID-19.
Collapse
Affiliation(s)
- Heike C. Hawerkamp
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Adam H. Dyer
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Department of Age-Related Healthcare, Tallaght University Hospital, Dublin, Ireland
- *Correspondence: Adam H. Dyer, ; Padraic G. Fallon,
| | - Neha D. Patil
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Matt McElheron
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Niamh O’Dowd
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Laura O’Doherty
- Department of Infectious Diseases, St James’s Hospital, Dublin, Ireland
| | - Aisling Ui Mhaonaigh
- Trinity Kidney Centre, Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland
| | - Angel M. George
- Trinity Kidney Centre, Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland
| | - Aisling M. O’Halloran
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Conor Reddy
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Rose Anne Kenny
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Mark A. Little
- Trinity Kidney Centre, Trinity Translational Medicine Institute, Trinity College, Dublin, Ireland
| | | | - Colm Bergin
- Department of Infectious Diseases, St James’s Hospital, Dublin, Ireland
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Sean P. Kennelly
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
- Department of Age-Related Healthcare, Tallaght University Hospital, Dublin, Ireland
| | - Seamas C. Donnelly
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Clinical Medicine, Tallaght University Hospital, Dublin, Ireland
| | - Nollaig M. Bourke
- Department of Medical Gerontology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Aideen Long
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Jacklyn Sui
- Department of Immunology, St James’s Hospital, Dublin, Ireland
| | - Derek G. Doherty
- Department of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Niall Conlon
- Department of Immunology, St James’s Hospital, Dublin, Ireland
| | - Cliona Ni Cheallaigh
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Padraic G. Fallon
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- *Correspondence: Adam H. Dyer, ; Padraic G. Fallon,
| |
Collapse
|
17
|
Souquette A, Allen EK, Oshansky CM, Tang L, Wong SS, Jeevan T, Shi L, Pounds S, Elias G, Kuan G, Balmaseda A, Zapata R, Shaw-Saliba K, Damme PV, Tendeloo VV, Dib JC, Ogunjimi B, Webby R, Schultz-Cherry S, Pekosz A, Rothman R, Gordon A, Thomas PG. Integrated Drivers of Basal and Acute Immunity in Diverse Human Populations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.25.534227. [PMID: 36993205 PMCID: PMC10055315 DOI: 10.1101/2023.03.25.534227] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Prior studies have identified genetic, infectious, and biological associations with immune competence and disease severity; however, there have been few integrative analyses of these factors and study populations are often limited in demographic diversity. Utilizing samples from 1,705 individuals in 5 countries, we examined putative determinants of immunity, including: single nucleotide polymorphisms, ancestry informative markers, herpesvirus status, age, and sex. In healthy subjects, we found significant differences in cytokine levels, leukocyte phenotypes, and gene expression. Transcriptional responses also varied by cohort, and the most significant determinant was ancestry. In influenza infected subjects, we found two disease severity immunophenotypes, largely driven by age. Additionally, cytokine regression models show each determinant differentially contributes to acute immune variation, with unique and interactive, location-specific herpesvirus effects. These results provide novel insight into the scope of immune heterogeneity across diverse populations, the integrative effects of factors which drive it, and the consequences for illness outcomes.
Collapse
|
18
|
Parallel Dysregulated Immune Response in Severe Forms of COVID-19 and Bacterial Sepsis via Single-Cell Transcriptome Sequencing. Biomedicines 2023; 11:biomedicines11030778. [PMID: 36979757 PMCID: PMC10045101 DOI: 10.3390/biomedicines11030778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/20/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Critically ill COVID-19 patients start developing single respiratory organ failure that often evolves into multiorgan failure. Understanding the immune mechanisms in severe forms of an infectious disease (either critical COVID-19 or bacterial septic shock) would help to achieve a better understanding of the patient’s clinical trajectories and the success of potential therapies. We hypothesized that a dysregulated immune response manifested by the abnormal activation of innate and adaptive immunity might be present depending on the severity of the clinical presentation in both COVID-19 and bacterial sepsis. We found that critically ill COVID-19 patients demonstrated a different clinical endotype that resulted in an inflammatory dysregulation in mild forms of the disease. Mild cases (COVID-19 and bacterial non severe sepsis) showed significant differences in the expression levels of CD8 naïve T cells, CD4 naïve T cells, and CD4 memory T cells. On the other hand, in the severe forms of infection (critical COVID-19 and bacterial septic shock), patients shared immune patterns with upregulated single-cell transcriptome sequencing at the following levels: B cells, monocyte classical, CD4 and CD8 naïve T cells, and natural killers. In conclusion, we identified significant gene expression differences according to the etiology of the infection (COVID-19 or bacterial sepsis) in the mild forms; however, in the severe forms (critical COVID-19 and bacterial septic shock), patients tended to share some of the same immune profiles related to adaptive and innate immune response. Severe forms of the infections were similar independent of the etiology. Our findings might promote the implementation of co-adjuvant therapies and interventions to avoid the development of severe forms of disease that are associated with high mortality rates worldwide.
Collapse
|
19
|
Limmer A, Engler A, Kattner S, Gregorius J, Pattberg KT, Schulz R, Schwab J, Roth J, Vogl T, Krawczyk A, Witzke O, Zelinskyy G, Dittmer U, Brenner T, Berger MM. Patients with SARS-CoV-2-Induced Viral Sepsis Simultaneously Show Immune Activation, Impaired Immune Function and a Procoagulatory Disease State. Vaccines (Basel) 2023; 11:vaccines11020435. [PMID: 36851312 PMCID: PMC9960366 DOI: 10.3390/vaccines11020435] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/10/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND It is widely accepted that SARS-CoV-2 causes a dysregulation of immune and coagulation processes. In severely affected patients, viral sepsis may result in life endangering multiple organ dysfunction. Furthermore, most therapies for COVID-19 patients target either the immune system or coagulation processes. As the exact mechanism causing SARS-CoV-2-induced morbidity and mortality was unknown, we started an in-depth analysis of immunologic and coagulation processes. METHODS 127 COVID-19 patients were treated at the University Hospital Essen, Germany, between May 2020 and February 2022. Patients were divided according to their maximum COVID-19 WHO ordinal severity score (WHO 0-10) into hospitalized patients with a non-severe course of disease (WHO 4-5, n = 52) and those with a severe course of disease (WHO 6-10, n = 75). Non-infected individuals served as healthy controls (WHO 0, n = 42). Blood was analyzed with respect to cell numbers, clotting factors, as well as pro- and anti-inflammatory mediators in plasma. As functional parameters, phagocytosis and inflammatory responses to LPS and antigen-specific stimulation were determined in monocytes, granulocytes, and T cells using flow cytometry. FINDINGS In the present study, immune and coagulation systems were analyzed simultaneously. Interestingly, many severe COVID-19 patients showed an upregulation of pro-inflammatory mediators and at the same time clear signs of immunosuppression. Furthermore, severe COVID-19 patients not only exhibited a disturbed immune system, but in addition showed a pronounced pro-coagulation phenotype with impaired fibrinolysis. Therefore, our study adds another puzzle piece to the already complex picture of COVID-19 pathology implying that therapies in COVID-19 must be individualized. CONCLUSION Despite years of research, COVID-19 has not been understood completely and still no therapies exist, fitting all requirements and phases of COVID-19 disease. This observation is highly reminiscent to sepsis. Research in sepsis has been going on for decades, while the disease is still not completely understood and therapies fitting all patients are lacking as well. In both septic and COVID-19 patients, immune activation can be accompanied by immune paralysis, complicating therapeutic intervention. Accordingly, therapies that lower immune activation may cause detrimental effects in patients, who are immune paralyzed by viral infections or sepsis. We therefore suggest individualizing therapies and to broaden the spectrum of immunological parameters analyzed before therapy. Only if the immune status of a patient is understood, can a therapeutic intervention be successful.
Collapse
Affiliation(s)
- Andreas Limmer
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
- Department of Pediatric Cardiac Surgery, University Hospital Erlangen, 91054 Erlangen, Germany
- Correspondence:
| | - Andrea Engler
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Simone Kattner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Jonas Gregorius
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Kevin Thomas Pattberg
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Rebecca Schulz
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Jansje Schwab
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Johannes Roth
- Institute of Immunology, University of Münster, 48149 Münster, Germany
| | - Thomas Vogl
- Institute of Immunology, University of Münster, 48149 Münster, Germany
| | - Adalbert Krawczyk
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Gennadiy Zelinskyy
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Ulf Dittmer
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Thorsten Brenner
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Marc Moritz Berger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| |
Collapse
|
20
|
Liu S, Luo W, Szatmary P, Zhang X, Lin JW, Chen L, Liu D, Sutton R, Xia Q, Jin T, Liu T, Huang W. Monocytic HLA-DR Expression in Immune Responses of Acute Pancreatitis and COVID-19. Int J Mol Sci 2023; 24:3246. [PMID: 36834656 PMCID: PMC9964039 DOI: 10.3390/ijms24043246] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Acute pancreatitis is a common gastrointestinal disease with increasing incidence worldwide. COVID-19 is a potentially life-threatening contagious disease spread throughout the world, caused by severe acute respiratory syndrome coronavirus 2. More severe forms of both diseases exhibit commonalities with dysregulated immune responses resulting in amplified inflammation and susceptibility to infection. Human leucocyte antigen (HLA)-DR, expressed on antigen-presenting cells, acts as an indicator of immune function. Research advances have highlighted the predictive values of monocytic HLA-DR (mHLA-DR) expression for disease severity and infectious complications in both acute pancreatitis and COVID-19 patients. While the regulatory mechanism of altered mHLA-DR expression remains unclear, HLA-DR-/low monocytic myeloid-derived suppressor cells are potent drivers of immunosuppression and poor outcomes in these diseases. Future studies with mHLA-DR-guided enrollment or targeted immunotherapy are warranted in more severe cases of patients with acute pancreatitis and COVID-19.
Collapse
Affiliation(s)
- Shiyu Liu
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenjuan Luo
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Peter Szatmary
- Liverpool Pancreatitis Research Group, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3BE, UK
| | - Xiaoying Zhang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing-Wen Lin
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lu Chen
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Dan Liu
- Department of Respiratory and Critical Care Medicine, Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Robert Sutton
- Liverpool Pancreatitis Research Group, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3BE, UK
| | - Qing Xia
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Jin
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tingting Liu
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Huang
- West China Centre of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
21
|
Abstract
Fatigue is defined as a symptom leading to the inability to continue functioning at the expected activity level. It is a highly prevalent symptom, challenging to frame into monodimensional pathophysiological mechanisms. As a result, fatigue is often underestimated in the clinical setting and is wrongly considered an unavoidable consequence of ageing. Several potential mechanisms responsible for fatigue have been proposed, including sleep patterns, autonomic nervous system abnormalities and biological complexity. Inflammation and mitochondrial dysfunction are among the most promising mechanisms through which malnutrition may cause fatigue. Not surprisingly, fatigue is highly prevalent in inflammatory conditions (e.g. COVID-19 infection). The nutritional status may also represent a critical factor in the development and presentation of fatigue, which may mimic the exhaustion of the individual's metabolic reserves. For example, the insufficient dietary intake of energy and proteins may determine the catabolism of body fat and muscles, disrupt the homeostatic balance and cause the onset of fatigue. It is necessary to conduct research on fatigue. By characterising its pathophysiological mechanisms, it will be possible to (1) support the design and development of targeted interventions, (2) improve the quality of life of many persons by acting on the symptom and (3) reduce the direct and indirect costs of a burdening condition typical of advancing age. In the present review, we provide an overview of the role that nutrition may play as a determinant of fatigue in older people, also in the context of the COVID-19 pandemic.
Collapse
|
22
|
Almada L, Angiolini SC, Dho ND, Dutto J, Gazzoni Y, Manzone-Rodríguez C, Marín C, Ponce NE, Arroyo DS, Quiróz JN, Iribarren P, Cerbán FM, Morón G, Amezcua Vesely MC, Cervi L, Chiapello LS, Fozzatti L, Icely PA, Maccioni M, Montes CL, Motrán CC, Rodríguez-Galán MC, Stempin CC, Viano ME, Mena C, Bertone M, Abiega CD, Escudero D, Kahn A, Caeiro JP, Maletto BA, Acosta Rodríguez EV, Gruppi A, Sotomayor CE. Different cytokine and chemokine profiles in hospitalized patients with COVID-19 during the first and second outbreaks from Argentina show no association with clinical comorbidities. Front Immunol 2023; 14:1111797. [PMID: 36817433 PMCID: PMC9929547 DOI: 10.3389/fimmu.2023.1111797] [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: 11/30/2022] [Accepted: 01/09/2023] [Indexed: 02/04/2023] Open
Abstract
Background COVID-19 severity has been linked to an increased production of inflammatory mediators called "cytokine storm". Available data is mainly restricted to the first international outbreak and reports highly variable results. This study compares demographic and clinical features of patients with COVID-19 from Córdoba, Argentina, during the first two waves of the pandemic and analyzes association between comorbidities and disease outcome with the "cytokine storm", offering added value to the field. Methods We investigated serum concentration of thirteen soluble mediators, including cytokines and chemokines, in hospitalized patients with moderate and severe COVID-19, without previous rheumatic and autoimmune diseases, from the central region of Argentina during the first and second infection waves. Samples from healthy controls were also assayed. Clinical and biochemical parameters were collected. Results Comparison between the two first COVID-19 waves in Argentina highlighted that patients recruited during the second wave were younger and showed less concurrent comorbidities than those from the first outbreak. We also recognized particularities in the signatures of systemic cytokines and chemokines in patients from both infection waves. We determined that concurrent pre-existing comorbidities did not have contribution to serum concentration of systemic cytokines and chemokines in COVID-19 patients. We also identified immunological and biochemical parameters associated to inflammation which can be used as prognostic markers. Thus, IL-6 concentration, C reactive protein level and platelet count allowed to discriminate between death and discharge in patients hospitalized with severe COVID-19 only during the first but not the second wave. Conclusions Our data provide information that deepens our understanding of COVID-19 pathogenesis linking demographic features of a COVID-19 cohort with cytokines and chemokines systemic concentration, presence of comorbidities and different disease outcomes. Altogether, our findings provide information not only at local level by delineating inflammatory/anti-inflammatory response of patients but also at international level addressing the impact of comorbidities and the infection wave in the variability of cytokine and chemokine production upon SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Laura Almada
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Sofía Carla Angiolini
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Nicolás Daniel Dho
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Jeremías Dutto
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Yamila Gazzoni
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Clarisa Manzone-Rodríguez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Constanza Marín
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Nicolás Eric Ponce
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Daniela Soledad Arroyo
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Juan Nahuel Quiróz
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Pablo Iribarren
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Fabio Marcelo Cerbán
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Gabriel Morón
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - María Carolina Amezcua Vesely
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Laura Cervi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Laura Silvina Chiapello
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Laura Fozzatti
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Paula Alejandra Icely
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Mariana Maccioni
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Carolina Lucia Montes
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Claudia Cristina Motrán
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - María Cecilia Rodríguez-Galán
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Cinthia Carolina Stempin
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - María Estefanía Viano
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Cristian Mena
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Mariana Bertone
- Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Hospital Privado Universitario de Córdoba, Córdoba, Argentina
| | - Claudio Daniel Abiega
- Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Hospital Privado Universitario de Córdoba, Córdoba, Argentina
| | - Daiana Escudero
- Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Hospital Privado Universitario de Córdoba, Córdoba, Argentina
| | - Adrián Kahn
- Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Hospital Privado Universitario de Córdoba, Córdoba, Argentina
| | - Juan Pablo Caeiro
- Instituto Universitario de Ciencias Biomédicas de Córdoba (IUCBC), Hospital Privado Universitario de Córdoba, Córdoba, Argentina
| | - Belkys Angélica Maletto
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Eva Virginia Acosta Rodríguez
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Adriana Gruppi
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Claudia Elena Sotomayor
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| |
Collapse
|
23
|
Sejdic A, Frische A, Jørgensen CS, Rasmussen LD, Trebbien R, Dungu A, Holler JG, Ostrowski SR, Eriksson R, Søborg C, Nielsen TL, Fischer TK, Lindegaard B, Franck KT, Harboe ZB. High titers of neutralizing SARS-CoV-2 antibodies six months after symptom onset are associated with increased severity in COVID-19 hospitalized patients. Virol J 2023; 20:14. [PMID: 36698135 PMCID: PMC9875770 DOI: 10.1186/s12985-023-01974-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Viral shedding and neutralizing antibody (NAb) dynamics among patients hospitalized with severe coronavirus disease 2019 (COVID-19) and immune correlates of protection have been key questions throughout the pandemic. We investigated the duration of reverse transcriptase-polymerase chain reaction (RT-PCR) positivity, infectious viral shedding and NAb titers as well as the association between NAb titers and disease severity in hospitalized COVID-19 patients in Denmark 2020-2021. MATERIALS AND METHODS Prospective single-center observational cohort study of 47 hospitalized COVID-19 patients. Oropharyngeal swabs were collected at eight time points during the initial 30 days of inclusion. Serum samples were collected after a median time of 7 (IQR 5 - 10), 37 (IQR 35 - 38), 97 (IQR 95 - 100), and 187 (IQR 185 - 190) days after symptom onset. NAb titers were determined by an in-house live virus microneutralization assay. Viral culturing was performed in Vero E6 cells. RESULTS Patients with high disease severity had higher mean log2 NAb titers at day 37 (1.58, 95% CI [0.34 -2.81]), 97 (2.07, 95% CI [0.53-3.62]) and 187 (2.49, 95% CI [0.20- 4.78]) after symptom onset, compared to patients with low disease severity. Peak viral load (0.072, 95% CI [- 0.627 - 0.728]), expressed as log10 SARS-CoV-2 copies/ml, was not associated with disease severity. Virus cultivation attempts were unsuccessful in almost all (60/61) oropharyngeal samples collected shortly after hospital admission. CONCLUSIONS We document an association between high disease severity and high mean NAb titers at days 37, 97 and 187 after symptom onset. However, peak viral load during admission was not associated with disease severity. TRIAL REGISTRATION The study is registered at https://clinicaltrials.gov/ (NCT05274373).
Collapse
Affiliation(s)
- Adin Sejdic
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark.
- Statens Serum Institut, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | - Arnold Dungu
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
| | - Jon G Holler
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Immunology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Robert Eriksson
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
- Department of Infectious Diseases, Karolinska Institutet, Solna, Sweden
| | - Christian Søborg
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
| | - Thyge L Nielsen
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
| | - Thea K Fischer
- Department of Clinical Research, Copenhagen University Hospital - North Zealand, Copenhagen, Denmark
- Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Birgitte Lindegaard
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Zitta Barrella Harboe
- Department of Pulmonary and Infectious Diseases, Copenhagen University Hospital, North Zealand, Hillerød, Denmark
- Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
24
|
Dobrijević Z, Gligorijević N, Šunderić M, Penezić A, Miljuš G, Tomić S, Nedić O. The association of human leucocyte antigen (HLA) alleles with COVID-19 severity: A systematic review and meta-analysis. Rev Med Virol 2023; 33:e2378. [PMID: 35818892 PMCID: PMC9349710 DOI: 10.1002/rmv.2378] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/10/2022] [Accepted: 07/04/2022] [Indexed: 01/28/2023]
Abstract
Due to their pivotal role in orchestrating the immune response, HLA loci were recognized as candidates for genetic association studies related to the severity of COVID-19. Since the findings on the effects of HLA alleles on the outcome of SARS-CoV-2 infection remain inconclusive, we aimed to elucidate the potential involvement of genetic variability within HLA loci in the molecular genetics of COVID-19 by classifying the articles according to different disease severity/outcomes and by conducting a systematic review with meta-analysis. Potentially eligible studies were identified by searching PubMed, Scopus and Web of Science literature databases. A total of 28 studies with 13,073 participants were included in qualitative synthesis, while the results of 19 studies with 10,551 SARS-CoV-2-positive participants were pooled in the meta-analysis. According to the results of quantitative data synthesis, association with COVID-19 severity or with the lethal outcome was determined for the following alleles and allele families: HLA-A*01, HLA-A*03, HLA-A*11, HLA-A*23, HLA-A*31, HLA-A*68, HLA-A*68:02, HLA-B*07:02, HLA-B*14, HLA-B*15, HLA-B*40:02, HLA-B*51:01, HLA-B*53, HLA-B*54, HLA-B*54:01, HLA-C*04, HLA-C*04:01, HLA-C*06, HLA-C*07:02, HLA-DRB1*11, HLA-DRB1*15, HLA-DQB1*03 and HLA-DQB1*06 (assuming either allelic or dominant genetic model). We conclude that alleles of HLA-A, -B, -C, -DRB1 and -DQB1 loci may represent potential biomarkers of COVID-19 severity and/or mortality, which needs to be confirmed in a larger set of studies.
Collapse
Affiliation(s)
- Zorana Dobrijević
- Department for MetabolismUniversity of Belgrade—Institute for the Application of Nuclear EnergyBelgradeSerbia
| | - Nikola Gligorijević
- Department for MetabolismUniversity of Belgrade—Institute for the Application of Nuclear EnergyBelgradeSerbia
| | - Miloš Šunderić
- Department for MetabolismUniversity of Belgrade—Institute for the Application of Nuclear EnergyBelgradeSerbia
| | - Ana Penezić
- Department for MetabolismUniversity of Belgrade—Institute for the Application of Nuclear EnergyBelgradeSerbia
| | - Goran Miljuš
- Department for MetabolismUniversity of Belgrade—Institute for the Application of Nuclear EnergyBelgradeSerbia
| | - Sergej Tomić
- Department for Immunology and ImmunoparasitologyUniversity of Belgrade—Institute for the Application of Nuclear EnergyBelgradeSerbia
| | - Olgica Nedić
- Department for MetabolismUniversity of Belgrade—Institute for the Application of Nuclear EnergyBelgradeSerbia
| |
Collapse
|
25
|
Hypercoagulability in critically ill patients with COVID 19, an observational prospective study. PLoS One 2022; 17:e0277544. [PMID: 36417476 PMCID: PMC9683576 DOI: 10.1371/journal.pone.0277544] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/30/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE COVID 19 is often associated with hypercoagulability and thromboembolic (TE) events. The aim of this study was to assess the characteristics of hypercoagulability and its relationship with new-onset TE events and the composite outcome of need for intubation and/or death in intensive care unit (ICU) patients admitted for COVID. DESIGN Prospective observational study. SETTING Monocentric, intensive care, University Hospital of Clermont Ferrand, France. PATIENTS Patients admitted to intensive care from January 2020 to May 2021 for COVID-19 pneumonia. INTERVENTIONS Standard hemostatic tests and rotational thromboelastometry (ROTEM) were performed on admission and on day 4. Hypercoagulability was defined by at least one of the following criteria: D-dimers > 3000 μg/dL, fibrinogen > 8 g/L, EXTEM CFT below the normal range, EXTEM A5, MCF, Li 60 above the normal range, and EXTEM G-score ((5000 x MCF) / (100-MCF)) ≥ 11 dyne/cm2. MEASUREMENTS AND MAIN RESULTS Of the 133 patients included, 17 (12.7%) developed new-onset TE events, and 59 (44.3%) required intubation and/or died in the ICU. ROTEM was performed in 133 patients on day 1 and in 67 on day 4. Hypercoagulability was present on day 1 in 115 (86.4%) patients. None of the hypercoagulability indices were associated with subsequent new-onset TE events on days 1 and 4 nor with the need for intubation and/or ICU death. Hyperfibrinogenemia > 8g/dL, higher D-dimers and higher EXTEM Li 60 on day 4 were predictive of need for intubation and/or of ICU death. CONCLUSIONS Our study confirmed that most COVID-19 ICU patients have hypercoagulability on admission and almost all on day 4. Hyperfibrinogenemia or fibrinolysis shutdown on day 4 were associated with unfavorable outcome.
Collapse
|
26
|
Messing M, Sekhon MS, Hughes MR, Stukas S, Hoiland RL, Cooper J, Ahmed N, Hamer MS, Li Y, Shin SB, Tung LW, Wellington CL, Sin DD, Leslie KB, McNagny KM. Prognostic peripheral blood biomarkers at ICU admission predict COVID-19 clinical outcomes. Front Immunol 2022; 13:1010216. [DOI: 10.3389/fimmu.2022.1010216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
The COVID-19 pandemic continues to challenge the capacities of hospital ICUs which currently lack the ability to identify prospectively those patients who may require extended management. In this study of 90 ICU COVID-19 patients, we evaluated serum levels of four cytokines (IL-1β, IL-6, IL-10 and TNFα) as well as standard clinical and laboratory measurements. On 42 of these patients (binned into Initial and Replication Cohorts), we further performed CyTOF-based deep immunophenotyping of peripheral blood mononuclear cells with a panel of 38 antibodies. All measurements and patient samples were taken at time of ICU admission and retrospectively linked to patient clinical outcomes through statistical approaches. These analyses resulted in the definition of a new measure of patient clinical outcome: patients who will recover after short ICU stays (< 6 days) and those who will subsequently die or recover after long ICU stays (≥6 days). Based on these clinical outcome categories, we identified blood prognostic biomarkers that, at time of ICU admission, prospectively distinguish, with 91% sensitivity and 91% specificity (positive likelihood ratio 10.1), patients in the two clinical outcome groups. This is achieved through a tiered evaluation of serum IL-10 and targeted immunophenotyping of monocyte subsets, specifically, CD11clow classical monocytes. Both immune biomarkers were consistently elevated ( ≥15 pg/ml and ≥2.7 x107/L for serum IL-10 and CD11clow classical monocytes, respectively) in those patients who will subsequently die or recover after long ICU stays. This highly sensitive and specific prognostic test could prove useful in guiding clinical resource allocation.
Collapse
|
27
|
Sun X, Gao C, Zhao K, Yang Y, Rassadkina Y, Fajnzylber J, Regan J, Li JZ, Lichterfeld M, Yu XG. Immune-profiling of SARS-CoV-2 viremic patients reveals dysregulated innate immune responses. Front Immunol 2022; 13:984553. [PMID: 36439166 PMCID: PMC9682031 DOI: 10.3389/fimmu.2022.984553] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 09/01/2022] [Indexed: 09/08/2024] Open
Abstract
SARS-CoV-2 plasma viremia has been associated with severe disease and death in COVID-19. However, the effects of viremia on immune responses in blood cells remain unclear. The current study comprehensively examined transcriptional signatures of PBMCs involving T cells, B cells, NK cells, monocytes, myeloid dendritic cells (mDCs), and plasmacytoid dendritic cells (pDCs) respectively, from three different groups including individuals with moderate (nM), or severe disease with (vS) or without (nS) detectable plasma viral load. Whole transcriptome analysis demonstrated that all seven immune cell subsets were associated with disease severity regardless of cell type. Supervised clustering analysis demonstrated that mDCs and pDCs gene signatures could distinguish disease severity. Notably, transcriptional signatures of the vS group were enriched in pathways related to DNA repair, E2F targets, and G2M checkpoints; in contrast, transcriptional signatures of the nM group were enriched in interferon responses. Moreover, we observed an impaired induction of interferon responses accompanied by imbalanced cell-intrinsic immune sensing and an excessive inflammatory response in patients with severe disease (nS and vS). In sum, our study provides detailed insights into the systemic immune response to SARS-CoV-2 infection and reveals profound alterations in seven major immune cells in COVID-19 patients.
Collapse
Affiliation(s)
- Xiaoming Sun
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, United States
| | - Ce Gao
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, United States
- Infectious Disease Division, Massachusetts General Hospital, Boston, MA, United States
| | - Ke Zhao
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Yanhui Yang
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | | | - Jesse Fajnzylber
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, United States
| | - James Regan
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, United States
| | - Jonathan Z. Li
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, United States
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, United States
- Infectious Disease Division, Massachusetts General Hospital, Boston, MA, United States
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, United States
| | - Xu G. Yu
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, United States
- Infectious Disease Division, Massachusetts General Hospital, Boston, MA, United States
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, MA, United States
| |
Collapse
|
28
|
Noor R. How do the severe acute respiratory coronavirus 2 (SARS-CoV-2) and its variants escape the host protective immunity and mediate pathogenesis? BULLETIN OF THE NATIONAL RESEARCH CENTRE 2022; 46:255. [PMID: 36254244 PMCID: PMC9556142 DOI: 10.1186/s42269-022-00945-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/06/2022] [Indexed: 05/10/2023]
Abstract
Background To protect the global population from the ongoing COVID-19 pandemic caused by the severe acute respiratory β-coronavirus 2 (SARS-CoV-2), a number of vaccines are currently being used in three dosages (i.e., along with the booster dose) to induce the immunity required to combat the SARS-CoV-2 and its variants. So far, several antivirals and the commercial vaccines have been found to evoke the required humoral and cellular immunity within a huge population around world. However, an important aspect to consider is the avoidance mechanism of the host protective immunity by SARS-CoV-2 variants. Main body of the abstract Indeed, such an immune escape strategy has been noticed previously in case of SARS-CoV-1 and the Middle East Respiratory Syndrome coronavirus (MERS-CoV). Regarding the SARS-CoV-2 variants, the most important aspect on vaccine development is to determine whether the vaccine is actually capable to elicit the immune response or not, especially the viral spike (S) protein. Short conclusion Present review thus focused on such elicitation of immunity as well as pondered to the avoidance of host immunity by the SARS-CoV-2 Wuhan strain and its variants.
Collapse
Affiliation(s)
- Rashed Noor
- Department of Life Sciences (DLS), School of Environment and Life Sciences (SELS), Independent University, Bangladesh (IUB), Plot 16, Block B, Aftabuddin Ahmed Road, Bashundhara, Dhaka 1229 Bangladesh
| |
Collapse
|
29
|
Özbay Kurt FG, Lepper A, Gerhards C, Roemer M, Lasser S, Arkhypov I, Bitsch R, Bugert P, Altevogt P, Gouttefangeas C, Neumaier M, Utikal J, Umansky V. Booster dose of mRNA vaccine augments waning T cell and antibody responses against SARS-CoV-2. Front Immunol 2022; 13:1012526. [PMID: 36311732 PMCID: PMC9597683 DOI: 10.3389/fimmu.2022.1012526] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/26/2022] [Indexed: 12/03/2022] Open
Abstract
A gradual decay in humoral and cellular immune responses over time upon SAR1S-CoV-2 vaccination may cause a lack of protective immunity. We conducted a longitudinal analysis of antibodies, T cells, and monocytes in 25 participants vaccinated with mRNA or ChAdOx1-S up to 12 weeks after the 3rd (booster) dose with mRNA vaccine. We observed a substantial increase in antibodies and CD8 T cells specific for the spike protein of SARS-CoV-2 after vaccination. Moreover, vaccination induced activated T cells expressing CD69, CD137 and producing IFN-γ and TNF-α. Virus-specific CD8 T cells showed predominantly memory phenotype. Although the level of antibodies and frequency of virus-specific T cells reduced 4-6 months after the 2nd dose, they were augmented after the 3rd dose followed by a decrease later. Importantly, T cells generated after the 3rd vaccination were also reactive against Omicron variant, indicated by a similar level of IFN-γ production after stimulation with Omicron peptides. Breakthrough infection in participants vaccinated with two doses induced more SARS-CoV-2-specific T cells than the booster vaccination. We found an upregulation of PD-L1 expression on monocytes but no accumulation of myeloid cells with MDSC-like immunosuppressive phenotype after the vaccination. Our results indicate that the 3rd vaccination fosters antibody and T cell immune response independently from vaccine type used for the first two injections. However, such immune response is attenuated over time, suggesting thereby the need for further vaccinations.
Collapse
Affiliation(s)
- Feyza Gül Özbay Kurt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Alisa Lepper
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Catharina Gerhards
- Institute for Clinical Chemistry, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Mathis Roemer
- Institute for Clinical Chemistry, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Samantha Lasser
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ihor Arkhypov
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rebekka Bitsch
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Peter Bugert
- German Red Cross Blood Service Baden-Württemberg – Hessen, Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Cécile Gouttefangeas
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Michael Neumaier
- Institute for Clinical Chemistry, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
- DKFZ-Hector Cancer Institute, University Medical Center Mannheim, Mannheim, Germany
- Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
30
|
SARS-CoV-2 pneumonia and bacterial pneumonia patients differ in a second hit immune response model. Sci Rep 2022; 12:15485. [PMID: 36109525 PMCID: PMC9476429 DOI: 10.1038/s41598-022-17368-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 07/25/2022] [Indexed: 11/10/2022] Open
Abstract
Secondary infections have been shown to complicate the clinical course and worsen the outcome of critically ill patients. Severe Coronavirus Disease 2019 (COVID-19) may be accompanied by a pronounced cytokine release, and immune competence of these patients towards most pathogenic antigens remains uncompromised early in the disease. Patients with bacterial sepsis also exhibit excessive cytokine release with systemic hyper-inflammation, however, typically followed by an anti-inflammatory phase, causing immune paralysis. In a second hit immune response model, leukocyte activation capacity of severely ill patients with pneumonia caused by SARS-CoV-2 or by bacteria were compared upon ICU admission and at days 4 and 7 of the ICU stay. Blood cell count and release of the pro-inflammatory cytokines IL-2, IFNγ and TNF were assessed after whole-blood incubation with the potent immune stimulus pokeweed mitogen (PWM). For comparison, patients with bacterial sepsis not originating from pneumonia, and healthy volunteers were included. Lymphopenia and granulocytosis were less pronounced in COVID-19 patients compared to bacterial sepsis patients. After PWM stimulation, COVID-19 patients showed a reduced release of IFNγ, while IL-2 levels were found similar and TNF levels were increased compared to healthy controls. Interestingly, concentrations of all three cytokines were significantly higher in samples from COVID-19 patients compared to samples from patients with bacterial infection. This fundamental difference in immune competence during a second hit between COVID-19 and sepsis patients may have implications for the selection of immune suppressive or enhancing therapies in personalized medicine.
Collapse
|
31
|
Hottz ED, Martins-Gonçalves R, Palhinha L, Azevedo-Quintanilha IG, de Campos MM, Sacramento CQ, Temerozo JR, Soares VC, Dias SSG, Teixeira L, Castro Í, Righy C, Souza TML, Kurtz P, Andrade BB, Nakaya HI, Monteiro RQ, Bozza FA, Bozza PT. Platelet-monocyte interaction amplifies thromboinflammation through tissue factor signaling in COVID-19. Blood Adv 2022; 6:5085-5099. [PMID: 35420680 PMCID: PMC9015715 DOI: 10.1182/bloodadvances.2021006680] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 04/08/2022] [Indexed: 11/20/2022] Open
Abstract
Accumulating evidence into the pathogenesis of COVID-19 highlights a hypercoagulability state with high risk of life-threatening thromboembolic complications. However, the mechanisms of hypercoagulability and their link to hyperinflammation remain poorly understood. Here, we investigate functions and mechanisms of platelet activation and platelet-monocyte interactions in inflammatory amplification during SARS-CoV-2 infection. We used a combination of immunophenotyping, single-cell analysis, functional assays, and pharmacological approaches to gain insights on mechanisms. Critically ill patients with COVID-19 exhibited increased platelet-monocyte aggregates formation. We identified a subset of inflammatory monocytes presenting high CD16 and low HLA-DR expression as the subset mainly interacting with platelets during severe COVID-19. Single-cell RNA-sequencing analysis indicated enhanced fibrinogen receptor Mac-1 in monocytes from patients with severe COVID-19. Monocytes from patients with severe COVID-19 displayed increased platelet binding and hyperresponsiveness to P-selectin and fibrinogen with respect to tumor necrosis factor-α and interleukin-1β secretion. Platelets were able to orchestrate monocyte responses driving tissue factor (TF) expression, inflammatory activation, and inflammatory cytokines secretion in SARS-CoV-2 infection. Platelet-monocyte interactions ex vivo and in SARS-CoV-2 infection model in vitro reciprocally activated monocytes and platelets, inducing the heightened secretion of a wide panel of inflammatory mediators. We identified platelet adhesion as a primary signaling mechanism inducing mediator secretion and TF expression, whereas TF signaling played major roles in amplifying inflammation by inducing proinflammatory cytokines, especially tumor necrosis factor-α and interleukin-1β. Our data identify platelet-induced TF expression and activity at the crossroad of coagulation and inflammation in severe COVID-19.
Collapse
Affiliation(s)
- Eugenio D. Hottz
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Laboratory of Immunothrombosis, Department of Biochemistry, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Brazil
| | - Remy Martins-Gonçalves
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Lohanna Palhinha
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Mariana M. de Campos
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Carolina Q. Sacramento
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Jairo R. Temerozo
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Laboratory on Thymus Research, and
- National Institute for Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Vinicius Cardoso Soares
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Immunology and inflammation (IMPPG), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Suelen S. Gomes Dias
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Lívia Teixeira
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Ícaro Castro
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Cassia Righy
- Paulo Niemeyer State Brain Institute, Rio de Janeiro, Brazil
- National Institute of Infectious Disease Evandro Chagas, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Thiago Moreno L. Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Pedro Kurtz
- Paulo Niemeyer State Brain Institute, Rio de Janeiro, Brazil
- D’Or institute for Research and Education, Rio de Janeiro, Brazil
| | - Bruno B. Andrade
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Laboratory of Inflammation and Biomarkers, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil
| | - Helder I. Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil; and
| | - Robson Q. Monteiro
- Institute of Medical Biochemistry Leopoldo de Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Fernando A. Bozza
- National Institute of Infectious Disease Evandro Chagas, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- D’Or institute for Research and Education, Rio de Janeiro, Brazil
| | - Patrícia T. Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Bulka CM, Enggasser AE, Fry RC. Epigenetics at the Intersection of COVID-19 Risk and Environmental Chemical Exposures. Curr Environ Health Rep 2022; 9:477-489. [PMID: 35648356 PMCID: PMC9157479 DOI: 10.1007/s40572-022-00353-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW Several environmental contaminants have been implicated as contributors to COVID-19 susceptibility and severity. Immunomodulation and epigenetic regulation have been hypothesized as mediators of this relationship, but the precise underlying molecular mechanisms are not well-characterized. This review examines the evidence for epigenetic modification at the intersection of COVID-19 and environmental chemical exposures. RECENT FINDINGS Numerous environmental contaminants including air pollutants, toxic metal(loid)s, per- and polyfluorinated substances, and endocrine disrupting chemicals are hypothesized to increase susceptibility to the SARS-CoV-2 virus and the risk of severe COVID-19, but few studies currently exist. Drawing on evidence that many environmental chemicals alter the epigenetic regulation of key immunity genes and pathways, we discuss how exposures likely perturb host antiviral responses. Specific mechanisms vary by contaminant but include general immunomodulation as well as regulation of viral entry and recognition, inflammation, and immunologic memory pathways, among others. Associations between environmental contaminants and COVID-19 are likely mediated, in part, by epigenetic regulation of key immune pathways involved in the host response to SARS-CoV-2.
Collapse
Affiliation(s)
- Catherine M Bulka
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam E Enggasser
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina at Chapel Hill, 166A Rosenau Hall, CB #7431, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
33
|
Rocco JM, Laghetti P, Di Stefano M, Sereti I, Ortega-Villa A, Wang J, Rupert A, Chironna M, Ye's P, Liu X, Anderson MV, Burbelo PB, Fiore JR, Saracino A, Lisco A. Impact of Innate Immunity, Endothelial Damage, and Metabolic Biomarkers on COVID-19 Severity and Mortality. Open Forum Infect Dis 2022; 9:ofac427. [PMID: 36111172 PMCID: PMC9452087 DOI: 10.1093/ofid/ofac427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/18/2022] [Indexed: 01/24/2023] Open
Abstract
In this study, abnormal levels of myeloid activation, endothelial damage, and innate immune markers were associated with severe coronavirus disease 2019 (COVID-19), while higher levels of metabolic biomarkers (irisin, leptin) demonstrated a protective effect. These data support a model for COVID-19 immunopathogenesis linking robust inflammation and endothelial damage in metabolically predisposed individuals.
Collapse
Affiliation(s)
- Joseph M Rocco
- Correspondence: J. Rocco, MD, National Institute of Allergy & Infectious Diseases, National Institutes of Health, 10 Center Dr, Building. 10 Room 11B17, Bethesda, MD 20892 ()
| | | | | | | | - Ana Ortega-Villa
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jing Wang
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Adam Rupert
- Leidos Biomedical Research, Inc., Frederick, Maryland, USA
| | - Maria Chironna
- Hygiene Section, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Peiying Ye's
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Xiangdong Liu
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Megan V Anderson
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter B Burbelo
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | |
Collapse
|
34
|
Fan Q, Shi J, Yang Y, Tang G, Jiang M, Li J, Tang J, Li L, Wen X, Zhang L, Deng X, Wang Y, Lan Y, Li L, Peng P, Tong Y, Lu H, Yan L, Liu Y, Cai S, Li Y, Mo X, Li M, Deng X, Hu Z, Yu H, Hu F, Liu J, Tang X, Li F. Clinical characteristics and immune profile alterations in vaccinated individuals with breakthrough Delta SARS-CoV-2 infections. Nat Commun 2022; 13:3979. [PMID: 35810174 PMCID: PMC9271076 DOI: 10.1038/s41467-022-31693-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/29/2022] [Indexed: 11/25/2022] Open
Abstract
Despite timely immunization programs, and efficacious vaccines conveying protection against SARS-CoV-2 infection, breakthrough infections in vaccinated individuals have been reported. The Delta variant of concern (VOC) outbreak in Guangzhou resulted in local transmission in vaccinated and non-vaccinated residents, providing a unique opportunity to study the protective effects of the inactivated vaccines in breakthrough infection. Here, we find that the 2-dose vaccinated group has similar peak viral titers and comparable speeds of viral RNA clearance to the non-vaccinated group but accelerated viral suppression in the middle course of the disease. We quantitatively demonstrate that peak viral pneumonia is significantly mitigated in the 2-dose vaccine group (median 0.298%) compared with the non-vaccinated (5.77%) and 1-dose vaccine (3.34%) groups. Pneumonia absorbance is approximately 6 days ahead in the 2-dose group (median 10 days) than in the non-vaccinated group (16 days) (p = 0.003). We also observe reduced cytokine inflammation and markedly undisturbed gene transcription profiles of peripheral blood mononuclear cells (PBMCs) in the 2-dose group. In short, our study demonstrates that prior vaccination substantially restrains pneumonia development, reduces cytokine storms, and facilitates clinical recovery. SARS-CoV-2 breakthrough infections in vaccinated individuals are a public health concern. Here, the authors analyse the clinical characteristics and profile immune alterations among vaccinated and non-vaccinated residents with Delta SARS-CoV-2 infection in Guangzhou.
Collapse
Affiliation(s)
- Qinghong Fan
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jingrong Shi
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yanhong Yang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Guofang Tang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mengling Jiang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiaojiao Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jingyan Tang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lu Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xueliang Wen
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lieguang Zhang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xizi Deng
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yaping Wang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yun Lan
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Liya Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ping Peng
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuwei Tong
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huan Lu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lili Yan
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ying Liu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shuijiang Cai
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yueping Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiaoneng Mo
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Meiyu Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xilong Deng
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhongwei Hu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Haisheng Yu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Fengyu Hu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jinxin Liu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Xiaoping Tang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China. .,Guangzhou Laboratory, Bio-Island, Guangzhou, China.
| | - Feng Li
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China. .,Guangzhou Laboratory, Bio-Island, Guangzhou, China.
| |
Collapse
|
35
|
Gasparyan AY, Kitas GD. Hyperinflammation due to COVID-19 and the Targeted Use of Interleukin-1 Inhibitors. Mediterr J Rheumatol 2022; 33:173-175. [PMID: 36128214 PMCID: PMC9450206 DOI: 10.31138/mjr.33.2.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 12/02/2022] Open
Affiliation(s)
- Armen Yuri Gasparyan
- Departments of Rheumatology and Research and Development, Dudley Group NHS Foundation Trust (Teaching Trust of the University of Birmingham, UK), Russells Hall Hospital, Dudley, West Midlands, United Kingdom
| | - George D. Kitas
- Departments of Rheumatology and Research and Development, Dudley Group NHS Foundation Trust (Teaching Trust of the University of Birmingham, UK), Russells Hall Hospital, Dudley, West Midlands, United Kingdom
- Centre for Epidemiology versus Arthritis, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
36
|
Rodríguez-Hernández MÁ, Carneros D, Núñez-Núñez M, Coca R, Baena R, López-Ruiz GM, Cano-Serrano ME, Martínez-Tellería A, Fuentes-López A, Praena-Fernandez JM, Garbers C, Hernández-Quero J, García F, Rose-John S, Bustos M. Identification of IL-6 Signalling Components as Predictors of Severity and Outcome in COVID-19. Front Immunol 2022; 13:891456. [PMID: 35634332 PMCID: PMC9137400 DOI: 10.3389/fimmu.2022.891456] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
IL-6 is one of the major mediators of the hyper-inflammatory responses with complex biological functions as it can signal via different modes of action. IL-6 by classical signalling has anti-inflammatory and antibacterial activities, while trans-signalling mediates pro-inflammatory effects. The net biological effect of IL-6 is established by multiple factors beyond its absolute concentration. Here, we assess the relationship between IL-6 signalling variables [IL-6, soluble IL-6R (sIL-6R) and soluble gp130 (sgp130)] and outcomes in a cohort of 366 COVID-19 patients. The potential trans-signalling was evaluated by a ratio between the pro-inflammatory binary IL-6:sIL-6R complex and the inactive ternary IL-6:sIL-6R:sgp130 complex (binary/ternary complex) and the fold molar excess of sgp130 over sIL-6R (FME). Our data provide new evidence that high levels of IL-6, sIL-6R, sgp130, binary/ternary complex ratio, and low FME are independent predictors of COVID-19 severity in survivor patients (without death), and the combination of IL-6 + sIL-6R + sgp130 exhibited the most robust classification capacity. Conversely, in a subgroup of patients with a very poor prognosis, we found that high levels of IL-6 and low levels of sIL-6R, sgp130, and binary/ternary complex ratio were predictors of death. In this context, the highest predictive capacity corresponded to the combined analysis of IL-6 + FME + lymphopenia + creatinine. Herein, we present IL-6 signalling variables as a helpful tool for the early identification and stratification of patients with clear implications for treatment and clinical decision-making.
Collapse
Affiliation(s)
- María Ángeles Rodríguez-Hernández
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBIS), Spanish National Research Council (CSIC) - University of Seville (US) - Virgen del Rocio University Hospital (HUVR), Seville, Spain
- *Correspondence: María Ángeles Rodríguez-Hernández, ; Matilde Bustos,
| | - David Carneros
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBIS), Spanish National Research Council (CSIC) - University of Seville (US) - Virgen del Rocio University Hospital (HUVR), Seville, Spain
| | - María Núñez-Núñez
- Department of Pharmacy, San Cecilio University Hospital, Granada, Spain
- Infectious Disease Unit, San Cecilio University Hospital, Granada, Spain
- Biosanitary Research Institute of Granada (ibs.Granada), Granada, Spain
| | - Ramón Coca
- Department of Clinical Analysis, Virgen de las Nieves University Hospital, Granada, Spain
| | - Rosario Baena
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Pamplona, Spain
| | - Gema M. López-Ruiz
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBIS), Spanish National Research Council (CSIC) - University of Seville (US) - Virgen del Rocio University Hospital (HUVR), Seville, Spain
| | | | | | - Ana Fuentes-López
- Department of Microbiology, San Cecilio University Hospital, Granada, Spain
| | | | - Christoph Garbers
- Department of Pathology, Medical Faculty, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | | | - Federico García
- Biosanitary Research Institute of Granada (ibs.Granada), Granada, Spain
- Department of Microbiology, San Cecilio University Hospital, Granada, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), ISCIII, Madrid, Spain
| | | | - Matilde Bustos
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBIS), Spanish National Research Council (CSIC) - University of Seville (US) - Virgen del Rocio University Hospital (HUVR), Seville, Spain
- *Correspondence: María Ángeles Rodríguez-Hernández, ; Matilde Bustos,
| |
Collapse
|
37
|
Syenina A, Gan ES, Toh JZN, de Alwis R, Lin LZ, Tham CYL, Yee JX, Leong YS, Sam H, Cheong C, Teh YE, Wee ILE, Ng DHL, Chan KR, Sim JXY, Kalimuddin S, Ong EZ, Low JG, Ooi EE. Adverse effects following anti-COVID-19 vaccination with mRNA-based BNT162b2 are alleviated by altering the route of administration and correlate with baseline enrichment of T and NK cell genes. PLoS Biol 2022; 20:e3001643. [PMID: 35639676 PMCID: PMC9154185 DOI: 10.1371/journal.pbio.3001643] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/22/2022] [Indexed: 12/14/2022] Open
Abstract
Ensuring high vaccination and even booster vaccination coverage is critical in preventing severe Coronavirus Disease 2019 (COVID-19). Among the various COVID-19 vaccines currently in use, the mRNA vaccines have shown remarkable effectiveness. However, systemic adverse events (AEs), such as postvaccination fatigue, are prevalent following mRNA vaccination, and the underpinnings of which are not understood. Herein, we found that higher baseline expression of genes related to T and NK cell exhaustion and suppression were positively correlated with the development of moderately severe fatigue after Pfizer-BioNTech BNT162b2 vaccination; increased expression of genes associated with T and NK cell exhaustion and suppression reacted to vaccination were associated with greater levels of innate immune activation at 1 day postvaccination. We further found, in a mouse model, that altering the route of vaccination from intramuscular (i.m.) to subcutaneous (s.c.) could lessen the pro-inflammatory response and correspondingly the extent of systemic AEs; the humoral immune response to BNT162b2 vaccination was not compromised. Instead, it is possible that the s.c. route could improve cytotoxic CD8 T-cell responses to BNT162b2 vaccination. Our findings thus provide a glimpse of the molecular basis of postvaccination fatigue from mRNA vaccination and suggest a readily translatable solution to minimize systemic AEs.
Collapse
Affiliation(s)
- Ayesa Syenina
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Esther S. Gan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Justin Z. N. Toh
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore
- School of Life Sciences, Nanyang Polytechnic, Singapore
| | - Ruklanthi de Alwis
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Lowell Z. Lin
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Christine Y. L. Tham
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Jia Xin Yee
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Yan Shan Leong
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Huizhen Sam
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Charlene Cheong
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Yii Ean Teh
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Ian L. E. Wee
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Dorothy H. L. Ng
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Kuan Rong Chan
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Jean X. Y. Sim
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Shirin Kalimuddin
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Eugenia Z. Ong
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore
| | - Jenny G. Low
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore
- Department of Infectious Diseases, Singapore General Hospital, Singapore
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore
- Department of Infectious Diseases, Singapore General Hospital, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
| |
Collapse
|
38
|
Hammad R, Kotb HG, Eldesoky GAR, Mosaad AM, El-Nasser AM, Abd El Hakam FELZ, Eldesoky NAR, Mashaal A, Farhoud H. Utility of Monocyte Expression of HLA-DR versus T Lymphocyte Frequency in the Assessment of COVID-19 Outcome. Int J Gen Med 2022; 15:5073-5087. [PMID: 35615469 PMCID: PMC9126655 DOI: 10.2147/ijgm.s359690] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/09/2022] [Indexed: 12/26/2022] Open
Abstract
Background Dysregulated immunity is a hallmark of SARS-CoV-2 infection. Immune suppression is indicated by low monocyte expression of human leukocyte antigen D-related (mHLA-DR). T cells are important antiviral cells. We aimed to assess the role of mHLA-DR and T lymphocyte frequency in predicting COVID-19 severity. Patients and Methods This cross-sectional study enrolled 97 SARS-CoV-2 positive patients, including mild to moderate (n = 49) and severe cases admitted to intensive care unit (ICU) (n = 48). These ICU cases were further subdivided into survivors (n = 35) and non-survivors (n = 13). Results Severe cases had a significant decrease in the mHLA-DR mean fluorescence intensity (MFI) and T lymphocyte percentage compared to mild to moderate cases (P<0.001). Non-survivors had a lower T lymphocyte percentage (P=0.004) than survivors. The mHLA-DR MFI and T lymphocyte percentage correlated with oxygen saturation (r=0.632, P<0.001) and (r=0.669, P<0.001), respectively. According to the ROC curves, mHLA-DR MFI, at a cutoff of 143 and an AUC of 0.9, is a reliable biomarker for distinguishing severe COVID-19 cases, with 89.6% sensitivity and 81.6% specificity, while T lymphocyte frequency had 81.3% sensitivity and 81.6% specificity at a cutoff of 54.4% and an AUC of 0.9. The T lymphocyte percentage as a predictor of ICU survival at a cutoff of 38.995% exhibited 100% sensitivity and 57.1% specificity. According to multivariate regression analysis, reduced mHLA-DR MFI and T lymphocyte percentage are independent predictors of COVID-19 severity (OR = 0.976, 95% CI: 0.955–0.997, P = 0.025) and (OR = 0.849, 95% CI: 0.741–0.972, P = 0.018), respectively. Conclusion Reduced mHLA-DR expression and T-lymphocyte percentage are independent predictors of COVID-19 severity. Oxygen saturation percentage is correlated with mHLA-DR MFI and T lymphocyte frequency. The T lymphocyte frequency is a proposed predictor of COVID-19 survival in ICU admitted patients.
Collapse
Affiliation(s)
- Reham Hammad
- Clinical Pathology Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo, Egypt
| | - Hend G Kotb
- Internal Medicine Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo, Egypt
| | - Gehan Abdel-Rahman Eldesoky
- Anesthesia, Intensive Care and Pain Management Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo, Egypt
| | - Alshaimaa Mohamed Mosaad
- Hepatogastroenterology and Infectious Diseases Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo, Egypt
| | - Asmaa M El-Nasser
- Medical Microbiology & Immunology Department, Faculty of Medicine (for Girls), Al-Azhar University, Cairo, Egypt
| | | | - Noha Abdel-Rahman Eldesoky
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (for Girls), Al-Azhar University, Cairo, Egypt
| | - Alya Mashaal
- Immunology, Zoology & Entomology Department, Faculty of Science (for Girls), Al-Azhar University, Cairo, Egypt
- Correspondence: Alya Mashaal, Immunology, Zoology & Entomology Department, Faculty of Science (for Girls), Al-Azhar University, Cairo, Egypt, Email
| | - Hesham Farhoud
- Orthopedic Surgery Department, Former Dean of Faculty of Medicine (for Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
39
|
Salvia miltiorrhiza Bunge as a Potential Natural Compound against COVID-19. Cells 2022; 11:cells11081311. [PMID: 35455990 PMCID: PMC9028742 DOI: 10.3390/cells11081311] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 02/06/2023] Open
Abstract
Salvia miltiorrhiza Bunge, commonly called danshen, is widely used in traditional Chinese medicine for its cardiovascular and neuroprotective effects, which include antioxidative, anti-inflammatory, and antifibrotic properties. The purpose of this study was to evaluate the preclinical potential of S. miltiorrhiza extracts for the treatment of COVID-19. First, the impact of the extract on the binding between SARS-CoV-2 and the cellular ACE2 receptors was assessed using atomic force microscopy (AFM), showing a significant reduction in binding by the extract at concentrations in the µg/mL range. Second, the interference of this extract with the inflammatory response of blood mononuclear cells (PBMCs) was determined, demonstrating potent inhibitory properties in the same concentration range on pro-inflammatory cytokine release and interference with the activation of NFκB signaling. Together, these in vitro data demonstrate the potential of S. miltiorrhiza against COVID-19, consisting first of the blockade of the binding of SARS-CoV-2 to the ACE2 receptor and the mitigation of the inflammatory response from leukocytes by interfering with NFκB signaling. This dataset prompts the launch of a clinical trial to address in vivo the clinical benefits of this promising agent.
Collapse
|
40
|
Venet F, Gossez M, Bidar F, Bodinier M, Coudereau R, Lukaszewicz AC, Tardiveau C, Brengel-Pesce K, Cheynet V, Cazalis MA, Pescarmona R, Garnier L, Ortillon M, Buisson M, Bouscambert-Duchamp M, Morfin-Sherpa F, Casalegno JS, Conti F, Rimmelé T, Argaud L, Cour M, Saadatian-Elahi M, Henaff L, Vanhems P, Monneret G. T cell response against SARS-CoV-2 persists after one year in patients surviving severe COVID-19. EBioMedicine 2022; 78:103967. [PMID: 35349827 PMCID: PMC8957405 DOI: 10.1016/j.ebiom.2022.103967] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/10/2022] [Accepted: 03/10/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In critically ill COVID-19 patients, the initial response to SARS-CoV-2 infection is characterized by major immune dysfunctions. The capacity of these severe patients to mount a robust and persistent SARS-CoV-2 specific T cell response despite the presence of severe immune alterations during the ICU stay is unknown. METHODS Critically ill COVID-19 patients were sampled five times during the ICU stay and 9 and 13 months afterwards. Immune monitoring included counts of lymphocyte subpopulations, HLA-DR expression on monocytes, plasma IL-6 and IL-10 concentrations, anti-SARS-CoV-2 IgG levels and T cell proliferation in response to three SARS-CoV-2 antigens. FINDINGS Despite the presence of major lymphopenia and decreased monocyte HLA-DR expression during the ICU stay, convalescent critically ill COVID-19 patients consistently generated adaptive and humoral immune responses against SARS-CoV-2 maintained for more than one year after hospital discharge. Patients with long hospital stays presented with stronger anti-SARS-CoV-2 specific T cell response but no difference in anti-SARS-CoV2 IgG levels. INTERPRETATION Convalescent critically ill COVID-19 patients consistently generated a memory immune response against SARS-CoV-2 maintained for more than one year after hospital discharge. In recovered individuals, the intensity of SARS-CoV-2 specific T cell response was dependent on length of hospital stay. FUNDING This observational study was supported by funds from the Hospices Civils de Lyon, Fondation HCL, Claude Bernard Lyon 1 University and Région Auvergne Rhône-Alpes and by partial funding by REACTing (Research and ACTion targeting emerging infectious diseases) INSERM, France and a donation from Fondation AnBer (http://fondationanber.fr/).
Collapse
Affiliation(s)
- Fabienne Venet
- Immunology Laboratory, Hôpital E. Herriot - Hospices Civils de Lyon, 5 place d'Arsonval, Lyon 69437 CEDEX 03, France; Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, Lyon, France.
| | - Morgane Gossez
- Immunology Laboratory, Hôpital E. Herriot - Hospices Civils de Lyon, 5 place d'Arsonval, Lyon 69437 CEDEX 03, France; Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, Lyon, France
| | - Frank Bidar
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Lyon 69003, France; Anesthesia and Critical Care Medicine Department, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon 69437, France
| | - Maxime Bodinier
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Lyon 69003, France
| | - Rémy Coudereau
- Immunology Laboratory, Hôpital E. Herriot - Hospices Civils de Lyon, 5 place d'Arsonval, Lyon 69437 CEDEX 03, France; EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Lyon 69003, France
| | - Anne-Claire Lukaszewicz
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Lyon 69003, France; Anesthesia and Critical Care Medicine Department, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon 69437, France
| | - Claire Tardiveau
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Lyon 69003, France
| | - Karen Brengel-Pesce
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Lyon 69003, France
| | - Valérie Cheynet
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Lyon 69003, France
| | - Marie-Angélique Cazalis
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Lyon 69003, France
| | - Rémi Pescarmona
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, Lyon, France; Immunology Laboratory, Lyon-Sud University Hospital - Hospices Civils de Lyon, Pierre-Bénite, France
| | - Lorna Garnier
- Immunology Laboratory, Lyon-Sud University Hospital - Hospices Civils de Lyon, Pierre-Bénite, France
| | - Marine Ortillon
- Immunology Laboratory, Hôpital E. Herriot - Hospices Civils de Lyon, 5 place d'Arsonval, Lyon 69437 CEDEX 03, France
| | - Marielle Buisson
- Centre d'Investigation Clinique de Lyon (CIC 1407 Inserm), Hospices Civils de Lyon, Lyon F-69677, France
| | - Maude Bouscambert-Duchamp
- Virology Laboratory, CNR des virus des infections Respiratoires, Institut des Agents Infectieux, Hospices Civils de Lyon, Hôpital de la Croix Rousse, Lyon, France
| | - Florence Morfin-Sherpa
- Virology Laboratory, CNR des virus des infections Respiratoires, Institut des Agents Infectieux, Hospices Civils de Lyon, Hôpital de la Croix Rousse, Lyon, France
| | - Jean-Sébastien Casalegno
- Virology Laboratory, CNR des virus des infections Respiratoires, Institut des Agents Infectieux, Hospices Civils de Lyon, Hôpital de la Croix Rousse, Lyon, France
| | - Filippo Conti
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Lyon 69003, France
| | - Thomas Rimmelé
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Lyon 69003, France; Anesthesia and Critical Care Medicine Department, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon 69437, France
| | - Laurent Argaud
- Medical intensive Care Department, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon 69437, France
| | - Martin Cour
- Medical intensive Care Department, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon 69437, France
| | - Mitra Saadatian-Elahi
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, Lyon, France; Service Hygiène, Epidémiologie, Infectiovigilance et Prévention, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon 69437, France
| | - Laetitia Henaff
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, Lyon, France; Service Hygiène, Epidémiologie, Infectiovigilance et Prévention, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon 69437, France
| | - Philippe Vanhems
- Centre International de Recherche en Infectiologie (CIRI), Inserm U1111, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Université Claude Bernard-Lyon 1, Lyon, France; Service Hygiène, Epidémiologie, Infectiovigilance et Prévention, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon 69437, France
| | - Guillaume Monneret
- Immunology Laboratory, Hôpital E. Herriot - Hospices Civils de Lyon, 5 place d'Arsonval, Lyon 69437 CEDEX 03, France; EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon - bioMérieux), Joint Research Unit HCL-bioMérieux, Lyon 69003, France
| | | |
Collapse
|
41
|
Fedorchenko Y, Zimba O. CYTOKINES AS POTENTIAL MARKERS OF COVID-19 SEVERITY AND OUTCOMES. CENTRAL ASIAN JOURNAL OF MEDICAL HYPOTHESES AND ETHICS 2022. [DOI: 10.47316/cajmhe.2022.3.1.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The continual propagation of SARS-CoV-2 has changed health care systems globally. Ranging degrees of clinical severity in COVID-19 patients have been noted in numerous literature sources. Cytokines play a crucial role in the development of key immunological processes in COVID-19. SARS-CoV-2 causes imbalance of the immune system and might culminate in cytokine storm and multiple organ involvement. The prevailing role of some special cytokines might serve as indicators of disease severity. Further stratification of patients in the context of specific cytokines can be beneficial for diagnosing disease stages. It can prevent critical states owing to timely diagnosis and targeted therapy. Targeting peculiar cytokines can markedly reduce complications. The aim of this article is to comprehensively overview the role of the main cytokines in COVID-19 pathogenesis and distinguish prognostic factors. Insights into specific cytokine involvement in COVID-19 pathogenesis may open new avenues for diagnosing hyperinflammatory COVID-19, predicting its outcomes and providing individualized cytokine-targeted therapeutic approaches.
Collapse
|
42
|
Silverstein NJ, Wang Y, Manickas-Hill Z, Carbone C, Dauphin A, Boribong BP, Loiselle M, Davis J, Leonard MM, Kuri-Cervantes L, Meyer NJ, Betts MR, Li JZ, Walker BD, Yu XG, Yonker LM, Luban J. Innate lymphoid cells and COVID-19 severity in SARS-CoV-2 infection. eLife 2022; 11:e74681. [PMID: 35275061 PMCID: PMC9038195 DOI: 10.7554/elife.74681] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 03/11/2022] [Indexed: 11/21/2022] Open
Abstract
Background Risk of severe COVID-19 increases with age, is greater in males, and is associated with lymphopenia, but not with higher burden of SARS-CoV-2. It is unknown whether effects of age and sex on abundance of specific lymphoid subsets explain these correlations. Methods Multiple regression was used to determine the relationship between abundance of specific blood lymphoid cell types, age, sex, requirement for hospitalization, duration of hospitalization, and elevation of blood markers of systemic inflammation, in adults hospitalized for severe COVID-19 (n = 40), treated for COVID-19 as outpatients (n = 51), and in uninfected controls (n = 86), as well as in children with COVID-19 (n = 19), recovering from COVID-19 (n = 14), MIS-C (n = 11), recovering from MIS-C (n = 7), and pediatric controls (n = 17). Results This observational study found that the abundance of innate lymphoid cells (ILCs) decreases more than 7-fold over the human lifespan - T cell subsets decrease less than 2-fold - and is lower in males than in females. After accounting for effects of age and sex, ILCs, but not T cells, were lower in adults hospitalized with COVID-19, independent of lymphopenia. Among SARS-CoV-2-infected adults, the abundance of ILCs, but not of T cells, correlated inversely with odds and duration of hospitalization, and with severity of inflammation. ILCs were also uniquely decreased in pediatric COVID-19 and the numbers of these cells did not recover during follow-up. In contrast, children with MIS-C had depletion of both ILCs and T cells, and both cell types increased during follow-up. In both pediatric COVID-19 and MIS-C, ILC abundance correlated inversely with inflammation. Blood ILC mRNA and phenotype tracked closely with ILCs from lung. Importantly, blood ILCs produced amphiregulin, a protein implicated in disease tolerance and tissue homeostasis. Among controls, the percentage of ILCs that produced amphiregulin was higher in females than in males, and people hospitalized with COVID-19 had a lower percentage of ILCs that produced amphiregulin than did controls. Conclusions These results suggest that, by promoting disease tolerance, homeostatic ILCs decrease morbidity and mortality associated with SARS-CoV-2 infection, and that lower ILC abundance contributes to increased COVID-19 severity with age and in males. Funding This work was supported in part by the Massachusetts Consortium for Pathogen Readiness and NIH grants R37AI147868, R01AI148784, F30HD100110, 5K08HL143183.
Collapse
Affiliation(s)
- Noah J Silverstein
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
- Medical Scientist Training Program, University of Massachusetts Medical SchoolWorcesterUnited States
- Massachusetts Consortium on Pathogen ReadinessBostonUnited States
| | - Yetao Wang
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
- Massachusetts Consortium on Pathogen ReadinessBostonUnited States
| | - Zachary Manickas-Hill
- Massachusetts Consortium on Pathogen ReadinessBostonUnited States
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
| | - Claudia Carbone
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Ann Dauphin
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
| | - Brittany P Boribong
- Massachusetts General Hospital, Mucosal Immunology and Biology Research CenterBostonUnited States
- Massachusetts General Hospital, Department of PediatricsBostonUnited States
- Harvard Medical SchoolBostonUnited States
| | - Maggie Loiselle
- Massachusetts General Hospital, Mucosal Immunology and Biology Research CenterBostonUnited States
| | - Jameson Davis
- Massachusetts General Hospital, Mucosal Immunology and Biology Research CenterBostonUnited States
| | - Maureen M Leonard
- Massachusetts General Hospital, Mucosal Immunology and Biology Research CenterBostonUnited States
- Massachusetts General Hospital, Department of PediatricsBostonUnited States
- Harvard Medical SchoolBostonUnited States
| | - Leticia Kuri-Cervantes
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute for Immunology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Nuala J Meyer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Pennsylvania Perelman School of MedicinePhiladelphiaUnited States
| | - Michael R Betts
- Department of Microbiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute for Immunology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Jonathan Z Li
- Massachusetts Consortium on Pathogen ReadinessBostonUnited States
- Department of Medicine, Brigham and Women’s HospitalBostonUnited States
| | - Bruce D Walker
- Massachusetts Consortium on Pathogen ReadinessBostonUnited States
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
- Howard Hughes Medical InstituteChevy ChaseUnited States
- Department of Biology and Institute of Medical Engineering and Science, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Xu G Yu
- Massachusetts Consortium on Pathogen ReadinessBostonUnited States
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
- Department of Medicine, Brigham and Women’s HospitalBostonUnited States
| | - Lael M Yonker
- Massachusetts General Hospital, Mucosal Immunology and Biology Research CenterBostonUnited States
- Massachusetts General Hospital, Department of PediatricsBostonUnited States
- Harvard Medical SchoolBostonUnited States
| | - Jeremy Luban
- Program in Molecular Medicine, University of Massachusetts Medical SchoolWorcesterUnited States
- Massachusetts Consortium on Pathogen ReadinessBostonUnited States
- Ragon Institute of MGH, MIT and HarvardCambridgeUnited States
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Medical SchoolWorcesterUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
| |
Collapse
|
43
|
Affiliation(s)
- Domenico Azzolino
- Department of Clinical and Community Sciences, University of Milan, 20138 Milan, Italy
- Geriatric Unit, IRCCS Istituti Clinici Scientifici Maugeri, Milan, Italy
| | - Matteo Cesari
- Department of Clinical and Community Sciences, University of Milan, 20138 Milan, Italy
- Geriatric Unit, IRCCS Istituti Clinici Scientifici Maugeri, Milan, Italy
| |
Collapse
|
44
|
Giamarellos-Bourboulis EJ. Complex immune deregulation in severe COVID-19: More than a mechanism of pathogenesis. EBioMedicine 2021; 73:103673. [PMID: 34742130 PMCID: PMC8564503 DOI: 10.1016/j.ebiom.2021.103673] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023] Open
Affiliation(s)
- Evangelos J Giamarellos-Bourboulis
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, ATTIKON University Hospital, 1 Rimini Str, Athens 12462, Greece.
| |
Collapse
|