1
|
Abba Moussa D, Vazquez M, Chable-Bessia C, Roux-Portalez V, Tamagnini E, Pedotti M, Simonelli L, Ngo G, Souchard M, Lyonnais S, Chentouf M, Gros N, Marsile-Medun S, Dinter H, Pugnière M, Martineau P, Varani L, Juan M, Calderon H, Naranjo-Gomez M, Pelegrin M. Discovery of a pan anti-SARS-CoV-2 monoclonal antibody with highly efficient infected cell killing capacity for novel immunotherapeutic approaches. Emerg Microbes Infect 2025; 14:2432345. [PMID: 39584380 PMCID: PMC11632933 DOI: 10.1080/22221751.2024.2432345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/24/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024]
Abstract
Unlocking the potential of broadly reactive coronavirus monoclonal antibodies (mAbs) and their derivatives offers a transformative therapeutic avenue against severe COVID-19, especially crucial for safeguarding high-risk populations. Novel mAb-based immunotherapies may help address the reduced efficacy of current vaccines and neutralizing mAbs caused by the emergence of variants of concern (VOCs). Using phage display technology, we discovered a pan-SARS-CoV-2 mAb (C10) that targets a conserved region within the receptor-binding domain (RBD) of the virus. Noteworthy, C10 demonstrates exceptional efficacy in recognizing all assessed VOCs, including recent Omicron variants. While C10 lacks direct neutralization capacity, it efficiently binds to infected lung epithelial cells and induces their lysis via natural killer (NK) cell-mediated antibody-dependent cellular cytotoxicity (ADCC). Building upon this pan-SARS-CoV-2 mAb, we engineered C10-based, Chimeric Antigen Receptor (CAR)-T cells endowed with efficient killing capacity against SARS-CoV-2-infected lung epithelial cells. Notably, NK and CAR-T-cell mediated killing of lung infected cells effectively reduces viral titers. These findings highlight the potential of non-neutralizing mAbs in providing immune protection against emerging infectious diseases. Our work reveals a pan-SARS-CoV-2 mAb effective in targeting infected cells and demonstrates the proof-of-concept for the potential application of CAR-T cell therapy in combating SARS-CoV-2 infections. Furthermore, it holds promise for the development of innovative antibody-based and cell-based therapeutic strategies against severe COVID-19 by expanding the array of therapeutic options available for high-risk populations.Trial registration: ClinicalTrials.gov identifier: NCT04093596.
Collapse
Affiliation(s)
| | - Mario Vazquez
- IDIBAPS, Immunogenetics and Immunotherapy in Autoinflammatory and Immune Responses, Barcelona, Spain
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Vincent Roux-Portalez
- IRCM, University of Montpellier, ICM, INSERM, Montpellier, France
- GenAc, Siric Plateform, INSERM, Montpellier, France
| | - Elia Tamagnini
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Mattia Pedotti
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Luca Simonelli
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Giang Ngo
- IRCM, University of Montpellier, ICM, INSERM, Montpellier, France
- PPM, BioCampus Plateforme de Protéomique de Montpellier CNRS, Montpellier, France
| | - Manon Souchard
- IRMB, University of Montpellier, INSERM, CNRS, Montpellier, France
| | | | - Myriam Chentouf
- IRCM, University of Montpellier, ICM, INSERM, Montpellier, France
- GenAc, Siric Plateform, INSERM, Montpellier, France
| | - Nathalie Gros
- CEMIPAI, University of Montpellier, UAR3725 CNRS, Montpellier, France
| | | | - Heiko Dinter
- IRMB, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Martine Pugnière
- IRCM, University of Montpellier, ICM, INSERM, Montpellier, France
- PPM, BioCampus Plateforme de Protéomique de Montpellier CNRS, Montpellier, France
| | - Pierre Martineau
- IRCM, University of Montpellier, ICM, INSERM, Montpellier, France
- GenAc, Siric Plateform, INSERM, Montpellier, France
| | - Luca Varani
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Manel Juan
- IDIBAPS, Immunogenetics and Immunotherapy in Autoinflammatory and Immune Responses, Barcelona, Spain
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Hugo Calderon
- IDIBAPS, Immunogenetics and Immunotherapy in Autoinflammatory and Immune Responses, Barcelona, Spain
- Department of Immunology, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Mireia Pelegrin
- IRMB, University of Montpellier, INSERM, CNRS, Montpellier, France
| |
Collapse
|
2
|
Song Y, Lu J, Qin P, Chen H, Chen L. Interferon-I modulation and natural products: Unraveling mechanisms and therapeutic potential in severe COVID-19. Cytokine Growth Factor Rev 2025; 82:18-30. [PMID: 39261232 DOI: 10.1016/j.cytogfr.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to pose a significant global public health threat, particularly to older adults, pregnant women, and individuals with underlying chronic conditions. Dysregulated immune responses to SARS-CoV-2 infection are believed to contribute to the progression of COVID-19 in severe cases. Previous studies indicates that a deficiency in type I interferon (IFN-I) immunity accounts for approximately 15 %-20 % of patients with severe pneumonia caused by COVID-19, highlighting the potential therapeutic importance of modulating IFN-I signals. Natural products and their derivatives, due to their structural diversity and novel scaffolds, play a crucial role in drug discovery. Some of these natural products targeting IFN-I have demonstrated applications in infectious diseases and inflammatory conditions. However, the immunomodulatory potential of IFN-I in critical COVID-19 pneumonia and the natural compounds regulating the related signal pathway remain not fully understood. In this review, we offer a comprehensive assessment of the association between IFN-I and severe COVID-19, exploring its mechanisms and integrating information on natural compounds effective for IFN-I regulation. Focusing on the primary targets of IFN-I, we also summarize the regulatory mechanisms of natural products, their impact on IFNs, and their therapeutic roles in viral infections. Collectively, by synthesizing these findings, our goal is to provide a valuable reference for future research and to inspire innovative treatment strategies for COVID-19.
Collapse
Affiliation(s)
- Yuheng Song
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiani Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Pengcheng Qin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Henan University, Kaifeng 475001, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 200032, China
| | - Lili Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
3
|
Zhou F, Guo Y, Li W, Hu Y, Yang L, Fu S, Bao X, Tong H, Ye Y, Ding Z. Tetrastigma hemsleyanum polysaccharide protects against "two-hit" induced severe pneumonia via TLR4/NF-κB signaling pathway. Int J Biol Macromol 2025; 303:140639. [PMID: 39909274 DOI: 10.1016/j.ijbiomac.2025.140639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
Severe pneumonia, frequently accompanied by cytokine storms, stands as a perilous respiratory condition with alarmingly high mortality rates. Tetrastigma hemsleyanum polysaccharide (THP), a pivotal constituent derived from Tetrastigma hemsleyanum Diels et Gilg (TH), has demonstrated efficacy in treating lung inflammation. However, its precise efficacy and underlying mechanisms in the context of severe pneumonia remain elusive. Our research aims to elucidate THP's protective effects in a "two-hit" severe pneumonia model. Our observations indicate that THP administration markedly shields the lungs from injury, reduces pulmonary apoptosis, balances the formation of immune thrombus and alleviates oxidative stress in pneumonia-induced mice. Furthermore, THP significantly decreases the levels of pro-inflammatory cytokines, suggesting its robust anti-inflammatory capabilities. Notably, THP also plays a crucial role in normalizing gut microbiota imbalance, which is vital in the pathogenesis of severe pneumonia. Metabolomic analysis further validates THP's restorative effects on plasma metabolites, indicating its involvement in regulating energy metabolism and immune homeostasis. Mechanistically, THP targets the TLR4/NF-κB signaling pathway, a core mediator of inflammation, thereby dampening the inflammatory cascade. In summary, our findings underscore that THP, through its multifaceted actions targeting inflammation, oxidative stress, immune thrombus formation, gut microbiota regulation, and metabolic modulation, emerges as a promising therapeutic approach for severe pneumonia. This study provides invaluable insights into the potential applications of natural polysaccharides in treating severe pneumonia and highlights the significance of the TLR4/NF-κB pathway in the disease's progression.
Collapse
Affiliation(s)
- Fangmei Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Ying Guo
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Wenxuan Li
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Yiwen Hu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Liu Yang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Siyu Fu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Xiaodan Bao
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Hongbin Tong
- Hangzhou HealthBank Medical Laboratory Co., Ltd., Hangzhou, Zhejiang 310053, China
| | - Yujian Ye
- Department of Dermatology, Third People's Hospital of Hangzhou, Hangzhou, China.
| | - Zhishan Ding
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
4
|
Sha J, Kong G, Fu L, Wang P, Zhang L, Wang T, Song F, Chu Y, Meng M. Impact of Early Administration of Albumin on Mortality Among Severe COVID-19 Patients, China. Infect Drug Resist 2025; 18:1539-1549. [PMID: 40123713 PMCID: PMC11930246 DOI: 10.2147/idr.s510245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/15/2025] [Indexed: 03/25/2025] Open
Abstract
Purpose Hypoalbuminemia is commonly observed in patients with severe Coronavirus Disease 2019 (COVID-19) and is independently associated with adverse outcomes. However, the efficacy of albumin administration on the clinical prognosis of these patients remains uncertain. Patients and Methods This multicenter retrospective study enrolled 458 patients with severe COVID-19 in four medical centers from December 1, 2022, to June 1, 2024. Clinical features and laboratory variables were collected through electronic medical records. The cohorts were divided into two groups: albumin administration and non-albumin administration. Propensity score matching (PSM) was used for minimizing confounding effect. Statistical analyses were conducted to assess the relationship between early albumin administration and 28-day mortality. Results Four hundred and fifty-eight severe COVID-19 cases were included in the study, of which 167 (36.5%) received early albumin administration, while 291 (63.5%) did not. Among these patients, 140 experienced in-hospital mortality and 318 survived. Compared to survivors, non-survivors exhibited significantly lower serum albumin levels (29.1g/L vs.33.8g/L, p < 0.05). In comparison to patients with admission albumin levels ≥30 g/L, those with albumin levels <30 g/L had a significantly higher in-hospital mortality (48.4% vs 21.1%, p < 0.001). Prior to PSM, the albumin administration group demonstrated significantly higher 28-day and in-hospital cumulative survival rates compared to the non-albumin group (both p < 0.001). However, no significant differences were observed between the two groups following PSM (p = 0.21 and p = 0.41, respectively). Conclusion Hypoalbuminemia was correlated with adverse outcomes in severe COVID-19 patients. However, early albumin administration did not reduce 28-day mortality and in-hospital mortality in these patients, and more relative RCTs were required for validation.
Collapse
Affiliation(s)
- Jing Sha
- Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People’s Republic of China
| | - Guiqing Kong
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Lin Fu
- Department of Critical Care Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Peng Wang
- Neurocritical Care Unit, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Lin Zhang
- Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, People’s Republic of China
| | - Tao Wang
- Department of Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong, People’s Republic of China
| | - Fangqiang Song
- Department of Critical Care Medicine, Tengzhou Central People’s Hospital, Tengzhou, Shandong, People’s Republic of China
| | - Yufeng Chu
- Neurocritical Care Unit, Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Mei Meng
- Department of Critical Care Medicine, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, People’s Republic of China
| |
Collapse
|
5
|
Hu Y, Lu Y, Dong J, Xia D, Li J, Wang H, Rao M, Wang C, Tong W. Epidemiological and clinical characteristics of COVID-19 mortality: a retrospective study. Front Med (Lausanne) 2025; 12:1464274. [PMID: 40130249 PMCID: PMC11930819 DOI: 10.3389/fmed.2025.1464274] [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: 08/17/2024] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
Background The global impact of SARS-CoV-2 and its associated coronavirus disease (COVID-19) has necessitated urgent characterization of prognostic biomarkers. This study aimed to delineate the epidemiological and clinical predictors of mortality among hospitalized COVID-19 patients. Methods A retrospective cohort study was conducted on 123 patients with laboratory-confirmed COVID-19 admitted to Huoshenshan Hospital (Wuhan, China) from 1 February 2020 to 30 April 2020. Kaplan-Meier curve and multivariate Cox regression were used to assess the independent factors with survival time. Statistical significance was set at a p-value of <0.05. Results The cohort exhibited a mortality rate of 49.6% (61/123), with the critical clinical type (HR = 7.970, p = 0.009), leukocytosis (HR = 3.408, p = 0.006), and lymphopenia (HR = 0.817, p = 0.038) emerging as independent predictors of reduced survival. Critical-type patients demonstrated significantly elevated inflammatory markers (neutrophils: 10.41 ± 6.23 × 109/L; CRP: 104.47 ± 29.18 mg/L) and coagulopathy (D-dimer: 5.21 ± 2.34 μg/ml) compared to non-critical cases. Deceased patients exhibited pronounced metabolic derangements, including hyperglycemia (9.81 ± 2.07 mmol/L) and hepatic dysfunction (ALP: 174.03 ± 30.13 U/L). Conclusion We revealed the epidemiological and clinical features of different clinical types of SARS-CoV-2 as summarized in this paper. We found that critical type, leukocyte, and lymphocyte are risk factors that affect survival time, which could be an early and helpful marker to improve management of COVID-19 patients.
Collapse
Affiliation(s)
- Yaohua Hu
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - You Lu
- Department of Respiratory Medicine, Shanghai Tenth People’s Hospital, Shanghai, China
| | - Jiagui Dong
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - Delin Xia
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - Jin Li
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - Hong Wang
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - Min Rao
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - Chenxing Wang
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| | - Wanning Tong
- Department of Respiratory and Critical Care Medicine, Naval Medical Center of People’s Liberation Army, Shanghai, China
| |
Collapse
|
6
|
de Oliveira JN, Fernandes CYM, de Godoy SM, Frantine-Silva W, de Souza Cassela PLC, Trigo GL, Lozovoy MAB, Tano ZN, Simão ANC, de Oliveira KB. Association of IL10 gene SNVs rs1800896 (A>G), rs1800871 (C>T), rs1800872 (C>A) and haplotypes with COVID-19 severity and outcome in the Brazilian population. Hum Immunol 2025; 86:111261. [PMID: 39933261 DOI: 10.1016/j.humimm.2025.111261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Elevated concentrations of IL-10 have been detected in coronavirus disease (COVID-19) patients and are a possible disease severity marker. Single nucleotide variants (SNVs) and their haplotypes can be associated with differences in IL-10 levels and with viral disease susceptibility. AIM Evaluate the associations of SNVs and their haplotypes in Brazilian patients with COVID-19 severity and outcome. METHODS In this cross-sectional and case-control study, the patients were selected from the University Hospital of State University of Londrina (HU-UEL) (n = 367) and were subdivided into mild (n = 165), moderate (n = 72) and severe (n = 130) groups. The DNA samples of the participants were subjected to real-time PCR for the detection of rs1800896 (A>G), rs1800871 (C>T) and rs1800872 (C>A) genotypes. The haplotypes were inferred with PHASE v2.1.1. RESULTS The severe cases of COVID-19 were independently associated with the GG genotype (rs1800896) (P = 0.038, OR 2.522, 95 % CI 1.053-6.038) as well as with the GCC haplotype in homozygosity (P = 0.037, OR 2.767, 95 % CI 1.065-7.191). CONCLUSION These results showed that the GG genotype of rs1800896 or the GCC haplotype are associated with COVID-19 severity in Brazilian patients.
Collapse
Affiliation(s)
- Janaina Nicolau de Oliveira
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, Pr 445 km 380 Celso Garcia Cid Highway 86.057-970 PR, Brazil
| | - Caroline Yukari Motoori Fernandes
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, Pr 445 km 380 Celso Garcia Cid Highway 86.057-970 PR, Brazil
| | - Sara Mataroli de Godoy
- Laboratory for Studies and Analysis of Polymorphisms, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina 86.057-970 PR, Brazil
| | - Wilson Frantine-Silva
- Laboratory for Studies and Analysis of Polymorphisms, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina 86.057-970 PR, Brazil
| | | | - Guilherme Lerner Trigo
- Department of Applied Pathology, Clinical and Toxicological Analysis, State University of Londrina 86.057-970 PR, Brazil
| | | | - Zuleica Naomi Tano
- Department of Clinical Medicine, University of Londrina, Londrina, PR, Brazil
| | - Andrea Name Colado Simão
- Department of Applied Pathology, Clinical and Toxicological Analysis, State University of Londrina 86.057-970 PR, Brazil
| | - Karen Brajão de Oliveira
- Laboratory of Molecular Genetics and Immunology, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina, Pr 445 km 380 Celso Garcia Cid Highway 86.057-970 PR, Brazil; Laboratory for Studies and Analysis of Polymorphisms, Department of Immunology, Parasitology and General Pathology, Center of Biological Sciences, State University of Londrina 86.057-970 PR, Brazil.
| |
Collapse
|
7
|
Kaya H, Argun Baris S, Gultepe B, Basyigit I, Boyaci H. The predictive value of the LDH-albumin ratio on poor clinical course and mortality in COVID-19 patients: A single-center study. Medicine (Baltimore) 2025; 104:e41660. [PMID: 40020123 PMCID: PMC11875623 DOI: 10.1097/md.0000000000041660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 02/06/2025] [Indexed: 03/05/2025] Open
Abstract
There are studies evaluating the association of serum lactate dehydrogenase (LDH) and albumin levels with mortality in COVID-19 patients. The aim of our study was to evaluate the predictive effect of the LDH/albumin ratio (LAR) on mortality and poor clinical course in COVID-19 patients. A total of 2093 patients for whom LDH and albumin tests were available were included in the study. Demographic data, length of hospitalization, and signs of poor clinical course were recorded and compared with the LAR value at the time of hospitalization. The study included 1010 female (48.3%) and 1083 male (51.7%) patients. Notably, 1408 (67.3%) of the patients had at least 1 comorbidity. Oxygen was required in 860 patients (41.1%) and intensive care unit was required in 215 patients (10.3%). The mortality rate was 8.1% (n: 170). The median LAR value was 8.05. A positive correlation was observed between LAR and length of hospitalization. The LAR value was significantly higher in patients who died compared with those who survived, in patients who required intensive care compared with those who did not, and in patients who required oxygen compared to those who did not. The cutoff value for LAR in predicting mortality was calculated as 10.48. The sensitivity and specificity were determined as 73.5% and 73.7%. In conclusion, serum LAR at the time of admission is predictive of poor clinical course and mortality in COVID-19 patients. Patients with LAR values higher than the cutoff value should be closely monitored for poor clinical course.
Collapse
Affiliation(s)
- Huseyin Kaya
- Department of Chest Diseases, Kocaeli City Hospital, Izmit, Kocaeli, Turkey
| | - Serap Argun Baris
- Department of Chest Diseases, University of Kocaeli, Izmit, Kocaeli, Turkey
| | | | - Ilknur Basyigit
- Department of Chest Diseases, University of Kocaeli, Izmit, Kocaeli, Turkey
| | - Hasim Boyaci
- Department of Chest Diseases, University of Kocaeli, Izmit, Kocaeli, Turkey
| |
Collapse
|
8
|
Fratta Pasini AM, Stranieri C, Di Leo EG, Bertolone L, Aparo A, Busti F, Castagna A, Vianello A, Chesini F, Friso S, Girelli D, Cominacini L. Identification of Early Biomarkers of Mortality in COVID-19 Hospitalized Patients: A LASSO-Based Cox and Logistic Approach. Viruses 2025; 17:359. [PMID: 40143288 PMCID: PMC11946718 DOI: 10.3390/v17030359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/06/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
This study aimed to identify possible early biomarkers of mortality among clinical and biochemical parameters, iron metabolism parameters, and cytokines detected within 24 h from admission in hospitalized COVID-19 patients. We enrolled 80 hospitalized patients (40 survivors and 40 non-survivors) with COVID-19 pneumonia and acute respiratory failure. The median time from the onset of COVID-19 symptoms to hospital admission was lower in non-survivors than survivors (p < 0.05). Respiratory failure, expressed as the ratio of arterial oxygen partial pressure to the fraction of inspired oxygen (P/F), was more severe in non-survivors than survivors (p < 0.0001). Comorbidities were similar in both groups. Among biochemical parameters and cytokines, eGFR and interleukin (IL)-1β were found to be significantly lower (p < 0.05), while LDH, IL-10, and IL-8 were significantly higher in non-survivors than in survivors (p < 0.0005, p < 0.05 and p < 0.005, respectively). Among other parameters, LDH values distribution showed the most significant difference between study groups (p < 0.0001). LASSO feature selection combined with Cox proportional hazards and logistic regression models was applied to identify features distinguishing between survivors and non-survivors. Both approaches highlighted LDH as the strongest predictor, with IL-22 and creatinine emerging in the Cox model, while IL-10, eGFR, and creatinine were influential in the logistic model (AUC = 0.744 for Cox, 0.723 for logistic regression). In a similar manner, we applied linear regression for predicting LDH levels, identifying the P/F ratio as the top predictor, followed by IL-10 and eGFR (NRMSE = 0.128). Collectively, these findings underscore LDH's critical role in mortality prediction, with P/F and IL-10 as key determinants of LDH increases in this Italian COVID-19 cohort.
Collapse
Affiliation(s)
- Anna Maria Fratta Pasini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Chiara Stranieri
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Edoardo Giuseppe Di Leo
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Lorenzo Bertolone
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Antonino Aparo
- Interdepartmental Laboratory of Medical Research, Research Center LURM, University of Verona, 37134 Verona, Italy;
| | - Fabiana Busti
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Annalisa Castagna
- Department of Medicine, Section of Internal Medicine B, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy (S.F.)
| | - Alice Vianello
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Fabio Chesini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Simonetta Friso
- Department of Medicine, Section of Internal Medicine B, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy (S.F.)
| | - Domenico Girelli
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| | - Luciano Cominacini
- Department of Medicine, Section of Internal Medicine D, University of Verona, Policlinico G.B. Rossi, Piazzale L.A. Scuro 10, 37134 Verona, Italy; (C.S.); (E.G.D.L.); (L.B.); (F.B.); (A.V.); (F.C.); (D.G.); (L.C.)
| |
Collapse
|
9
|
Parlițeanu OA, Bălteanu MA, Zaharia DC, Constantinescu T, Cristea AM, Dumitrache-Rujinscki Ș, Nica AE, Voineag C, Alexe OS, Tabacu E, Croitoru A, Strâmbu I, Nemeș RM, Mahler B. The Impact of SARS-CoV-2 Infection on Glucose Homeostasis in Hospitalized Patients with Pulmonary Impairment. Diagnostics (Basel) 2025; 15:554. [PMID: 40075801 PMCID: PMC11898410 DOI: 10.3390/diagnostics15050554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/22/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Background and Objectives: We conducted a retrospective observational study to evaluate the impact of elevated blood glucose levels in patients with SARS-CoV-2 infection and a prior diagnosis of diabetes mellitus (DM) or newly diagnosed hyperglycemia. Materials and Methods: This study analyzed 6065 patients admitted to the COVID-19 departments of the "Marius Nasta" National Institute of Pulmonology in Bucharest, Romania, between 26 October 2020 and 5 January 2023. Of these, 813 patients (13.40%) were selected for analysis due to either a pre-existing diagnosis of DM or hyperglycemia at the time of hospital admission. Results: The erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) levels were elevated in patients with blood glucose levels exceeding 300 mg/dL. These elevations correlated with the presence of respiratory failure and increased mortality rates. Additionally, oxygen requirements were significantly higher at elevated blood glucose levels (p < 0.001), with a direct relationship between glycemia and oxygen demand. This was accompanied by lower oxygen saturation levels (p < 0.001). Maximum blood glucose levels were associated with the severity of respiratory failure (AUC 0.6, 95% CI: 0.56-0.63, p < 0.001). We identified cut-off values for blood glucose at admission (217.5 mg/dL) and maximum blood glucose during hospitalization (257.5 mg/dL), both of which were associated with disease severity and identified as risk factors for increased mortality. Conclusions: High blood glucose levels, both at admission and during hospitalization, were identified as risk factors for poor prognosis and increased mortality in patients with SARS-CoV-2 infection, regardless of whether the hyperglycemia was due to a prior diagnosis of DM or was newly developed during the hospital stay. These findings underscore the importance of glycemic control in the management of hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Oana-Andreea Parlițeanu
- Institutul Național de Pneumoftizologie Marius Nasta, 050159 București, Romania; (O.-A.P.); (D.C.Z.); (T.C.); (A.M.C.); (Ș.D.-R.); (E.T.); (A.C.); (I.S.); (R.M.N.); (B.M.)
| | - Mara-Amalia Bălteanu
- Institutul Național de Pneumoftizologie Marius Nasta, 050159 București, Romania; (O.-A.P.); (D.C.Z.); (T.C.); (A.M.C.); (Ș.D.-R.); (E.T.); (A.C.); (I.S.); (R.M.N.); (B.M.)
| | - Dragoș Cosmin Zaharia
- Institutul Național de Pneumoftizologie Marius Nasta, 050159 București, Romania; (O.-A.P.); (D.C.Z.); (T.C.); (A.M.C.); (Ș.D.-R.); (E.T.); (A.C.); (I.S.); (R.M.N.); (B.M.)
- Department of Pneumology, Universitatea de Medicină și Farmacie Carol Davila, 050474 Bucrești, Romania;
| | - Tudor Constantinescu
- Institutul Național de Pneumoftizologie Marius Nasta, 050159 București, Romania; (O.-A.P.); (D.C.Z.); (T.C.); (A.M.C.); (Ș.D.-R.); (E.T.); (A.C.); (I.S.); (R.M.N.); (B.M.)
- Department of Pneumology, Universitatea de Medicină și Farmacie Carol Davila, 050474 Bucrești, Romania;
| | - Alexandra Maria Cristea
- Institutul Național de Pneumoftizologie Marius Nasta, 050159 București, Romania; (O.-A.P.); (D.C.Z.); (T.C.); (A.M.C.); (Ș.D.-R.); (E.T.); (A.C.); (I.S.); (R.M.N.); (B.M.)
- Department of Pneumology, Universitatea de Medicină și Farmacie Carol Davila, 050474 Bucrești, Romania;
| | - Ștefan Dumitrache-Rujinscki
- Institutul Național de Pneumoftizologie Marius Nasta, 050159 București, Romania; (O.-A.P.); (D.C.Z.); (T.C.); (A.M.C.); (Ș.D.-R.); (E.T.); (A.C.); (I.S.); (R.M.N.); (B.M.)
- Department of Pneumology, Universitatea de Medicină și Farmacie Carol Davila, 050474 Bucrești, Romania;
| | - Andra Elena Nica
- Department of Pneumology, Universitatea de Medicină și Farmacie Carol Davila, 050474 Bucrești, Romania;
| | - Cristiana Voineag
- Department of Diabetes, Universitatea Dunărea de Jos, 800201 Galați, Romania; (C.V.); (O.S.A.)
| | - Octavian Sabin Alexe
- Department of Diabetes, Universitatea Dunărea de Jos, 800201 Galați, Romania; (C.V.); (O.S.A.)
| | - Emilia Tabacu
- Institutul Național de Pneumoftizologie Marius Nasta, 050159 București, Romania; (O.-A.P.); (D.C.Z.); (T.C.); (A.M.C.); (Ș.D.-R.); (E.T.); (A.C.); (I.S.); (R.M.N.); (B.M.)
- Department of Pneumology, Universitatea de Medicină și Farmacie Carol Davila, 050474 Bucrești, Romania;
| | - Alina Croitoru
- Institutul Național de Pneumoftizologie Marius Nasta, 050159 București, Romania; (O.-A.P.); (D.C.Z.); (T.C.); (A.M.C.); (Ș.D.-R.); (E.T.); (A.C.); (I.S.); (R.M.N.); (B.M.)
- Department of Pneumology, Universitatea de Medicină și Farmacie Carol Davila, 050474 Bucrești, Romania;
| | - Irina Strâmbu
- Institutul Național de Pneumoftizologie Marius Nasta, 050159 București, Romania; (O.-A.P.); (D.C.Z.); (T.C.); (A.M.C.); (Ș.D.-R.); (E.T.); (A.C.); (I.S.); (R.M.N.); (B.M.)
- Department of Pneumology, Universitatea de Medicină și Farmacie Carol Davila, 050474 Bucrești, Romania;
| | - Roxana Maria Nemeș
- Institutul Național de Pneumoftizologie Marius Nasta, 050159 București, Romania; (O.-A.P.); (D.C.Z.); (T.C.); (A.M.C.); (Ș.D.-R.); (E.T.); (A.C.); (I.S.); (R.M.N.); (B.M.)
| | - Beatrice Mahler
- Institutul Național de Pneumoftizologie Marius Nasta, 050159 București, Romania; (O.-A.P.); (D.C.Z.); (T.C.); (A.M.C.); (Ș.D.-R.); (E.T.); (A.C.); (I.S.); (R.M.N.); (B.M.)
- Department of Pneumology, Universitatea de Medicină și Farmacie Carol Davila, 050474 Bucrești, Romania;
| |
Collapse
|
10
|
Casetti R, Sacchi A, Mazzotta V, Cristofanelli F, Grassi G, Gili S, Cimini E, Notari S, Bordoni V, Mastrorosa I, Giancola ML, Vergori A, Tempestilli M, Vita S, Mariotti D, Rosati S, Lalle E, Meschi S, Colavita F, Garbuglia AR, Girardi E, Nicastri E, Antinori A, Agrati C. Innate and SARS-CoV-2 specific adaptive immune response kinetic in neutralizing monoclonal antibody successfully treated COVID-19 patients. Int Immunopharmacol 2025; 148:113934. [PMID: 39832460 DOI: 10.1016/j.intimp.2024.113934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/06/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025]
Abstract
The impact of anti-Spike monoclonal antibody (mAbs) treatment on the immune response of COVID19-patients is poorly explored. In particular, a comparison of the immunological influence of different therapeutic regimens has not yet been performed. Aim of the study was to compare the kinetic of innate and adaptive immune response as well as the SARS-CoV-2 specific humoral and T cell response in two groups of SARS-CoV-2-infected patients treated with two different mAbs regimens: Bamlanivimab/Etesevimab (BAM/ETE) or Casirivimab/Imdevimab (CAS/IMD). SARS-CoV-2-infected patients (n = 39) with mild/moderate disease were enrolled before (T0) and after 7 days (T7) and 30 day (T30) from mAbs infusion. Patients were divided in two groups on the basis of the mAb regimen: BAM/ETE (n = 15) and CAS/IMD (n = 24). The phenotype/function of immune cell subsets was evaluated by flow-cytometry and by ELISA. The Spike-specific T cell response (IFN-γ) and anti-Nucleocapside IgG were evaluated by chemiluminescence assay. SARS CoV-2 RNA in nasal swabs was evaluated by RT-PCR. Eleven out of the thirty-nine enrolled patients tested negative at T7, among which nine (81.8 %) had been treated with CAS/IMD regimen. A comparable increase in CD4 and CD8 T cells was observed in both treatment groups. Moreover, a reduction of CD38 expression on T (CD4, CD8 and Vδ2) and on NK cells was observed in both groups, as well as a reduction overtime of the perforin expression in T (CD8, Vδ2) and in NK cells reaching significance only in CAS/IMD-treated patients. The SARS-CoV-2-specific T cells response increased at T7 in BAM/ETE-treated patients and at T30 in CAS/IND group. Of note, at T30 SARS-CoV2-specific T cells was higher in CAS/IMD than in BAM/ETE group. Furthermore, the titre of anti-N IgG increased overtime in both groups with a faster kinetic in CAS/IMD group. The spontaneous production of inflammatory cytokines by monocytes and neutrophils was similar the two mAb regimens, as well as the level of plasmatic IL-6. Finally, patients were also analysed according to sex. The male group showed a higher frequency of activated CD4 T cells, NKG2A-expressing CD8 T cells and perforin-expressing Vδ2 T cells compared to female group. Moreover, a higher specific T cell response at T30 was observed in the male compared to female group. In conclusion, these results show similar effects of both mAb regimens in restoring T and NK cell homeostasis and in reducing inflammation. In contrast, CAS/IMD allows a better humoral and cellular SARS-CoV2 specific immunization.
Collapse
Affiliation(s)
- Rita Casetti
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Alessandra Sacchi
- Molecular Virology and Antimicrobic Immunity Laboratory, Department of Science, University of Rome Three, 00146 Rome, Italy.
| | | | - Flavia Cristofanelli
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Germana Grassi
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Simona Gili
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Eleonora Cimini
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Stefania Notari
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Veronica Bordoni
- Unit of Pathogen Specific Immunity, Bambino Gesù Children's Hospital, IRCCS, Rome 00146 Italy.
| | | | | | | | - Massimo Tempestilli
- Cellular Immunology and Pharmacology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Serena Vita
- Clinical Department, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Davide Mariotti
- Virology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Silvia Rosati
- Clinical Department, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Eleonora Lalle
- Virology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Silvia Meschi
- Virology Laboratory, INMI L. Spallanzani, 00149 Rome, Italy.
| | | | | | - Enrico Girardi
- Scientific Directorate, INMI L. Spallanzani, 00149 Rome, Italy.
| | | | - Andrea Antinori
- Clinical Department, INMI L. Spallanzani, 00149 Rome, Italy.
| | - Chiara Agrati
- Unit of Pathogen Specific Immunity, Bambino Gesù Children's Hospital, IRCCS, Rome 00146 Italy.
| |
Collapse
|
11
|
Lee C, Khan R, Mantsounga CS, Sharma S, Pierce J, Amelotte E, Butler CA, Farinha A, Parry C, Caballero O, Morrison JA, Uppuluri S, Whyte JJ, Kennedy JL, Zhang X, Choudhary G, Olson RM, Morrison AR. IL-1β-driven NF-κB transcription of ACE2 as a Mechanism of Macrophage Infection by SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.24.630260. [PMID: 39763770 PMCID: PMC11703209 DOI: 10.1101/2024.12.24.630260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Coronavirus disease 2019 (COVID-19), caused by infection with the enveloped RNA betacoronavirus, SARS-CoV-2, led to a global pandemic involving over 7 million deaths. Macrophage inflammatory responses impact COVID-19 severity; however, it is unclear whether macrophages are infected by SARS-CoV-2. We sought to identify mechanisms regulating macrophage expression of ACE2, the primary receptor for SARS-CoV-2, and to determine if macrophages are susceptible to productive infection. We developed a humanized ACE2 (hACE2) mouse whereby hACE2 cDNA was cloned into the mouse ACE2 locus under control of the native promoter. We validated the susceptibility of hACE2 mice to SARS-CoV-2 infection relative to wild-type mice and an established K18-hACE2 model of acute fulminating disease. Intranasal exposure to SARS-CoV-2 led to pulmonary consolidations with cellular infiltrate, edema, and hemorrhage, consistent with pneumonia, yet unlike the K18-hACE2 model, hACE2 mice survived and maintained stable weight. Infected hACE2 mice also exhibited a unique plasma chemokine, cytokine, and growth factor inflammatory signature relative to K18-hACE2 mice. Infected hACE2 mice demonstrated evidence of viral replication in infiltrating lung macrophages, and infection of macrophages in vitro revealed a transcriptional profile indicative of altered RNA and ribosomal processing machinery as well as activated cellular antiviral defense. Macrophage IL-1β-driven NF-κB transcription of ACE2 was an important mechanism of dynamic ACE2 upregulation, promoting macrophage susceptibility to infection. Experimental models of COVID-19 that make use of native hACE2 expression will allow for mechanistic insight into factors that can either promote host resilience or increase susceptibility to worsening severity of infection.
Collapse
Affiliation(s)
- Cadence Lee
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Rachel Khan
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Chris S. Mantsounga
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Sheila Sharma
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Julia Pierce
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Elizabeth Amelotte
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Celia A. Butler
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Andrew Farinha
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Crystal Parry
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Olivya Caballero
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Jeremi A. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Saketh Uppuluri
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | - Jeffrey J. Whyte
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri Division of Research, Innovation and Impact, Columbia, Missouri, USA
| | - Joshua L. Kennedy
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Arkansas Children’s Research Institute, Little Rock, Arkansas, USA
| | - Xuming Zhang
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Gaurav Choudhary
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
- Cardiovascular Research Center, Lifespan Cardiovascular Research Institute, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Rachel M. Olson
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
- Laboratory for Infectious Disease Research, University of Missouri Division of Research, Innovation and Impact, Columbia, Missouri, USA
| | - Alan R. Morrison
- Vascular Research Laboratory, Providence VA Medical Center, Providence, Rhode Island 02908, USA
- Ocean State Research Institute, Inc., Providence, Rhode Island 02908, USA
- Department of Internal Medicine, Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
- Lead contact and corresponding author
| |
Collapse
|
12
|
Adamopoulos PG, Bartzoka N, Tsiakanikas P, Scorilas A. Characterization of novel ACE2 mRNA transcripts: The potential role of alternative splicing in SARS-CoV-2 infection. Gene 2025; 936:149092. [PMID: 39549777 DOI: 10.1016/j.gene.2024.149092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/25/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024]
Abstract
The human angiotensin converting enzyme 2 (ACE2) gene encodes a type I transmembrane protein, which is homologous to angiotensin I-converting enzyme (ACE) and belongs to the angiotensin-converting enzyme family of dipeptidyl carboxypeptidases. As highlighted by the COVID-19 pandemic, ACE2 is not only crucial for the renin-angiotensin-aldosterone system (RAAS), but also displays great affinity with the SARS-CoV-2 spike protein, representing the major receptor of the virus. Given the significance of ACE2 in COVID-19, especially among cancer patients, the present study aims to explore the transcriptional landscape of ACE2 in human cancer and non-cancerous cell lines through the design and implementation of a custom targeted long-read sequencing approach. Bioinformatics analysis of the massive parallel sequencing data led to the identification of novel ACE2 mRNA splice variants (ACE2 sv.7-sv.12) that demonstrate previously uncharacterized exon-skipping events as well as 5' and/or 3' alternative splice sites. Demultiplexing of the sequencing data elucidated the differential expression profile of the identified splice variants in multiple human cell types, whereas in silico analysis suggests that some of the novel splice variants could produce truncated ACE2 isoforms with altered functionalities, potentially influencing their interaction with the SARS-CoV-2 spike protein. In summary, our study sheds light on the complex alternative splicing landscape of the ACE2 gene in cancer cell lines, revealing novel splice variants that could have significant implications for SARS-CoV-2 susceptibility in cancer patients. These findings contribute to the increased understanding of ACE2's role in COVID-19 and highlight the importance of considering alternative splicing as a key factor in viral pathogenesis. Undoubtably, further research is needed to explore the functional roles of these variants and their potential as therapeutic targets in the ongoing fight against COVID-19.
Collapse
Affiliation(s)
- Panagiotis G Adamopoulos
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Natalia Bartzoka
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Tsiakanikas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
13
|
Zhao Y, Tang Y, Wang QY, Li J. Ocular neuroinflammatory response secondary to SARS-CoV-2 infection-a review. Front Immunol 2025; 16:1515768. [PMID: 39967658 PMCID: PMC11832381 DOI: 10.3389/fimmu.2025.1515768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
With the consistent occurrence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the prevalence of various ocular complications has increased over time. SARS-CoV-2 infection has been shown to have neurotropism and therefore to lead to not only peripheral inflammatory responses but also neuroinflammation. Because the receptor for SARS-CoV-2, angiotensin-converting enzyme 2 (ACE2), can be found in many intraocular tissues, coronavirus disease 2019 (COVID-19) may also contribute to persistent intraocular neuroinflammation, microcirculation dysfunction and ocular symptoms. Increased awareness of neuroinflammation and future research on interventional strategies for SARS-CoV-2 infection are important for improving long-term outcomes, reducing disease burden, and improving quality of life. Therefore, the aim of this review is to focus on SARS-CoV-2 infection and intraocular neuroinflammation and to discuss current evidence and future perspectives, especially possible connections between conditions and potential treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Jia Li
- Department of Glaucoma, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Subramaniam S, Jose A, Kenney D, O’Connell AK, Bosmann M, Douam F, Crossland N. Challenging the notion of endothelial infection by SARS-CoV-2: insights from the current scientific evidence. Front Immunol 2025; 16:1443932. [PMID: 39967675 PMCID: PMC11832389 DOI: 10.3389/fimmu.2025.1443932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 01/14/2025] [Indexed: 02/20/2025] Open
Affiliation(s)
- Saravanan Subramaniam
- Department of Pharmacology and Toxicology, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
- Renal Section, Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Asha Jose
- Renal Section, Department of Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Devin Kenney
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Aoife K. O’Connell
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Markus Bosmann
- Department of Medicine, Pulmonary Center, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| | - Florian Douam
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
| | - Nicholas Crossland
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, United States
| |
Collapse
|
15
|
Lespezeanu DA, Kraft A, Moldovan C, Ungureanu D, Bacalbasa N. The short-term follow-up of patients with diabetes mellitus presenting with COVID-19. J Med Life 2025; 18:116-124. [PMID: 40134446 PMCID: PMC11932506 DOI: 10.25122/jml-2025-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 02/23/2025] [Indexed: 03/27/2025] Open
Abstract
The COVID-19 pandemic has disproportionately affected individuals with diabetes mellitus (DM), significantly increasing their risk of adverse outcomes. This retrospective study aimed to explore the underlying factors contributing to the heightened vulnerability of individuals with DM to severe COVID-19. We reviewed medical records of patients diagnosed with DM from August 2020 to August 2022 and identified 60 equally divided into two groups. Group A (n = 30) included those diagnosed with an associated COVID-19 infection, while Group B (n = 30) served as the control group without a COVID-19 infection. Inflammatory biomarkers, venous blood glucose levels, and other parameters were assessed at hospital admission and again after a 14-day treatment period. Statistical analysis confirmed a strong association between diabetes and COVID-19 infection. In COVID-19 patients treated with Empagliflozin, correlations were observed between IL-1, TNF-alpha, IL-6, and blood glucose levels. Patients in Group B did not show significant improvements in inflammatory markers or blood glucose control. In contrast, in the first group, better correlations between interleukin levels and blood glucose were demonstrated, suggesting a higher success rate for that treatment. Our findings indicate that while Empagliflozin had limited efficacy in managing long-term diabetes-related complications, it might offer significant benefits in the acute phase of illness.
Collapse
Affiliation(s)
- Delia-Andreea Lespezeanu
- Ion Pavel Diabetes Center, Prof. Dr. N. C. Paulescu National Institute of Diabetes, Nutrition and Metabolic Diseases, Bucharest, Romania
- Doctoral School, Titu Maiorescu University of Bucharest, Romania
| | - Alin Kraft
- Department of General Surgery, Regina Maria Military Emergency Hospital, Brasov, Romania
| | - Cosmin Moldovan
- Department of Medical-Surgical and Prophylactic Disciplines, Faculty of Medicine, Titu Maiorescu University, Bucharest, Romania
- Department of General Surgery, Witting Clinical Hospital, Bucharest, Romania
| | - Dan Ungureanu
- Doctoral School, Titu Maiorescu University of Bucharest, Romania
| | - Nicolae Bacalbasa
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Visceral Surgery, Center of Excellence in Translational Medicine, Fundeni Clinical Institute, Bucharest, Romania
- Department of Visceral Surgery, Center of Digestive Diseases and Liver Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| |
Collapse
|
16
|
García AMG, Arias Arias AJ, Muñoz FL, García-Rico E. Allostatic Load as a Short-Term Prognostic and Predictive Marker. Stress Health 2025; 41:e3527. [PMID: 39789760 DOI: 10.1002/smi.3527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/22/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
It would be highly valuable to possess a tool for evaluating disease progression and identifying patients at risk of experiencing a more severe clinical course and potentially worse outcomes. The concept of allostatic load, which represents the overall strain on the body from repeated stress responses, has been recognized as a precursor to the development of chronic illnesses. It functions as a cumulative measure of the body's capacity to adapt to stress. Numerous studies have demonstrated that elevated allostatic load levels are associated with various negative health outcomes, both physical and mental, and are more predictive of mortality than individual biomarkers. Leveraging the unique circumstances presented by the COVID-19 pandemic, we evaluated different clinical and laboratory parameters in hospitalised COVID-19 patients to assess allostatic load. Our results indicated that allostatic load acts as a strong predictor of prolonged hospitalisation, increased ICU days, and mortality. This highlights its efficacy as a precise gauge of biological dysregulation linked to the response to COVID-19 during disease progression. Allostatic load is easily obtainable and provides an early, cost-effective indication of disease prognosis. Additionally, it has the potential to forecast the necessity for ICU admission. As a result, this parameter, indicative of the comprehensive physiological disruption in response to stress, emerges as a promising prognostic marker for hospitalised patients, extending beyond COVID-19.
Collapse
Affiliation(s)
- Ana M Gómez García
- Internal Medicine Unit, Hospital Universitario HM Madrid, Madrid, Spain
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Villafranca del Castillo, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | - Angel Jesús Arias Arias
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Villafranca del Castillo, Spain
| | - Francisco López Muñoz
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Villafranca del Castillo, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
| | - E García-Rico
- Facultad HM de Ciencias de la Salud de la Universidad Camilo José Cela, Villafranca del Castillo, Spain
- Instituto de Investigación Sanitaria HM Hospitales, Madrid, Spain
- Medical Oncology Unit, Hospital Universitario HM Torrelodones, Madrid, Spain
| |
Collapse
|
17
|
Sobh A, Elnagdy MH, Mosa DM, Korkor MS, Alawfi AD, Alshengeti AM, Al-Mazroea AH, Bafail R, Samman WA, El-Agamy DS, Abo-Haded HM. Longitudinal cytokine profile in severe COVID-19 and multisystem inflammatory syndrome in children: A single centre study from Egypt. J Paediatr Child Health 2025; 61:249-261. [PMID: 39679634 DOI: 10.1111/jpc.16746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 10/22/2024] [Accepted: 11/28/2024] [Indexed: 12/17/2024]
Abstract
AIM The severity of COVID-19 is influenced by uncontrolled hyper-inflammatory response with excessive release of many cytokines and chemokines. The understanding of the temporal change in the cytokine levels that underlies the diverse clinical presentations of COVID-19 can help in the prediction of the disease outcome and in the design of proper treatment strategies. METHOD Data were collected from children (<18 years old) hospitalised with severe COVID-19 or severe MIS-C who were compared to a group of healthy control children. Patient demographics, clinical, laboratory data and cytokines profiles were evaluated. Blood samples were collected within 24 h of admission for all enrolled children and on Day 14. RESULTS Twenty-five children with severe COVID-19 and 23 cases with severe MIS-C were included in the study. The biochemical and inflammatory markers tend to be elevated in MIS-C group. There was a significant difference between studied cases and the control group in the following cytokines: G-CSF, IL-10, HMGB1, TNF-α, IL-6, IL-8 and INF-gamma (P < 0.05). While there was a significant difference between severe COVID-19 and MIS-C groups in the following cytokines at Day 1 of admission; IL-10, IL-6, IL-8 and INF-gamma; while at Day 14, there was a significant difference only for G-CSF, IL-10 and IL-6, all other cytokines were comparable. CONCLUSION Our study underpinned patterns of cytokine response in severe COVID-19 and MIS-C. There is a significant upregulation in pro-inflammatory cytokines (mainly G-CSF, IL-10, HMGB1, TNF-α, IL-6, IL-8 and INF-gamma). These biomarkers that could imply on the severity rating and treatment strategies, should be preferentially assessed in SARS-CoV-2 associated immunological events.
Collapse
Affiliation(s)
- Ali Sobh
- Department of Pediatrics, Mansoura University Children's Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Marwa H Elnagdy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Doaa Mosad Mosa
- Department of Rheumatology and Rehabilitation, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mai S Korkor
- Department of Pediatrics, Mansoura University Children's Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Abdulsalam D Alawfi
- Department of Pediatrics, College of Medicine, Taibah University, Madinah, Saudi Arabia
| | - Amer M Alshengeti
- Department of Pediatrics, College of Medicine, Taibah University, Madinah, Saudi Arabia
| | | | - Rawan Bafail
- Department of Pharmaceutics & Pharmaceutical Technology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Waad A Samman
- Department of Pharmacology & Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Dina S El-Agamy
- Department of Pharmacology, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Hany M Abo-Haded
- Department of Pediatrics, Mansoura University Children's Hospital, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
18
|
Alirezaee A, Mirmoghtadaei M, Heydarlou H, Akbarian A, Alizadeh Z. Interferon therapy in alpha and Delta variants of SARS-CoV-2: The dichotomy between laboratory success and clinical realities. Cytokine 2025; 186:156829. [PMID: 39693873 DOI: 10.1016/j.cyto.2024.156829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024]
Abstract
The COVID-19 pandemic has caused significant morbidity and mortality worldwide. The emergence of the Alpha and Delta variants of SARS-CoV-2 has led to a renewed interest in using interferon therapy as a potential treatment option. Interferons are a group of signaling proteins produced by host cells in response to viral infections. They play a critical role in the innate immune response to viral infections by inducing an antiviral state in infected and neighboring cells. Interferon therapy has shown promise as a potential treatment option for COVID-19. In this review paper, we review the current knowledge regarding interferon therapy in the context of the Alpha and Delta variants of SARS-CoV-2 and discuss the challenges that must be overcome to translate laboratory findings into effective clinical treatments.
Collapse
Affiliation(s)
- Atefe Alirezaee
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Milad Mirmoghtadaei
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Hanieh Heydarlou
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Asiye Akbarian
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Zahra Alizadeh
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran; Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Li B, Jiang AY, Raji I, Atyeo C, Raimondo TM, Gordon AGR, Rhym LH, Samad T, MacIsaac C, Witten J, Mughal H, Chicz TM, Xu Y, McNamara RP, Bhatia S, Alter G, Langer R, Anderson DG. Enhancing the immunogenicity of lipid-nanoparticle mRNA vaccines by adjuvanting the ionizable lipid and the mRNA. Nat Biomed Eng 2025; 9:167-184. [PMID: 37679571 DOI: 10.1038/s41551-023-01082-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/27/2023] [Indexed: 09/09/2023]
Abstract
To elicit optimal immune responses, messenger RNA vaccines require intracellular delivery of the mRNA and the careful use of adjuvants. Here we report a multiply adjuvanted mRNA vaccine consisting of lipid nanoparticles encapsulating an mRNA-encoded antigen, optimized for efficient mRNA delivery and for the enhanced activation of innate and adaptive responses. We optimized the vaccine by screening a library of 480 biodegradable ionizable lipids with headgroups adjuvanted with cyclic amines and by adjuvanting the mRNA-encoded antigen by fusing it with a natural adjuvant derived from the C3 complement protein. In mice, intramuscular or intranasal administration of nanoparticles with the lead ionizable lipid and with mRNA encoding for the fusion protein (either the spike protein or the receptor-binding domain of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)) increased the titres of antibodies against SARS-CoV-2 tenfold with respect to the vaccine encoding for the unadjuvanted antigen. Multiply adjuvanted mRNA vaccines may improve the efficacy, safety and ease of administration of mRNA-based immunization.
Collapse
MESH Headings
- Animals
- Nanoparticles/chemistry
- SARS-CoV-2/immunology
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- Lipids/chemistry
- Adjuvants, Immunologic
- mRNA Vaccines/immunology
- COVID-19 Vaccines/immunology
- Mice
- COVID-19/prevention & control
- COVID-19/immunology
- Female
- Mice, Inbred BALB C
- Antibodies, Viral/immunology
- Adjuvants, Vaccine
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Administration, Intranasal
- Humans
- Immunogenicity, Vaccine
- Liposomes
Collapse
Affiliation(s)
- Bowen Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Allen Yujie Jiang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Idris Raji
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caroline Atyeo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Division of Medical Sciences, Harvard University, Boston, MA, USA
| | - Theresa M Raimondo
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Akiva G R Gordon
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Luke H Rhym
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tahoura Samad
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Corina MacIsaac
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacob Witten
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Haseeb Mughal
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Taras M Chicz
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Yue Xu
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Ryan P McNamara
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Sangeeta Bhatia
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Wyss Institute at Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
20
|
Amoroso D, Bongo S, Copponi A, Rossi V, Di Giorgio R, Bernardini S, Ippoliti L, Morello M. A Review of the Hematological Picture of Severe COVID-19 Infection. Cureus 2025; 17:e78797. [PMID: 39931501 PMCID: PMC11808344 DOI: 10.7759/cureus.78797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2025] [Indexed: 02/13/2025] Open
Abstract
Numerous hematological abnormalities have been documented in COVID-19 patients. We conducted an analysis of 82 articles from PubMed, focusing on the hematological characteristics observed in survivors (S) and non-survivors (NS) with moderate and severe COVID-19 symptoms, respectively. Our review underlines neutrophilia, lymphopenia, and thrombocytopenia as hallmark features of the disease. In severe cases, blood cell microscopy revealed the following abnormalities: i) an increased number of neutrophils, often displaying granularity, toxic granulation, and vacuolization; ii) lymphocytes with a notably blue cytoplasm; iii) several monocytes that contain vacuoles; iv) platelet aggregation; and v) basophilic stippling in red blood cells. Furthermore, scattergram analysis of COVID-19 patients revealed two common features: i) an increased neutrophil population and ii) the presence of a distinctive "sandglass pattern". This review underscores the critical role of hematochemical and cytomorphological blood cell analysis in COVID-19 patients, aiding clinicians in better recognizing and understanding the indicators of disease severity.
Collapse
Affiliation(s)
- Dominga Amoroso
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Stefania Bongo
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Anna Copponi
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Vanessa Rossi
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Roberta Di Giorgio
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Sergio Bernardini
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| | - Lorenzo Ippoliti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, ITA
| | - Maria Morello
- Department of Experimental Medicine, Faculty of Medicine, University of Rome Tor Vergata, Rome, ITA
| |
Collapse
|
21
|
Zhang FL, Chen YL, Luo ZY, Song ZB, Chen Z, Zhang JX, Zheng ZZ, Tan XM. Huashi baidu granule alleviates inflammation and lung edema by suppressing the NLRP3/caspase-1/GSDMD-N pathway and promoting fluid clearance in a porcine reproductive and respiratory syndrome (PRRS) model. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119207. [PMID: 39653102 DOI: 10.1016/j.jep.2024.119207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huashi Baidu Granule (HSBDG), a traditional Chinese medicine (TCM), is used for treating coronavirus disease 2019 (COVID-19). Porcine reproductive and respiratory syndrome (PRRS) is considered the "COVID-19" for swine. According to the TCM theory, "dampness" is the main pathogenic factor in COVID-19 and PRRS, and "Huashi" means that this formula is good at removing "dampness". Studies have demonstrated that HSBDG's effect in COVID-19; but the mechanism of removing "dampness" remains elusive. AIM OF THE STUDY We aimed to assess the effect of HSBDG on PRRS, and elucidate its potential mechanism in removing "dampness". MATERIALS AND METHODS We established a PRRS-virus (PRRSV)-infected Marc-145 cells model, and performed qRT-PCR, Western blot analysis, and indirect immunofluorescence assay to examine the anti-PRRSV effects of HSBDG in vitro. PRRSV-infected pig model was established and used to investigate HSBDG's effect in PRRS and explore underlying mechanisms in removing "dampness" using ELISA and immunohistochemistry assay methods. RESULTS HSBDG exhibited anti-PRRSV activity and suppressed the viral replication and release phases. HSBDG treatment alleviated PRRS, lowered rectal temperature, reduced histopathological changes and viral load in lung tissues, and ameliorated organ lesions. Moreover, IL-1β, IL-6, IL-8, and TNF-α expressions were decreased in lung tissues. Mechanistically, HSBDG inhibited the NLRP3/Caspase-1/GSDMD-N pathway to reduce the inflammatory response and upregulated AQP1, AQP5, α-ENaC, and Na-K-ATPase expressions to promote lung fluid clearance. CONCLUSION HSBDG exerted anti-PRRSV effects and could attenuate PRRS. HSBDG potentially removes "dampness" by attenuating inflammation by suppressing the NLRP3/Caspase-1/GSDMD-N pathway and inhibiting pulmonary edema by upregulating the expression of AQP1, AQP5, α-ENaC, and Na-K-ATPase.
Collapse
Affiliation(s)
- Feng-Lin Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| | - Yi-Lin Chen
- South China Agricultural University College of Veterinary Medicine, Guangzhou, 510640, China.
| | - Zhen-Ye Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| | - Ze-Bu Song
- South China Agricultural University College of Veterinary Medicine, Guangzhou, 510640, China.
| | - Zhe Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| | - Jia-Xuan Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| | - Ze-Zhong Zheng
- South China Agricultural University College of Veterinary Medicine, Guangzhou, 510640, China.
| | - Xiao-Mei Tan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| |
Collapse
|
22
|
Wohlwend J, Nathan A, Shalon N, Crain CR, Tano-Menka R, Goldberg B, Richards E, Gaiha GD, Barzilay R. Deep learning enhances the prediction of HLA class I-presented CD8 + T cell epitopes in foreign pathogens. NAT MACH INTELL 2025; 7:232-243. [PMID: 40008296 PMCID: PMC11847706 DOI: 10.1038/s42256-024-00971-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/10/2024] [Indexed: 02/27/2025]
Abstract
Accurate in silico determination of CD8+ T cell epitopes would greatly enhance T cell-based vaccine development, but current prediction models are not reliably successful. Here, motivated by recent successes applying machine learning to complex biology, we curated a dataset of 651,237 unique human leukocyte antigen class I (HLA-I) ligands and developed MUNIS, a deep learning model that identifies peptides presented by HLA-I alleles. MUNIS shows improved performance compared with existing models in predicting peptide presentation and CD8+ T cell epitope immunodominance hierarchies. Moreover, application of MUNIS to proteins from Epstein-Barr virus led to successful identification of both established and novel HLA-I epitopes which were experimentally validated by in vitro HLA-I-peptide stability and T cell immunogenicity assays. MUNIS performs comparably to an experimental stability assay in terms of immunogenicity prediction, suggesting that deep learning can reduce experimental burden and accelerate identification of CD8+ T cell epitopes for rapid T cell vaccine development.
Collapse
Affiliation(s)
- Jeremy Wohlwend
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA USA
- Jameel Clinic, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Anusha Nathan
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA USA
- Program in Health Sciences and Technology, Harvard Medical School and Massachusetts Institute of Technology, Boston, MA USA
| | - Nitan Shalon
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA USA
- Jameel Clinic, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Charles R. Crain
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA USA
| | - Rhoda Tano-Menka
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA USA
| | | | - Emma Richards
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA USA
| | - Gaurav D. Gaiha
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA USA
- Program in Health Sciences and Technology, Harvard Medical School and Massachusetts Institute of Technology, Boston, MA USA
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA USA
| | - Regina Barzilay
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA USA
- Jameel Clinic, Massachusetts Institute of Technology, Cambridge, MA USA
| |
Collapse
|
23
|
Du S, Jin J, Tang C, Su Z, Wang L, Chen X, Zhang M, Zhu Y, Wang J, Ju C, Song X, Li S. Airway Basal Stem Cells Inflammatory Alterations in COVID-19 and Mitigation by Mesenchymal Stem Cells. Cell Prolif 2025:e13812. [PMID: 39865778 DOI: 10.1111/cpr.13812] [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: 09/25/2024] [Revised: 12/18/2024] [Accepted: 01/11/2025] [Indexed: 01/28/2025] Open
Abstract
SARS-CoV-2 infection and the resultant COVID-19 pneumonia cause significant damage to the airway and lung epithelium. This damage manifests as mucus hypersecretion, pulmonary inflammation and fibrosis, which often lead to long-term complications collectively referred to as long COVID or post-acute sequelae of COVID-19 (PASC). The airway epithelium, as the first line of defence against respiratory pathogens, depends on airway basal stem cells (BSCs) for regeneration. Alterations in BSCs are associated with impaired epithelial repair and may contribute to the respiratory complications observed in PASC. Given the critical role of BSCs in maintaining epithelial integrity, understanding their alterations in COVID-19 is essential for developing effective therapeutic strategies. This study investigates the intrinsic properties of BSCs derived from COVID-19 patients and evaluates the modulatory effects of mesenchymal stem cells (MSCs). Through a combination of functional assessments and transcriptomic profiling, we identified key phenotypic and molecular deviations in COVID-19 patient-derived BSCs, including goblet cell hyperplasia, inflammation and fibrosis, which may underlie their contribution to PASC. Notably, MSC co-culture significantly mitigated these adverse effects, potentially through modulation of the interferon signalling pathway. This is the first study to isolate BSCs from COVID-19 patients in the Chinese population and establish a COVID-19 BSC-based xenograft model. Our findings reveal critical insights into the role of BSCs in epithelial repair and their inflammatory alterations in COVID-19 pathology, with potential relevance to PASC and virus-induced respiratory sequelae. Additionally, our study highlights MSC-based therapies as a promising strategy to address respiratory sequelae and persistent symptoms.
Collapse
Affiliation(s)
- Sheng Du
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Jing Jin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chunli Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhuquan Su
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lulin Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xinyuan Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mengni Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yiping Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiaojiao Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chunrong Ju
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xinyu Song
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shiyue Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Guangzhou, China
| |
Collapse
|
24
|
Beltrami VA, Martins FRB, Martins DG, Queiroz-Junior CM, Félix FB, Resende LC, Santos FRDS, Lacerda LDSB, Costa VRDM, da Silva WN, Guimaraes PPG, Guimaraes G, Soriani FM, Teixeira MM, Costa VV, Pinho V. Selective phosphodiesterase 4 inhibitor roflumilast reduces inflammation and lung injury in models of betacoronavirus infection in mice. Inflamm Res 2025; 74:24. [PMID: 39862252 DOI: 10.1007/s00011-024-01985-3] [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: 07/31/2024] [Revised: 10/03/2024] [Accepted: 10/16/2024] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE We aimed to understand the potential therapeutic and anti-inflammatory effects of the phosphodiesterase-4 (PDE4) inhibitor roflumilast in models of pulmonary infection caused by betacoronaviruses. METHODS Mice were infected intranasally with murine hepatitis virus (MHV-3) or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Roflumilast was given to MHV-3-infected mice therapeutically at doses of 1 mg/kg or 10 mg/kg, or prophylactically at 10 mg/kg. In SARS-CoV-2-infected mice, roflumilast was given therapeutically at a dose of 10 mg/kg. Lung histopathology, chemokines (CXCL-1 and CCL2), cytokines (IL-1β, IL-6, TNF, IFN-γ, IL-10 and TGFβ), neutrophil immunohistochemical staining (Ly6G+ cells), macrophage immunofluorescence staining (F4/80+ cells), viral titration plaque assay, real-time PCR virus detection, and blood cell counts were examined. RESULTS Therapeutic treatment with roflumilast at 10 mg/kg reduced lung injury in SARS-CoV-2 or MHV-3-infected mice without compromising viral clearance. In MHV-3-infected mice, reduced lung injury was associated with decreased chemokines levels, prevention of neutrophil aggregates and reduced macrophage accumulation in the lung tissue. However, the prophylactic treatment strategy with roflumilast increased lung injury in MHV-3-infected mice. CONCLUSION Our findings indicate that therapeutic treatment with roflumilast reduced lung injury in MHV-3 and SARS-CoV-2 lung infections. Given the protection induced by roflumilast in inflammation, PDE4 targeting could be a promising therapeutic avenue worth exploring following severe viral infections of the lung.
Collapse
Affiliation(s)
- Vinícius Amorim Beltrami
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Flávia Rayssa Braga Martins
- Departamento Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Débora Gonzaga Martins
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Celso Martins Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Franciel Batista Félix
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Letícia Cassiano Resende
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Felipe Rocha da Silva Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Larisse de Souza Barbosa Lacerda
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Victor Rodrigues de Melo Costa
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Walison Nunes da Silva
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Pedro Pires Goulart Guimaraes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Goulart Guimaraes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Frederico Marianetti Soriani
- Departamento Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Vivian Vasconcelos Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| |
Collapse
|
25
|
Chu KA, Lai CY, Chen YH, Kuo FH, Chen IY, Jiang YC, Liu YL, Ko TL, Fu YS. An animal model of severe acute respiratory distress syndrome for translational research. Lab Anim Res 2025; 41:4. [PMID: 39856771 PMCID: PMC11758736 DOI: 10.1186/s42826-025-00235-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/30/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Despite the fact that an increasing number of studies have focused on developing therapies for acute lung injury, managing acute respiratory distress syndrome (ARDS) remains a challenge in intensive care medicine. Whether the pathology of animal models with acute lung injury in prior studies differed from clinical symptoms of ARDS, resulting in questionable management for human ARDS. To evaluate precisely the therapeutic effect of transplanted stem cells or medications on acute lung injury, we developed an animal model of severe ARDS with lower lung function, capable of keeping the experimental animals survive with consistent reproducibility. Establishing this animal model could help develop the treatment of ARDS with higher efficiency. RESULTS In this approach, we intratracheally delivered bleomycin (BLM, 5 mg/rat) into rats' left trachea via a needle connected with polyethylene tube, and simultaneously rotated the rats to the left side by 60 degrees. Within seven days after the injury, we found that arterial blood oxygen saturation (SpO2) significantly decreased to 83.7%, partial pressure of arterial oxygen (PaO2) markedly reduced to 65.3 mmHg, partial pressure of arterial carbon dioxide (PaCO2) amplified to 49.2 mmHg, and the respiratory rate increased over time. Morphologically, the surface of the left lung appeared uneven on Day 1, the alveoli of the left lung disappeared on Day 2, and the left lung shrank on Day 7. A histological examination revealed that considerable cell infiltration began on Day 1 and lasted until Day 7, with a larger area of cell infiltration. Serum levels of IL-5, IL-6, IFN-γ, MCP-1, MIP-2, G-CSF, and TNF-α substantially rose on Day 7. CONCLUSIONS This modified approach for BLM-induced lung injury provided a severe, stable, and one-sided (left-lobe) ARDS animal model with consistent reproducibility. The physiological symptoms observed in this severe ARDS animal model are entirely consistent with the characteristics of clinical ARDS. The establishment of this ARDS animal model could help develop treatment for ARDS.
Collapse
Affiliation(s)
- Kuo-An Chu
- Division of Chest Medicine, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
- School of Medicine, College of Medicine, National Sun Yat-Sen University, No. 70, Lienhai Rd., Kaohsiung, Taiwan, ROC
- School of Nursing, Fooyin University, Kaohsiung, Taiwan, ROC
- Department of Nursing, Shu-Zen Junior College of Medicine and Management, Kaohsiung, Taiwan, ROC
| | - Chia-Yu Lai
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Yu-Hui Chen
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Fu-Hsien Kuo
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - I-Yuan Chen
- Division of Chest Medicine, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - You-Cheng Jiang
- Division of Chest Medicine, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Ya-Ling Liu
- Division of Chest Medicine, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Tsui-Ling Ko
- School of Medicine, College of Medicine, National Sun Yat-Sen University, No. 70, Lienhai Rd., Kaohsiung, Taiwan, ROC.
| | - Yu-Show Fu
- Department of Anatomy and Cell Biology, School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nung Street, Taipei, Taiwan, ROC.
| |
Collapse
|
26
|
Moharram FA, Ibrahim RR, Mahgoub S, Abdel-Aziz MS, Said AM, Huang HC, Chen LY, Lai KH, Hashad N, Mady MS. Secondary metabolites of Alternaria alternate appraisal of their SARS-CoV-2 inhibitory and anti-inflammatory potentials. PLoS One 2025; 20:e0313616. [PMID: 39854441 PMCID: PMC11760621 DOI: 10.1371/journal.pone.0313616] [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: 09/05/2024] [Accepted: 10/28/2024] [Indexed: 01/26/2025] Open
Abstract
This study identifies the secondary metabolites from Alternaria alternate and evaluates their ACE-2: Spike RBD (SARS-CoV-2) inhibitory activity confirmed via immunoblotting in human lung microvascular endothelial cells. In addition, their in vitro anti-inflammatory potential was assessed using a cell-based assay in LPS-treated RAW 264.7 macrophage cells. Two novel compounds, altenuline (1), phthalic acid bis (7'/7'' pentyloxy) isohexyl ester (2), along with 1-deoxyrubralactone (3) alternariol-5-O-methyl ether (4) and alternariol (5) were identified. Molecular docking and in vitro studies showed that compounds 2 and 4 were promising to counteract SARS-CoV-2 attachment to human ACE-2. Thus, they are considered promising natural anti-viral agents. SwissADME in silico analysis was conducted to predict the drug-like potential. Immunoblotting analysis confirmed that the tested compounds (1-4) demonstrated downregulation of ACE-2 expression in the endothelial cells from the lungs with variable degrees. Furthermore, the tested compounds (1-4) showed promising anti-inflammatory activities through TNF-α: TNFR2 inhibitory activity and their inhibitory effect on the proinflammatory cytokines (TNF-α and IL-6) in LPS-stimulated monocytes. In conclusion, our study, for the first time, provides beneficial experimental confirmation for the efficiency of the A. alternate secondary metabolites for the treatment of COVID-19 as they hinder SARS-CoV-2 infection and lower inflammatory responses initiated by SARS-CoV-2. A. alternate and its metabolites are considered in developing preventative and therapeutic tactics for COVID-19.
Collapse
Affiliation(s)
- Fatma A. Moharram
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Reham R. Ibrahim
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Shahenda Mahgoub
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohamed S. Abdel-Aziz
- Genetic Engineering and Biotechnology Division, Microbial Chemistry Department, National Research Centre, Giza, Egypt
| | - Ahmed M. Said
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Hui-Chi Huang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Lo-Yun Chen
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Kuei-Hung Lai
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Nashwa Hashad
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohamed S. Mady
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| |
Collapse
|
27
|
Schniederova M, Bobcakova A, Grendar M, Markocsy A, Ceres A, Cibulka M, Dobrota D, Jesenak M. Lymphocyte Inhibition Mechanisms and Immune Checkpoints in COVID-19: Insights into Prognostic Markers and Disease Severity. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:189. [PMID: 40005306 PMCID: PMC11857393 DOI: 10.3390/medicina61020189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025]
Abstract
Background and Objectives: Immune checkpoint inhibitors such as PD-1 and TIM-3 play an important role in regulating the host immune response and are proposed as potential prognostic markers and therapeutic targets in severe cases of COVID-19. We evaluated the expression of PD-1 and TIM-3 on T cells, as well as the concentration of sPD-1 in plasma, to clarify the role of these molecules in patients infected with SARS-CoV-2. Materials and Methods: In this retrospective observational study, we analysed the expression of PD-1 and TIM-3 on CD4+ and CD8+ T cells upon admission and after 7 days of hospitalisation in 770 adult patients. We also evaluated sPD-1 levels in the plasma of 145 patients at different stages of COVID-19 and of 11 control subjects. Molecules were determined using conventional flow cytometry and ELISA and the data were statistically processed. Results: We observed a significantly higher expression of PD-1 on CD4+ cells in deceased patients than in those with mild-to-moderate disease. All patients with COVID-19 exhibited a significantly higher expression of TIM-3 on both CD4+ and CD8+ T cells compared to controls. After 1 week of hospitalisation, there was no significant change in PD-1 or TIM-3 expression on CD4+ or CD8+ T cells across the studied groups. sPD-1 concentrations were not significantly different between survivors and non-survivors. Plasma sPD-1 levels did not correlate with PD-1 expression on T cells, but a significant correlation was observed between CD4+ PD-1 and CD8+ PD-1. Using machine-learning algorithms, we supported our observations and confirmed immunological variables capable of predicting survival, with AUC = 0.786. Conclusions: Analysis of the immune response may be useful for monitoring and predicting the course of COVID-19 upon admission. However, it is essential to evaluate complex immune parameters in conjunction with other key clinical and laboratory indicators.
Collapse
Affiliation(s)
- Martina Schniederova
- Institute of Clinical Immunology and Medical Genetics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia; (M.S.); (A.B.); (A.M.); (M.C.)
| | - Anna Bobcakova
- Institute of Clinical Immunology and Medical Genetics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia; (M.S.); (A.B.); (A.M.); (M.C.)
- Department of Pulmonology and Phthisiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia
- Department of Paediatrics and Adolescent Medicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia
| | - Marian Grendar
- Biomed—Centre for Biomedicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03659 Martin, Slovakia
| | - Adam Markocsy
- Institute of Clinical Immunology and Medical Genetics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia; (M.S.); (A.B.); (A.M.); (M.C.)
- Department of Pulmonology and Phthisiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia
| | - Andrej Ceres
- Institute of Clinical Immunology and Medical Genetics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia; (M.S.); (A.B.); (A.M.); (M.C.)
| | - Michal Cibulka
- Institute of Clinical Immunology and Medical Genetics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia; (M.S.); (A.B.); (A.M.); (M.C.)
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03659 Martin, Slovakia;
| | - Dusan Dobrota
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03659 Martin, Slovakia;
- Department of Clinical Biochemisty, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia
| | - Milos Jesenak
- Institute of Clinical Immunology and Medical Genetics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia; (M.S.); (A.B.); (A.M.); (M.C.)
- Department of Pulmonology and Phthisiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia
- Department of Paediatrics and Adolescent Medicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin University Hospital, 03659 Martin, Slovakia
| |
Collapse
|
28
|
Guironnet-Paquet A, Hamzeh-Cognasse H, Berard F, Cognasse F, Richard JC, Yonis H, Mezidi M, Desebbe O, Delannoy B, Demeret S, Marois C, Saheb S, Le QV, Schoeffler M, Pugliesi PS, Debord S, Bastard P, Cobat A, Casanova JL, Pescarmona R, Viel S, Nicolas JF, Nosbaum A, Vocanson M, Hequet O. Therapeutic plasma exchange accelerates immune cell recovery in severe COVID-19. Front Immunol 2025; 15:1492672. [PMID: 39896810 PMCID: PMC11782122 DOI: 10.3389/fimmu.2024.1492672] [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: 09/07/2024] [Accepted: 12/04/2024] [Indexed: 02/04/2025] Open
Abstract
Background Immunological disturbances (anti-type I IFN auto-antibody production, cytokine storm, lymphopenia, T-cell hyperactivation and exhaustion) are responsible for disease exacerbation during severe COVID-19 infections. Methods In this study, we set up a prospective, randomised clinical trial (ClinicalTrials.gov ID: NCT04751643) and performed therapeutic plasma exchange (TPE) in severe COVID-19 patients in order to decrease excess cytokines and auto-antibodies and to assess whether adding TPE to the standard treatment (ST, including corticosteroids plus high-flow rate oxygen) could help restore immune parameters and limit the progression of acute respiratory distress syndrome (ARDS). Results As expected, performing TPE decreased the amount of anti-type I IFN auto-antibodies and improved the elimination or limited the production of certain inflammatory mediators (IL-18, IL-7, CCL2, CCL3, etc.) circulating in the blood of COVID-19 patients, compared to ST controls. Interestingly, while TPE did not influence changes in ARDS parameters throughout the protocol, it proved more effective than ST in reversing lymphopenia, preventing T-cell hyperactivation and reducing T-cell exhaustion, notably in a fraction of TPE patients who had an early favourable respiratory outcome. TPE also restored appropriate numbers of CD4+ and CD8+ T-cell memory populations and increased the number of circulating virus-specific T cells in these patients. Conclusion Our results therefore indicate that the addition of TPE sessions to the standard treatment accelerates immune cell recovery and contributes to the development of appropriate antiviral T-cell responses in some patients with severe COVID-19 disease.
Collapse
Affiliation(s)
- Aurelie Guironnet-Paquet
- Apheresis Unit, Etablissement Français du Sang Auvergne-Rhône-Alpes, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre Bénite, France
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
| | - Hind Hamzeh-Cognasse
- University of Jean Monnet, Mines Saint-Étienne, Institut National de la Santé et de la Recherche Médicale (INSERM), U 1059 SAINBIOSE, Saint-Étienne, France
| | - Frederic Berard
- Clinical Immunology and Allergology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Fabrice Cognasse
- University of Jean Monnet, Mines Saint-Étienne, Institut National de la Santé et de la Recherche Médicale (INSERM), U 1059 SAINBIOSE, Saint-Étienne, France
- Scientific Department, Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France
| | - Jean Christophe Richard
- Intensive Care Unit, Centre Hospitalier Croix–Rousse, Hospices Civils de Lyon (HCL), Lyon, France
| | - Hodane Yonis
- Intensive Care Unit, Centre Hospitalier Croix–Rousse, Hospices Civils de Lyon (HCL), Lyon, France
| | - Mehdi Mezidi
- Intensive Care Unit, Centre Hospitalier Croix–Rousse, Hospices Civils de Lyon (HCL), Lyon, France
| | - Olivier Desebbe
- Department of Anesthesiology and Perioperative Medicine, Sauvegarde Clinic, Ramsay Santé, Lyon, France
| | - Bertrand Delannoy
- Department of Anesthesiology and Perioperative Medicine, Sauvegarde Clinic, Ramsay Santé, Lyon, France
| | - Sophie Demeret
- Neuro-Intensive Care Unit, Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Clemence Marois
- Neuro-Intensive Care Unit, Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Paris, France
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute, Institut du Cerveau et de la Moelle (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Departement Médico-Universitaire (DMU) Neurosciences 6, Paris, France
- Groupe de Recherche Clinique en REanimation et Soins Intensifs du Patient en Insuffisance Respiratoire aiguE (GRC-RESPIRE), Sorbonne Université, Paris, France
| | - Samir Saheb
- Hemobiotherapy Unit, Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Quoc Viet Le
- Intensive Care Unit, Medipôle Lyon Villeurbanne, Villeurbanne, France
| | - Mathieu Schoeffler
- Department of Anesthesiology and Intensive Care Unit, Centre Hospitalier de Montélimar, Montélimar, France
| | - Paul Simon Pugliesi
- Intensive Care Unit, Centre Hospitalier William Morey, Chalon sur Saône, France
| | - Sophie Debord
- Department of Anesthesiology and Intensive Care Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon (HCL), Lyon, France
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique des Hopitaux de Paris (AP-HP), Paris, France
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States
| | - Jean Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique des Hopitaux de Paris (AP-HP), Paris, France
- Howards Hugues Medical Institute, New York, NY, United States
| | - Rémi Pescarmona
- Immun Monitorage Laboratory, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Sébastien Viel
- Plateforme de Biothérapies et de production de Médicaments de Thérapie Innovante (MTI), Hôpital Edouard Herriot, Hospices Civils de Lyon (HCL), Lyon, France
| | - Jean François Nicolas
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
- Clinical Immunology and Allergology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Audrey Nosbaum
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
- Clinical Immunology and Allergology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Marc Vocanson
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
| | - Olivier Hequet
- Apheresis Unit, Etablissement Français du Sang Auvergne-Rhône-Alpes, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre Bénite, France
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
| |
Collapse
|
29
|
Stegeman SK, Kourko O, Amsden H, Pellizzari Delano IE, Mamatis JE, Roth M, Colpitts CC, Gee K. RNA Viruses, Toll-Like Receptors, and Cytokines: The Perfect Storm? J Innate Immun 2025; 17:126-153. [PMID: 39820070 PMCID: PMC11845175 DOI: 10.1159/000543608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/13/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND The interactions between viruses and the host immune response are nuanced and intricate. The cytokine response arguably plays a central role in dictating the outcome of virus infection, balancing inflammation, and healing, which is crucial to resolving infection without destructive immunopathologies. SUMMARY Early innate immune responses are key to the generation of a beneficial or detrimental immune response. These initial responses are regulated by a plethora of surface bound, endosomal, and cytoplasmic innate immune receptors known as pattern recognition receptors. Of these, the Toll-like receptors (TLRs) play an important role in the induction of cytokines during virus infection. Recognizing pathogen-associated molecular patterns (PAMPs) such as viral proteins and/or nucleotide sequences, the TLRs act as sentinels for the initiation and propagation of immune responses. KEY MESSAGES TLRs are important receptors for initiating the innate response to single-stranded RNA (ssRNA) viruses like influenza A virus (IAV), severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1), SARS-CoV-2, Middle East respiratory syndrome coronavirus, dengue virus, and Ebola virus. Infection with these viruses is also associated with aberrant expression of proinflammatory cytokines that contribute to a harmful cytokine storm response. Herein we discuss the connections between these ssRNA viruses, cytokine storm, and the roles of TLRs. BACKGROUND The interactions between viruses and the host immune response are nuanced and intricate. The cytokine response arguably plays a central role in dictating the outcome of virus infection, balancing inflammation, and healing, which is crucial to resolving infection without destructive immunopathologies. SUMMARY Early innate immune responses are key to the generation of a beneficial or detrimental immune response. These initial responses are regulated by a plethora of surface bound, endosomal, and cytoplasmic innate immune receptors known as pattern recognition receptors. Of these, the Toll-like receptors (TLRs) play an important role in the induction of cytokines during virus infection. Recognizing pathogen-associated molecular patterns (PAMPs) such as viral proteins and/or nucleotide sequences, the TLRs act as sentinels for the initiation and propagation of immune responses. KEY MESSAGES TLRs are important receptors for initiating the innate response to single-stranded RNA (ssRNA) viruses like influenza A virus (IAV), severe acute respiratory syndrome coronavirus-1 (SARS-CoV-1), SARS-CoV-2, Middle East respiratory syndrome coronavirus, dengue virus, and Ebola virus. Infection with these viruses is also associated with aberrant expression of proinflammatory cytokines that contribute to a harmful cytokine storm response. Herein we discuss the connections between these ssRNA viruses, cytokine storm, and the roles of TLRs.
Collapse
Affiliation(s)
- Sophia K Stegeman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Olena Kourko
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Heather Amsden
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - John E Mamatis
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Madison Roth
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Che C Colpitts
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
30
|
dos Santos Brito WR, de Brito WB, dos Santos Ferreira F, Santana EGM, da Costa Lopes J, da Silva Graça Amoras E, Lima SS, dos Santos EF, da Costa FP, de Sarges KML, Cantanhede MHD, de Brito MTFM, da Silva ALS, de Meira Leite M, de Nazaré do Socorro de Almeida Viana M, Rodrigues FBB, da Silva R, Viana GMR, do Socorro Souza Chaves T, de Oliveira Lameira Veríssimo A, da Silva Carvalho M, Henriques DF, da Silva CP, Nunes JAL, Costa IB, Brasil-Costa I, Quaresma JAS, Cayres-Vallinoto IMV, Reis LO, Falcão LFM, dos Santos EJM, Vallinoto ACR, Queiroz MAF. Polymorphisms Influence the Expression of the Fas and FasL Genes in COVID-19. Int J Mol Sci 2025; 26:666. [PMID: 39859379 PMCID: PMC11765610 DOI: 10.3390/ijms26020666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
The apoptotic molecule Fas and its ligand FasL are involved in the process of T-lymphocyte death, which may lead to lymphopenia, a characteristic of severe coronavirus disease 2019 (COVID-19). In this study, we investigated the influence of polymorphisms in the FAS and FASL genes, FAS and FASL gene expression, and plasma cytokine levels on COVID-19 severity and long COVID occurrence. A total of 116 individuals with severe COVID-19 and 254 with the non-severe form of the disease were evaluated. In the post-COVID-19 period, samples from 196 individuals with long COVID and 67 from people who did not have long COVID were included. Genotyping and quantification of gene expression were performed via real-time PCR, and cytokine measurement was performed via flow cytometry. The AA genotype for FAS rs1800682 (A/G) and the TT genotype for FASL rs763110 (C/T) were associated with increased FAS and FASL gene expression, respectively (p < 0.005). Higher plasma IFN-γ levels were associated with higher FAS and FASL gene expression (p < 0.05). Among individuals with non-severe COVID-19, carriers of the AA genotype for FAS rs1800682 (A/G) had higher levels of FAS expression, more symptoms, and higher IFN-γ levels (p < 0.05). No association of the evaluated markers with long COVID were observed. The AA genotype of FAS rs1800682 (A/G) and the TT genotype of FASL rs763110 (C/T) influence the levels of FAS and FASL gene expression. Higher gene expression of FAS and FASL may lead to greater inflammation in COVID-19 patients, with higher levels of IFN-γ and T lymphocyte death.
Collapse
Affiliation(s)
- Wandrey Roberto dos Santos Brito
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| | - William Botelho de Brito
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
| | - Fabiane dos Santos Ferreira
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| | - Emmanuelle Giuliana Mendes Santana
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
| | - Jeferson da Costa Lopes
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| | - Ednelza da Silva Graça Amoras
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
| | - Sandra Souza Lima
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
| | - Erika Ferreira dos Santos
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Flávia Póvoa da Costa
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Kevin Matheus Lima de Sarges
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Marcos Henrique Damasceno Cantanhede
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Mioni Thieli Figueiredo Magalhães de Brito
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Andréa Luciana Soares da Silva
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Mauro de Meira Leite
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Maria de Nazaré do Socorro de Almeida Viana
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Fabíola Brasil Barbosa Rodrigues
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Rosilene da Silva
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Giselle Maria Rachid Viana
- Laboratory of Basic Research on Malaria, Parasitology Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (G.M.R.V.); (T.d.S.S.C.)
| | - Tânia do Socorro Souza Chaves
- Laboratory of Basic Research on Malaria, Parasitology Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (G.M.R.V.); (T.d.S.S.C.)
- School of Medicine, Institute of Medical Sciences, Federal University of Pará, Belém 66075-110, Brazil
| | | | | | - Daniele Freitas Henriques
- Arbovirology and Hemorrhagic Fevers Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (D.F.H.); (C.P.d.S.)
| | - Carla Pinheiro da Silva
- Arbovirology and Hemorrhagic Fevers Section, Evandro Chagas Institute, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (D.F.H.); (C.P.d.S.)
| | - Juliana Abreu Lima Nunes
- Laboratory of Immunology, Section of Virology, Instituto Evandro Chagas, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (J.A.L.N.); (I.B.C.); (I.B.-C.)
| | - Iran Barros Costa
- Laboratory of Immunology, Section of Virology, Instituto Evandro Chagas, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (J.A.L.N.); (I.B.C.); (I.B.-C.)
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua 66093-020, Brazil;
| | - Igor Brasil-Costa
- Laboratory of Immunology, Section of Virology, Instituto Evandro Chagas, Health and Environment Surveillance Secretariat, Brazilian Ministry of Health, Ananindeua 66093-020, Brazil; (J.A.L.N.); (I.B.C.); (I.B.-C.)
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua 66093-020, Brazil;
| | - Juarez Antônio Simões Quaresma
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua 66093-020, Brazil;
- Center of Biological and Health Sciences, University of the State of Pará, Belém 66087-670, Brazil;
| | - Izaura Maria Vieira Cayres-Vallinoto
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| | - Leonardo Oliveira Reis
- UroScience, Faculty of Medical Sciences, State University of Campinas, Campinas 13083-590, Brazil;
- ImmunOncology, Pontifical Catholic University of Campinas, Campinas 13060-904, Brazil
| | - Luiz Fábio Magno Falcão
- Center of Biological and Health Sciences, University of the State of Pará, Belém 66087-670, Brazil;
| | - Eduardo José Melo dos Santos
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Laboratory of Genetics of Complex Diseases, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (M.T.F.M.d.B.); (A.L.S.d.S.); (M.d.M.L.)
| | - Antonio Carlos Rosário Vallinoto
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
- Graduate Program in Virology, Evandro Chagas Institute, Department of Science, Technology, Innovation and Strategic Health Inputs, Ministry of Health of Brazil, Ananindeua 66093-020, Brazil;
| | - Maria Alice Freitas Queiroz
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (W.R.d.S.B.); (W.B.d.B.); (F.d.S.F.); (E.G.M.S.); (J.d.C.L.); (E.d.S.G.A.); (S.S.L.); (I.M.V.C.-V.); (A.C.R.V.)
- Graduate Program in Biology of Infectious and Parasitic Agents, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil; (E.F.d.S.); (F.P.d.C.); (K.M.L.d.S.); (M.H.D.C.); (M.d.N.d.S.d.A.V.); (F.B.B.R.); (R.d.S.); (E.J.M.d.S.)
| |
Collapse
|
31
|
Alicia LB, María Ángeles OG, Desirée MG, Maximino R, Marilina GA. Utility of Protein Markers in COVID-19 Patients. Int J Mol Sci 2025; 26:653. [PMID: 39859366 PMCID: PMC11766239 DOI: 10.3390/ijms26020653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
COVID-19 has been a challenge at the healthcare level not only in the early stages of the pandemic, but also in the subsequent appearance of long-term COVID-19. Several investigations have attempted to identify proteomic biomarkers in an attempt to improve clinical care, guide treatment and predict possible patient outcomes. Proteins such as C-reactive protein (CRP) or interleukin 6 (IL-6) are clear markers of severe disease, but many others have been proposed that could help in risk stratification and in the prediction of specific complications. This review aims to bring together the most relevant studies in this regard, providing information to identify the most notable biomarkers in relation to COVID-19 found to date.
Collapse
Affiliation(s)
- López-Biedma Alicia
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
| | - Onieva-García María Ángeles
- Preventive Medicine and Public Health Unit, Hospital Universitario Reina Sofia, 14004 Cordoba, Spain;
- Preventive Medicine and Public Health Research Group, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Cordoba, Spain
- Department of Medical and Surgical Sciences, University of Cordoba, 14004 Cordoba, Spain
| | - Martín-García Desirée
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain
| | - Redondo Maximino
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain
- Surgical Specialties, Biochemistry and Immunology Department, Faculty of Medicine, University of Málaga, 29010 Malaga, Spain
| | - García-Aranda Marilina
- Research and Innovation Unit, Hospital Costa del Sol, Autovía A-7 km 187, 29603 Marbella, Spain; (L.-B.A.); (M.-G.D.); (G.-A.M.)
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA-BIONAND), Severo Ochoa, 35, 29590 Malaga, Spain
- Red de Investigación en Servicios de Salud en Enfermedades Crónicas (REDISSEC) and Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud (RICAPPS), Instituto de Investigación Biomédica de Málaga (IBIMA), 29590 Malaga, Spain
| |
Collapse
|
32
|
de Sá NBR, Macieira KV, Coelho MRI, Goulart MN, Ribeiro-Alves M, Rosadas LADS, Geraldo KM, Ribeiro MPD, Cardoso SW, Grinsztejn B, Veloso VG, Cazote ADS, de Almeida DV, Giacoia-Gripp CBW, Côrtes FH, Morgado MG. COVID-19 and HIV: Clinical Outcomes and Inflammatory Markers in a Cohort from a Reference Hospital in Rio de Janeiro, Brazil. Viruses 2025; 17:91. [PMID: 39861879 PMCID: PMC11769093 DOI: 10.3390/v17010091] [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: 12/17/2024] [Revised: 01/07/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Severe COVID-19 presents a variety of clinical manifestations associated with inflammatory profiles. People living with HIV (PLWH) could face a higher risk of hospitalization and mortality from COVID-19, depending on their immunosuppression levels. This study describes inflammatory markers in COVID-19 clinical outcomes with and without HIV infection. METHODS We analyzed 112 inpatients of the Hospital Center for COVID-19 (INI/FIOCRUZ), including 22 cases of COVID-19 in PLWH (COVID/PLWH group). Plasma samples were tested for a panel of 15 cytokines by Luminex. Sociodemographic, clinical, and laboratory data were collected from patients' clinical records. RESULTS COVID-19 individuals were stratified according to the WHO clinical severity profiles at hospitalization. Significant differences in clinical scores, symptoms (coughs), and the occurrence of HIV infection were found among the groups. Clinical blood parameters and plasma cytokines were analyzed among COVID-19 groups with distinct severity profiles. Critical COVID-19 cases showed higher levels of inflammatory markers (Bilirubin, D-dimer, PCR, and urea, as well as IL-8, IL-10, TNF-α, INF-α, IL-1β, IL-17A, IL-23, IL-6) than moderate and severe groups. The COVID/PLWH group had lower CD4 counts (64 cells/mm3) and cytokine levels than other COVID-19 patients. CONCLUSIONS Overall, critically ill COVID-19 patients exhibited heightened inflammatory responses, while COVID/PLWH demonstrated unique immunological characteristics without increased mortality risk.
Collapse
Affiliation(s)
- Nathalia Beatriz Ramos de Sá
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Karine Venegas Macieira
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Mariana Rosa Inacio Coelho
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Milena Neira Goulart
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Marcelo Ribeiro-Alves
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Leonardo Azevedo da Silva Rosadas
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Kim Mattos Geraldo
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Maria Pia Diniz Ribeiro
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Sandra Wagner Cardoso
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Beatriz Grinsztejn
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Valdiléa G. Veloso
- Laboratório de Pesquisa Clínica em IST e AIDS, Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (M.R.-A.); (L.A.d.S.R.); (K.M.G.); (M.P.D.R.); (S.W.C.); (B.G.); (V.G.V.)
| | - Andressa da Silva Cazote
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Dalziza Victalina de Almeida
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Carmem Beatriz Wagner Giacoia-Gripp
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Fernanda Heloise Côrtes
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| | - Mariza Gonçalves Morgado
- Laboratório de AIDS & Imunologia Molecular, Instituto Oswaldo Cruz (IOC), FIOCRUZ, Rio de Janeiro 21040-360, Brazil; (K.V.M.); (M.R.I.C.); (M.N.G.); (A.d.S.C.); (D.V.d.A.); (C.B.W.G.-G.); (F.H.C.)
| |
Collapse
|
33
|
Gómez-Delgado I, López-Pastor AR, González-Jiménez A, Ramos-Acosta C, Hernández-Garate Y, Martínez-Micaelo N, Amigó N, Espino-Paisán L, Anguita E, Urcelay E. Long-term mitochondrial and metabolic impairment in lymphocytes of subjects who recovered after severe COVID-19. Cell Biol Toxicol 2025; 41:27. [PMID: 39792183 PMCID: PMC11723900 DOI: 10.1007/s10565-024-09976-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025]
Abstract
The underlying mechanisms explaining the differential course of SARS-CoV-2 infection and the potential clinical consequences after COVID-19 resolution have not been fully elucidated. As a dysregulated mitochondrial activity could impair the immune response, we explored long-lasting changes in mitochondrial functionality, circulating cytokine levels, and metabolomic profiles of infected individuals after symptoms resolution, to evaluate whether a complete recovery could be achieved. Results of this pilot study evidenced that different parameters of aerobic respiration in lymphocytes of individuals recuperated from a severe course lagged behind those shown upon mild COVID-19 recovery, in basal conditions and after simulated reinfection, and they also showed altered glycolytic capacity. The severe groups showed trends to enhanced superoxide production in parallel to lower OPA1-S levels. Unbalance of pivotal mitochondrial fusion (MFN2, OPA1) and fission (DRP1, FIS1) proteins was detected, suggesting a disruption in mitochondrial dynamics, as well as a lack of structural integrity in the electron transport chain. In serum, altered cytokine levels of IL-1β, IFN-α2, and IL-27 persisted long after clinical recovery, and growing amounts of the latter after severe infection correlated with lower basal and maximal respiration, ATP production, and glycolytic capacity. Finally, a trend for higher circulating levels of 3-hydroxybutyrate was found in individuals recovered after severe compared to mild course. In summary, long after acute infection, mitochondrial and metabolic changes seem to differ in a situation of full recovery after mild infection versus the one evolving from severe infection.
Collapse
Affiliation(s)
- Irene Gómez-Delgado
- Lab. Genetics and Molecular Bases of Complex Diseases, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
- Cooperative Research Networks Oriented to Health Results (RICORS, REI), ISCIII, 28029, Madrid, Spain
| | - Andrea R López-Pastor
- Lab. Genetics and Molecular Bases of Complex Diseases, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
- Cooperative Research Networks Oriented to Health Results (RICORS, REI), ISCIII, 28029, Madrid, Spain
| | - Adela González-Jiménez
- Lab. Genetics and Molecular Bases of Complex Diseases, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
- Cooperative Research Networks Oriented to Health Results (RICORS, REI), ISCIII, 28029, Madrid, Spain
| | - Carlos Ramos-Acosta
- Hematology Group, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | - Yenitzeh Hernández-Garate
- Lab. Genetics and Molecular Bases of Complex Diseases, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
| | | | - Núria Amigó
- Biosfer Teslab, 43201, Reus, Tarragona, Spain
- Department of Basic Medical Sciences, Rovira I Virgili University, 43007, Tarragona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Laura Espino-Paisán
- Lab. Genetics and Molecular Bases of Complex Diseases, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
- Cooperative Research Networks Oriented to Health Results (RICORS, REI), ISCIII, 28029, Madrid, Spain
| | - Eduardo Anguita
- Hematology Group, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain
- Department of Medicine, Medical School, Universidad Complutense de Madrid, 28040, Madrid, Spain
- Hematology Department, IML, Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - Elena Urcelay
- Lab. Genetics and Molecular Bases of Complex Diseases, Health Research Institute of Hospital Clínico San Carlos (IdISSC), 28040, Madrid, Spain.
- Cooperative Research Networks Oriented to Health Results (RICORS, REI), ISCIII, 28029, Madrid, Spain.
| |
Collapse
|
34
|
Tu TH, Bennani FE, Masroori N, Liu C, Nemati A, Rozza N, Grunbaum AM, Kremer R, Milhalcioiu C, Roy DC, Rudd CE. The identification of a SARs-CoV2 S2 protein derived peptide with super-antigen-like stimulatory properties on T-cells. Commun Biol 2025; 8:14. [PMID: 39762551 PMCID: PMC11704208 DOI: 10.1038/s42003-024-07350-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Severe COVID-19 can trigger a cytokine storm, leading to acute respiratory distress syndrome (ARDS) with similarities to superantigen-induced toxic shock syndrome. An outstanding question is whether SARS-CoV-2 protein sequences can directly induce inflammatory responses. In this study, we identify a region in the SARS-CoV-2 S2 spike protein with sequence homology to bacterial super-antigens (termed P3). Computational modeling predicts P3 binding to sites on MHC class I/II and the TCR that partially overlap with sites for the binding of staphylococcal enterotoxins B and H. Like SEB and SEH derived peptides, P3 stimulated 25-40% of human CD4+ and CD8 + T-cells, increasing IFN-γ and granzyme B production. viSNE and SPADE profiling identified overlapping and distinct IFN-γ+ and GZMB+ subsets. The super-antigenic properties of P3 were further evident by its selective expansion of T-cells expressing specific TCR Vα and Vβ chain repertoires. In vivo experiments in mice revealed that the administration of P3 led to a significant upregulation of proinflammatory cytokines IL-1β, IL-6, and TNF-α. While the clinical significance of P3 in COVID-19 remains unclear, its homology to other mammalian proteins suggests a potential role for this peptide family in human inflammation and autoimmunity.
Collapse
Affiliation(s)
- Thai Hien Tu
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
| | - Fatima Ezzahra Bennani
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Nasser Masroori
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Institut Universitaire d'Hématologie-Oncologie & Thérapie Cellulaire de Montréal, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Chen Liu
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
| | - Atena Nemati
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada
| | - Nicholas Rozza
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada
| | - Amichai Meir Grunbaum
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
| | - Richard Kremer
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
| | - Catalin Milhalcioiu
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada
- Department of Medical Oncology, McGill University Health Center, Montreal, QC, Canada
| | - Denis-Claude Roy
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada
- Institut Universitaire d'Hématologie-Oncologie & Thérapie Cellulaire de Montréal, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada
| | - Christopher E Rudd
- Department of Medicine, Universite de Montreal, Montreal, QC, Canada.
- Centre de Researche-Hopital Maisonneuve-Rosemont (CR-HMR), Montreal, QC, Canada.
- Department of Microbiology, Infection and Immunology, Universite de Montreal, Montreal, QC, Canada.
- Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco.
- Institut Universitaire d'Hématologie-Oncologie & Thérapie Cellulaire de Montréal, Hôpital Maisonneuve-Rosemont, Montreal, QC, Canada.
- Division of Experimental Medicine, Department of Medicine & Health Sciences, McGill University Health Centre, McGill University, Montreal, QC, Canada.
- Department of Medicine, McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
35
|
Balmakov Y, Mark T, Barnett I, Cipok M, Lev EI, Cohen A, Aviram E, Mayo A. Immature Platelets and Platelet Reactivity in Patients with COVID-19. Clin Appl Thromb Hemost 2025; 31:10760296251318320. [PMID: 39943824 PMCID: PMC11826839 DOI: 10.1177/10760296251318320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19) is associated with a high incidence of thromboembolic events, both venous and arterial. There are currently no specific clinical or laboratory markers to guide antithrombotic therapy for COVID-19 patients. Immature platelets represent a population of hyper-reactive platelets associated with arterial thrombotic events. This prospective study compared consecutive severe COVID-19 patients (n = 53, median age = 73 years) versus patients with sepsis from another origin (n = 41, median age = 69 years). Total platelet counts, immature platelet fraction (IPF) and immature platelet count (IPC) were determined by the Sysmex XN-3000 auto-analyzer on admission and at subsequent time-points. IPC levels three days after admission were significantly higher in the COVID-19 group compared to the sepsis group (13.4 × 109/ L [IQR 9.1-18.5] in the COVID-19 group vs 9 × 109/ L [5.5-14.7] in the sepsis group, P = 0.007). COVID-19 patients with respiratory disease show increased platelet turnover and reactivity, as seen in higher levels of immature platelet indices, especially IPC, compared to the sepsis control group. While these platelet indices remained high, CRP levels decreased, particularly in patients treated with tocilizumab. This reduction in CRP was not accompanied by any apparent clinical improvement. These findings suggest that immature platelets may serve as a biomarker for disease severity in COVID-19 patients and their CRP may not be a reliable marker for disease severity.
Collapse
Affiliation(s)
- Yulia Balmakov
- Department of Military Medicine and “Tzameret”, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel, and Medical Corps, Israel Defense Forces, Jerusalem, Israel
| | - Tomer Mark
- Intensive Care Department, Assuta Ashdod Medical Center, The Faculty of Health Sciences, Ben-Gurion University, Ashdod, Israel
- Samson Assuta Ashdod Hospital, Ashdod, Israel; Faculty of Medicine,
Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Itzik Barnett
- Intensive Care Department, Assuta Ashdod Medical Center, The Faculty of Health Sciences, Ben-Gurion University, Ashdod, Israel
- Samson Assuta Ashdod Hospital, Ashdod, Israel; Faculty of Medicine,
Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Michal Cipok
- Samson Assuta Ashdod Hospital, Ashdod, Israel; Faculty of Medicine,
Ben-Gurion University of the Negev, Be’er Sheva, Israel
- Laboratory Division, Assuta Ashdod Medical Center, The Faculty of Health Sciences, Ben-Gurion University, Ashdod, Israel
| | - Eli I. Lev
- Samson Assuta Ashdod Hospital, Ashdod, Israel; Faculty of Medicine,
Ben-Gurion University of the Negev, Be’er Sheva, Israel
- Cardiology Department, Assuta Ashdod Medical Center, The Faculty of Health Sciences, Ben-Gurion University, 7 Harefua St, Ashdod, Israel
| | - Amir Cohen
- Samson Assuta Ashdod Hospital, Ashdod, Israel; Faculty of Medicine,
Ben-Gurion University of the Negev, Be’er Sheva, Israel
- Cardiology Department, Assuta Ashdod Medical Center, The Faculty of Health Sciences, Ben-Gurion University, 7 Harefua St, Ashdod, Israel
| | - Eliad Aviram
- Samson Assuta Ashdod Hospital, Ashdod, Israel; Faculty of Medicine,
Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Ami Mayo
- Intensive Care Department, Assuta Ashdod Medical Center, The Faculty of Health Sciences, Ben-Gurion University, Ashdod, Israel
- Samson Assuta Ashdod Hospital, Ashdod, Israel; Faculty of Medicine,
Ben-Gurion University of the Negev, Be’er Sheva, Israel
| |
Collapse
|
36
|
Tripathy AS, Singh D, Trimbake D, Salwe S, Tripathy S, Kakrani A, Jali P, Chavan H, Yadav P, Sahay R, Sarje P, Babar P, Shete A, Nandapurkar A, Kulkarni M. Humoral and cellular immune response to AZD1222 /Covishield and BV152/Covaxin COVID-19 vaccines among adults in India. Hum Vaccin Immunother 2024; 20:2410579. [PMID: 39434214 PMCID: PMC11497953 DOI: 10.1080/21645515.2024.2410579] [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: 06/07/2024] [Revised: 09/06/2024] [Accepted: 09/25/2024] [Indexed: 10/23/2024] Open
Abstract
Several COVID-19 vaccines were developed using different approaches to prevent both symptomatic COVID-19 cases and fatalities. The adults were vaccinated with two doses of AZD1222/Covishield (n = 77) [manufactured by Serum Institute of India Pvt Ltd] vaccine and BV152/Covaxin (n = 99) [manufactured by Bharat Biotech] vaccine. They were assessed for immune response at pre-vaccination, 1 month post first and 6 months post second dose for anti-SARS-CoV-2 IgG antibody, surrogate neutralizing antibody (NAbs), immune phenotypes, antigen specific NK, B and T cell response, their effector functionality by ELISPOT and plasma cytokine profile. Both vaccines elicited enhanced IgG antibody and Nab levels compared to the baseline. BV152/Covaxin, the whole virus inactivated vaccine exhibited higher IgG (70% vs 100%), Nab (90% vs 100%), and robust T cell (31% vs 96%) responses at 6 months post second dose compared to 1 month post first dose justifying the utility of the second dose. Detection of SARS-CoV-2 WV and S1 specific CD4+ central T cell memory response in AZD1222/Covishield vaccinee at 6 months post second dose and higher CD4+ and CD8+ naïve and central memory T cell response in BV152/Covaxin vaccinee at 1 month post first dose indicated the involvement of memory T cells. Persistent IgG and NAb responses along with IgG+B and IgG+memory B cells in AZD1222/Covishield recipients at 6 months post second dose indicated sustained immune memory response. Continued heightened IFN-γ secreting T cell response (ELISPOT) displayed by both the vaccine platforms could serve as a co correlate of protection, further to evaluation in follow up studies. Overall, our data suggest that coordinated functions of humoral and cellular branches of adaptive immunity may pave ways toward protective immunity against COVID-19.
Collapse
Affiliation(s)
| | | | | | | | - Srikanth Tripathy
- Dr. D. Y. Patil Medical College, Hospital & Research Centre, Pune, India
| | - Arjun Kakrani
- Dr. D. Y. Patil Medical College, Hospital & Research Centre, Pune, India
| | - Priyanka Jali
- Dr. D. Y. Patil Medical College, Hospital & Research Centre, Pune, India
| | - Hanmant Chavan
- Dr. D. Y. Patil Medical College, Hospital & Research Centre, Pune, India
| | - Pragya Yadav
- ICMR-National Institute of Virology, Pune, India
| | - Rima Sahay
- ICMR-National Institute of Virology, Pune, India
| | | | - Prasad Babar
- ICMR-National Institute of Virology, Pune, India
| | - Anita Shete
- ICMR-National Institute of Virology, Pune, India
| | | | | |
Collapse
|
37
|
Vazquez-Alejo E, De La Sierra Espinar-Buitrago M, Magro-Lopez E, Tarancon-Diez L, Díez C, Bernardino JI, Rull A, De Los Santos I, Alonso R, Zamora A, Jiménez JL, Muñoz-Fernández MÁ. Deciphering long-term immune effects of HIV-1/SARS-CoV-2 co-infection: a longitudinal study. Med Microbiol Immunol 2024; 214:4. [PMID: 39724280 DOI: 10.1007/s00430-024-00813-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024]
Abstract
INTRODUCTION While the general immune response to Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) is well-understood, the long-term effects of Human Immunodeficiency Virus-1/Severe Acute Respiratory Syndrome-Coronavirus-2 (HIV-1/SARS-CoV-2) co-infection on the immune system remain unclear. This study investigates the immune response in people with HIV-1 (PWH) co-infected with SARS-CoV-2 to understand its long-term health consequences. METHODS A retrospective longitudinal study of PWH with suppressed viral load and SARS-CoV-2 infection was conducted. Cryopreserved peripheral blood mononuclear cells and plasma samples were collected at three time-points: HIV-1/pre-SARS-CoV-2 (n = 18), HIV-1/SARS-CoV-2 (n = 46), and HIV-1/post-SARS-CoV-2 (n = 36). Plasma levels of 25 soluble cytokines and chemokines, and anti-S/anti-N-IgG-SARS-CoV-2 antibodies were measured. Immunophenotyping of innate and adaptive immune components and HIV-1 and SARS-CoV-2-specific T/B-cell responses were assessed by flow cytometry. RESULTS HIV-1/SARS-CoV-2 co-infection was associated with long-lasting immune dysfunction, characterized by elevated levels of pro-inflammatory cytokines and a decrease in the MIG-IP10-ITAC chemokine axis at the HIV/SARS-CoV-2 time-point, which persisted one year later. Additionally, alterations in the distribution of subsets and increased activation (NKG2D/NKG2C) and maturation (TIM3) markers of NK and dendritic cells were observed at the HIV-1/SARS-CoV-2 time-point, persisting throughout the study. Effector memory CD4 T-cell subsets were decreased, while exhaustion/senescence (PD1/TIM3/CD57) markers were elevated at all three time-points. SARS-CoV-2-specific T/B-cell responses remained stable throughout the study, while HIV-1-specific T-cell responses decreased at the HIV-1/SARS-CoV-2 time-point and remained so. CONCLUSIONS Persistent immune dysfunction in HIV-1/SARS-CoV-2 co-infection increases the risk of future complications, even in PWH with mild symptoms. Exacerbated inflammation and alterations in immune cells may contribute to reduce vaccine efficacy and potential reinfections.
Collapse
Affiliation(s)
- Elena Vazquez-Alejo
- Immunology Section, Molecular Immuno-Biology Laboratory, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María De La Sierra Espinar-Buitrago
- Immunology Section, Molecular Immuno-Biology Laboratory, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Faculty of Pharmacy, Universidad Alfonso X el Sabio, Madrid, Spain
| | - Esmeralda Magro-Lopez
- Immunology Section, Molecular Immuno-Biology Laboratory, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Laura Tarancon-Diez
- Pediatric Infectious Diseases Unit, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Cristina Díez
- HIV and Infectious Diseases Unit, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - José Ignacio Bernardino
- HIV and Infectious Diseases Unit, Hospital Universitario La Paz, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Anna Rull
- Hospital Universitari de Tarragona Joan XXIII, Institut Investigació Sanitària Pere Virgili (IISPV), Tarragona, Spain
- Universitat Rovira i Virgili, Tarragona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Ignacio De Los Santos
- Infectious Diseases Unit, Hospital Universitario de La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Roberto Alonso
- Microbiology Section, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Angielys Zamora
- Biochemistry Section, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - José Luis Jiménez
- Immunology Section, Molecular Immuno-Biology Laboratory, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Spanish HIV-HGM BioBank, Madrid, Spain
| | - Mª Ángeles Muñoz-Fernández
- Immunology Section, Molecular Immuno-Biology Laboratory, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
- Spanish HIV-HGM BioBank, Madrid, Spain.
| |
Collapse
|
38
|
Liu P, Wu Q, Li M. Efficacy of Ulinastatin in the Treatment of COVID-19: A Retrospective Study. Int J Gen Med 2024; 17:6421-6430. [PMID: 39735165 PMCID: PMC11681785 DOI: 10.2147/ijgm.s486434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/03/2024] [Indexed: 12/31/2024] Open
Abstract
Purpose This retrospective study aimed to evaluate the efficacy of ulinastatin in the treatment of COVID-19 patients compared to conventional therapy. Patients and Methods A total of 437 COVID-19 patients admitted to the Respiratory Oncology Department of our hospital between December 31, 2022, and July 8, 2023, were included in the study. Patients were classified into the observation group (n=62) receiving ulinastatin in addition to standard treatment and the control group (n=347) receiving standard treatment only. Clinical information, laboratory results, and treatment outcomes were collected and analyzed. Results The observation group showed an improvement in lymphocyte count compared to the control group. The clinical improvement rate in patients receiving ulinastatin for 7 days or longer was 92.1%, significantly higher than that of patients treated for less than 7 days (62.5%) and those receiving standard treatment (71.0%). No significant difference in total length of hospitalization was observed between the two groups, and no related adverse events occurred in either group. Conclusion Ulinastatin treatment improves lymphocyte counts in severe COVID-19 patients, and the clinical improvement rate is significantly higher with treatment duration of 7 days or longer. Larger-scale randomized controlled trials are warranted to further explore the role of ulinastatin in the management of COVID-19.
Collapse
Affiliation(s)
- Peng Liu
- School of Graduate, Tianjin Medical University, Tianjin, People’s Republic of China
- Department of Respiratory and Critical Care Medicine, Cangzhou Fifth Hospital (People’s Hospital of Qingxian), Cangzhou, People’s Republic of China
| | - Qi Wu
- Department of Respiratory Medicine, General Hospital of Tianjin Medical University, Tianjin, People’s Republic of China
| | - Mengjie Li
- Department of Respiratory and Critical Care Medicine, Cangzhou Fifth Hospital (People’s Hospital of Qingxian), Cangzhou, People’s Republic of China
| |
Collapse
|
39
|
An Y, He L, Xu X, Piao M, Wang B, Liu T, Cao H. Gut microbiota in post-acute COVID-19 syndrome: not the end of the story. Front Microbiol 2024; 15:1500890. [PMID: 39777148 PMCID: PMC11703812 DOI: 10.3389/fmicb.2024.1500890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has led to major global health concern. However, the focus on immediate effects was assumed as the tip of iceberg due to the symptoms following acute infection, which was defined as post-acute COVID-19 syndrome (PACS). Gut microbiota alterations even after disease resolution and the gastrointestinal symptoms are the key features of PACS. Gut microbiota and derived metabolites disorders may play a crucial role in inflammatory and immune response after SARS-CoV-2 infection through the gut-lung axis. Diet is one of the modifiable factors closely related to gut microbiota and COVID-19. In this review, we described the reciprocal crosstalk between gut and lung, highlighting the participation of diet and gut microbiota in and after COVID-19 by destroying the gut barrier, perturbing the metabolism and regulating the immune system. Therefore, bolstering beneficial species by dietary supplements, probiotics or prebiotics and fecal microbiota transplantation (FMT) may be a novel avenue for COVID-19 and PACS prevention. This review provides a better understanding of the association between gut microbiota and the long-term consequences of COVID-19, which indicates modulating gut dysbiosis may be a potentiality for addressing this multifaceted condition.
Collapse
Affiliation(s)
| | | | | | | | | | - Tianyu Liu
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin, China
| | - Hailong Cao
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
40
|
Lin XB, Yao YZ, Wen QR, Liu FB, Cai YX, Chen RH, Han J. Calceolarioside B inhibits SARS-CoV-2 Omicron BA.2 variant cell entry and modulates immune response. Virol J 2024; 21:329. [PMID: 39707427 DOI: 10.1186/s12985-024-02566-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/31/2024] [Indexed: 12/23/2024] Open
Abstract
This study evaluated the inhibitory effects of calceolarioside B, extracted from the traditional Chinese herb Mutong (Akebia quinata Thumb), on the SARS-CoV-2 Omicron BA.2 variant. Molecular docking and molecular dynamics simulations predicted the binding sites and interactions between calceolarioside B and the Omicron BA.2 spike (S) protein. Biolayer interferometry (BLI) and immunofluorescence assays validated its high-affinity binding. Pseudovirus entry assays assessed the inhibitory effects of calceolarioside B on viral entry into host cells, while enzyme-linked immunosorbent assay (ELISA) measured inflammatory cytokine levels. Flow cytometry was used to analyze its effects on macrophage phenotype switching. Results demonstrated that calceolarioside B could bind to the Omicron BA.2 S protein with high affinity, and significantly inhibited viral entry into host cells by interfering with the binding of angiotensin-converting enzyme 2 (ACE2) receptor and S protein. Additionally, calceolarioside B reduced IL(interleukin)-6 expression levels and promoted the switch of macrophages from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype. These findings suggest that calceolarioside B possesses antiviral and immunomodulatory effects, making it a potential dual-function inhibitor for the treatment of COVID-19.
Collapse
Affiliation(s)
- Xiao-Bin Lin
- Department of Thyroid and Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong Province, China
| | - Yu-Zhi Yao
- Department of Thyroid and Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong Province, China
- Department of Paediatric Surgery Clinic, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong Province, China
| | - Qi-Rong Wen
- Department of Gynecologic Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong Province, China
| | - Fu-Bin Liu
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong Province, China
| | - Yuan-Xuan Cai
- Department of Thyroid and Breast Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong Province, China
| | - Rui-Hong Chen
- Department of Clinical Immunology, Institute of Clinical Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, 523000, Guangdong Province, China.
| | - Jin Han
- Prenatal Diagnosis Center, Guangzhou Women and Children's Medical Center,, Guangzhou Medical University, Guangzhou, 510623, Guangdong Province, China.
| |
Collapse
|
41
|
Keskin Sarıtaş Ç, Özsüt H, Benli A, Başaran S. Examination of Risk Factors Affecting the Development of BSI and Mortality in Critically Ill COVID-19 Patients Hospitalized in Intensive Care Unit (ICU): A Single-Center Retrospective Study. J Intensive Care Med 2024:8850666241305347. [PMID: 39704100 DOI: 10.1177/08850666241305347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
BACKGROUND Various studies have shown that the incidence of BSI is greater in COVID-19 patients hospitalized in the intensive care unit (ICU). AIMS Our study aimed to determine the risk factors for BSI, mortality rates, and factors affecting mortality in adult COVID-19 patients hospitalized in the ICU. METHODS All COVID-19 patients who met the study criteria and stayed in intensive care for more than 2 days at a tertiary university hospital during the two-year pandemic period were included in the study. Logistic regression analysis was used to determine the risk factors for BSI and mortality. RESULTS We found that respiratory rate (RR) ≥ 30 breaths per minute at the time of admission [OR: 2.342 (95% CI: 1.12-4.897)] and antibiotic use in the month before admission ICU [OR: 3.137 (95% CI: 1.321-7.451)] were independent risk factors for BSI in COVID-19 patients. Subanalysis was also performed according to the doses of immunomodulators such as anakinra, tocilizumab, and corticosteroids, and it was found that they had no effect on the BSI (P > .05). The predominant causative pathogens were K. pneumoniae, A. baumannii and enterococci. The multidrug resistant rate among bacteria was 78%. Although their comorbidities and disease severity at the time of ICU admission were similar, patients with BSIs had a higher mortality rate (58.1 to 81.9%, P = .000). The SAPS-2 score at ICU admission [OR: 3.095 (95% CI: 1.969-4.865)] and mechanical ventilation requirement throughout the ICU admission [OR: 9.314 (95% CI: 3.878-22.37)] were found to be independent risk factors for mortality by multivariate analysis. BSI was not found to be a risk factor for mortality (> .05). CONCLUSIONS Antibiotic use in patients with severe COVID-19 significantly increases the risk of BSI; unnecessary antibiotic use should be avoided.
Collapse
Affiliation(s)
- Çağla Keskin Sarıtaş
- Department of Infectious Diseases and Clinical Microbiology, Marmara University Training and Research Hospital, Istanbul, Turkey
| | - Halit Özsüt
- Department of Infectious Diseases and Clinical Microbiology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Aysun Benli
- Department of Infectious Diseases and Clinical Microbiology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Seniha Başaran
- Department of Infectious Diseases and Clinical Microbiology, Istanbul University Istanbul Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
42
|
Nakayama EE, Shioda T. Detrimental Effects of Anti-Nucleocapsid Antibodies in SARS-CoV-2 Infection, Reinfection, and the Post-Acute Sequelae of COVID-19. Pathogens 2024; 13:1109. [PMID: 39770368 PMCID: PMC11728538 DOI: 10.3390/pathogens13121109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Antibody-dependent enhancement (ADE) is a phenomenon in which antibodies enhance subsequent viral infections rather than preventing them. Sub-optimal levels of neutralizing antibodies in individuals infected with dengue virus are known to be associated with severe disease upon reinfection with a different dengue virus serotype. For Severe Acute Respiratory Syndrome Coronavirus type-2 infection, three types of ADE have been proposed: (1) Fc receptor-dependent ADE of infection in cells expressing Fc receptors, such as macrophages by anti-spike antibodies, (2) Fc receptor-independent ADE of infection in epithelial cells by anti-spike antibodies, and (3) Fc receptor-dependent ADE of cytokine production in cells expressing Fc receptors, such as macrophages by anti-nucleocapsid antibodies. This review focuses on the Fc receptor-dependent ADE of cytokine production induced by anti-nucleocapsid antibodies, examining its potential role in severe COVID-19 during reinfection and its contribution to the post-acute sequelae of COVID-19, i.e., prolonged symptoms lasting at least three months after the acute phase of the disease. We also discuss the protective effects of recently identified anti-spike antibodies that neutralize Omicron variants.
Collapse
Affiliation(s)
| | - Tatsuo Shioda
- Department of Viral Infections, Research Institute for Microbial Diseases, Osaka University, Suita 565-0871, Japan;
| |
Collapse
|
43
|
Nguyen CV, Luong CQ, Dao CX, Nguyen MH, Pham DT, Khuat NH, Pham QT, Hoang DT, Nguyen AD, Nguyen PM, Cao DD, Pham DT, Nguyen TQ, Nong VM, Dang DT, Nguyen DT, Nguyen VD, Le TQ, Nguyen VK, Ngo HD, Nguyen DV, Pham TT, Nguyen DT, Nguyen NT, Do TD, Huynh NT, Phan NT, Nguyen CD, Vo KH, Vu TT, Do CD, Dang TQ, Vu GV, Nguyen TC, Do SN. Predictive validity of interleukin 6 (IL-6) for the mortality in critically ill COVID-19 patients with the B.1.617.2 (Delta) variant in Vietnam: a single-centre, cross-sectional study. BMJ Open 2024; 14:e085971. [PMID: 39653572 PMCID: PMC11628983 DOI: 10.1136/bmjopen-2024-085971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024] Open
Abstract
OBJECTIVES To investigate the serum IL-6 levels and their rate of change in predicting the mortality of critically ill patients with COVID-19 in Vietnam. DESIGN A single-centre, cross-sectional study. SETTING An Intensive Care Centre for the Treatment of Critically Ill Patients with COVID-19 in Ho Chi Minh City, Vietnam. PARTICIPANTS We included patients aged 18 years or older who were critically ill with COVID-19 and presented to the study centre from 30 July 2021 to 15 October 2021. We excluded patients who did not have serum IL-6 measurements between admission and the end of the first day. PRIMARY OUTCOME MEASURES The primary outcome was hospital all-cause mortality. RESULTS Of 90 patients, 41.1% were men, the median age was 60.5 years (Q1-Q3: 52.0-71.0), and 76.7% of patients died in the hospital. Elevated IL-6 levels were observed on admission (41.79 pg/mL; Q1-Q3: 20.68-106.27) and on the third day after admission (72.00 pg/mL; Q1-Q3: 26.98-186.50), along with a significant rate of change in IL-6 during that period (839.5%; SD: 2753.2). While admission IL-6 level (areas under the receiver operator characteristic curve (AUROC): 0.610 (95% CI: 0.459 to 0.761); cut-off value ≥15.8 pg/mL) and rate of change in IL-6 on the third day of admission (AUROC: 0.586 (95% CI: 0.420 to 0.751); cut-off value ≥-58.7%) demonstrated poor discriminatory ability in predicting hospital mortality, the third day IL-6 rate of change from admission ≥-58.7% (adjusted OR: 12.812; 95% CI: 2.104 to 78.005) emerged as an independent predictor of hospital mortality. CONCLUSIONS This study focused on a highly selected cohort of critically ill COVID-19 patients with a high IL-6 level and mortality rate. Despite the poor discriminatory value of admission IL-6 levels, the rate of change in IL-6 proved valuable in predicting mortality. To identify critically ill COVID-19 patients with the highest risk for mortality, monitoring the serial serum IL-6 measurements and observing the rate of change in serum IL-6 levels over time are needed.
Collapse
Affiliation(s)
- Chi Van Nguyen
- Center for Emergency Medicine, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
| | - Chinh Quoc Luong
- Center for Emergency Medicine, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Co Xuan Dao
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| | - My Ha Nguyen
- Department of Health Organization and Management, Faculty of Public Health, Thai Binh University of Medicine and Pharmacy, Thai Binh, Viet Nam
| | - Dung Thi Pham
- Department of Nutrition and Food Safety, Faculty of Public Health, Thai Binh University of Medicine and Pharmacy, Thai Binh, Viet Nam
| | - Nhung Hong Khuat
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Intensive Care and Poison Control, Duc Giang General Hospital, Hanoi, Viet Nam
| | - Quynh Thi Pham
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Intensive Care Unit, University Medical Center Ho Chi Minh City, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Viet Nam
| | - Dat Tien Hoang
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| | - Anh Diep Nguyen
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Intensive Care Unit, Hanoi Heart Hospital, Hanoi, Viet Nam
| | - Phuong Minh Nguyen
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Intensive Care Unit, Thanh Nhan General Hospital, Hanoi, Viet Nam
| | - Duong Dai Cao
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Intensive Care and Poison Control, Ha Dong General Hospital, Hanoi, Viet Nam
| | - Dung Thuy Pham
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Stroke Center, Bach Mai Hospital, Hanoi, Viet Nam
| | - Thai Quoc Nguyen
- Center for Tropical Diseases, Bach Mai Hospital, Hanoi, Viet Nam
| | - Vuong Minh Nong
- Center for Tropical Diseases, Bach Mai Hospital, Hanoi, Viet Nam
| | - Dung Tuan Dang
- Center for Emergency Medicine, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Dat Tuan Nguyen
- Center for Emergency Medicine, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Vinh Duc Nguyen
- Center for Emergency Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| | - Thuan Quang Le
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Poison Control Center, Bach Mai Hospital, Hanoi, Viet Nam
| | - Viet Khoi Nguyen
- Radiology Centre, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Radiology, Hanoi Medical University, Hanoi, Viet Nam
| | - Hung Duc Ngo
- Center for Emergency Medicine, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
| | - Dung Van Nguyen
- Center for Tropical Diseases, Bach Mai Hospital, Hanoi, Viet Nam
| | - Thach The Pham
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| | - Dung Tien Nguyen
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Poison Control Center, Bach Mai Hospital, Hanoi, Viet Nam
| | - Nguyen Trung Nguyen
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
- Poison Control Center, Bach Mai Hospital, Hanoi, Viet Nam
| | - Tan Dang Do
- Radiology Centre, Bach Mai Hospital, Hanoi, Viet Nam
| | - Nhung Thi Huynh
- Department of Internal Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Nga Thu Phan
- Department of Health Organization and Management, Faculty of Public Health, Thai Binh University of Medicine and Pharmacy, Thai Binh, Viet Nam
| | - Cuong Duy Nguyen
- Department of Emergency and Critical Care Medicine, Thai Binh University of Medicine and Pharmacy, Thai Binh, Viet Nam
| | - Khoi Hong Vo
- Department of Neuro Intensive Care and Emergency Neurology, Neurology Center, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Neurology, Hanoi Medical University, Hanoi, Viet Nam
- Department of Neurology, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Thom Thi Vu
- Department of Basic Medical Sciences, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Cuong Duy Do
- Center for Tropical Diseases, Bach Mai Hospital, Hanoi, Viet Nam
- Department of Infectious Diseases, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
| | - Tuan Quoc Dang
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| | - Giap Van Vu
- Department of Internal Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Respiratory Center, Bach Mai Hospital, Hanoi, Viet Nam
| | - Tan Cong Nguyen
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| | - Son Ngoc Do
- Department of Emergency and Critical Care Medicine, Hanoi Medical University, Hanoi, Viet Nam
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
- Center for Critical Care Medicine, Bach Mai Hospital, Hanoi, Viet Nam
| |
Collapse
|
44
|
Cylwik B, Gan K, Kazberuk M, Gruszewska E, Panasiuk A, Chrostek L. Diagnostic Usefulness of Serum Hyaluronic Acid in Patients with SARS-CoV-2 Infection. J Clin Med 2024; 13:7471. [PMID: 39685929 DOI: 10.3390/jcm13237471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/06/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objective: The aim of our study is to comprehensively assess the diagnostic usefulness of serum hyaluronic acid (HA) determination in COVID-19 patients. Methods: The study group included 87 patients with COVID-19 disease and 45 healthy subjects. The HA concentration was measured using the immunochemical method. Results: The serum HA concentration was significantly higher in the COVID-19 patients before admission to hospital than that in the controls (p < 0.001). Differences were found in HA levels between the groups categorized according to disease severity (p = 002), being significantly higher in patients with critical as compared to moderate disease severity (p < 0.001). The HA concentration varied depending on the type of oxygen therapy (p = 0.004). It was significantly higher in patients on a ventilator than in those without oxygen therapy (p = 0.002). In patients who qualified for the steroid treatment and immunotherapy, the HA levels were significantly higher compared to those who did not qualify for such therapies (p = 0.043, p = 0.049, respectively). The HA levels were significantly higher in patients with cytokine storm compared to those without it (p < 0.001) and were significantly more elevated in non-survivors than in survivors (p < 0.001). HA had an excellent diagnostic power (AUC = 0.994) with sensitivity (83.3%) and specificity (97.8%) in identifying patients with critical disease severity and an excellent diagnostic power (AUC = 0.932) with sensitivity (88.2%) and specificity (95.6%) in identifying non-surviving patients. Conclusions: In summary, the results of our study indicate that HA is closely associated with severe SARS-CoV-2 infection and could be used as a novel serum biomarker to predict the risk of disease progression and as a predictor of COVID-19 mortality.
Collapse
Affiliation(s)
- Bogdan Cylwik
- Department of Paediatric Laboratory Diagnostics, Medical University of Bialystok, 15-274 Bialystok, Poland
| | - Kacper Gan
- Department of Gastroenterology, Hepatology and Internal Diseases, Provincial Welded Hospital, 15-278 Bialystok, Poland
| | - Marcin Kazberuk
- Department of Gastroenterology, Hepatology and Internal Diseases, Provincial Welded Hospital, 15-278 Bialystok, Poland
| | - Ewa Gruszewska
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Anatol Panasiuk
- Department of Gastroenterology, Hepatology and Internal Diseases, Provincial Welded Hospital, 15-278 Bialystok, Poland
- Department of Clinical Medicine, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Lech Chrostek
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
45
|
Wang J, Li X, Ren J, Rao Y, Qiao Y, Sun L, Liang Y, Chang C, Zhou Q, Sun Y. The Association of Blood Eosinophils and Neutrophils Expressing Eosinophilic Surface Markers with the Severity and Outcome of COVID-19. Microorganisms 2024; 12:2503. [PMID: 39770705 PMCID: PMC11727756 DOI: 10.3390/microorganisms12122503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 01/16/2025] Open
Abstract
(1) Background: The implication of type 2 (T2) inflammatory response in COVID-19 remains controversial. This study aimed to evaluate the association of eosinophils, neutrophils expressing eosinophilic surface markers and T2 cytokines with the severity and outcome of COVID-19. (2) Methods: Patients who were admitted to hospital due to COVID-19 from 18 December 2022 to 31 January 2023 were enrolled. Peripheral blood WBC and differentials, T2 cellular markers (subsets of eosinophils and neutrophils expressing eosinophilic surface markers) and cytokines at admission were measured and compared between subjects with different disease severities and outcomes. (3) Results: Ten mild-to-moderate and 22 severe-to-very severe cases were enrolled for analysis. Of these patients, seven died of severe-to-very severe disease. The severe-to-very severe patients showed a higher number of neutrophils, but lower numbers of eosinophils, lymphocytes cells and neutrophils expressing eosinophilic surface markers. Similarly, deceased cases were also characterized by increased neutrophils, but decreased eosinophils and neutrophils expressing eosinophilic surface markers. The levels of T2 cytokines failed to demonstrate a significant correlation with the severity or outcome of COVID-19. (4) Conclusions: Eosinophils and neutrophils expressing eosinophilic surface markers were associated with milder disease and better outcomes of COVID-19, suggesting that a T2 inflammatory response may confer a potential protective effect against the disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Research Center for Chronic Airway Diseases, Peking University Health Science Center, Beijing 100191, China; (J.W.)
| |
Collapse
|
46
|
Subbarayan K, Al-Samadi A, Schäfer H, Massa C, Salo T, Biehl K, Vaxevanis CK, Ulagappan K, Wahbi W, Reimers M, Drexler F, Moreira-Soto A, Bachmann M, Seliger B. Altered ACE2 and interferon landscape in the COVID-19 microenvironment correlate with the anti-PD-1 response in solid tumors. Cell Mol Life Sci 2024; 81:473. [PMID: 39625479 PMCID: PMC11615173 DOI: 10.1007/s00018-024-05520-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/03/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024]
Abstract
Angiotensensin-converting enzyme-2 (ACE2) is a receptor for SARS-CoV-2, allowing the virus to enter cells. Although tumor patients infected by SARS-CoV-2 often have a worse outcome, the expression, function and clinical relevance of ACE2 in tumors has not yet been thoroughly analyzed. In this study, RNA sequencing (RNA-seq) data from tumors, adjacent tissues and whole blood samples of COVID-19 patients from genome databases and from tumor cell lines and endothelial cells infected with different SARS-CoV-2 variants or transfected with an ACE2 expression vector (ACE2high) or mock (ACE2low) were analyzed for the expression of ACE2 and immune response relevant molecules in silico or by qPCR, flow cytometry, Western blot and/or RNA-seq. The differential expression profiles in ACE2high vs. ACE2low cells correlated with available SARS-CoV-2 RNA-seq datasets. ACE2high cells demonstrated upregulated mRNA and/or protein levels of HLA class I, programmed death ligand 1 (PD-L1), components of the antigen processing machinery (APM) and the interferon (IFN) signaling pathway compared to ACE2low cells. Co-cultures of ACE2high cells with peripheral blood mononuclear cells increased immune cell migration and infiltration towards ACE2high cells, apoptosis of ACE2high cells, release of innate immunity-related cytokines and altered NK cell-mediated cytotoxicity. Thus, ACE2 expression was associated in different model systems and upon SARS-CoV-2 infection with an altered host immunogenicity, which might influence the efficacy of immune checkpoint inhibitors. These results provide novel insights into the (patho)physiological role of ACE2 on immune response-relevant mechanisms and suggest an alternative strategy to reduce COVID-19 severity in infected tumor patients targeting the ACE2-induced IFN-PD-L1 axis.
Collapse
Affiliation(s)
- Karthikeyan Subbarayan
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Ahmed Al-Samadi
- Institute of Dentistry, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Joensuu, Finland
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
| | - Helene Schäfer
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Chiara Massa
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
- Institute of Translational Immunology, Brandenburg an der Havel, Germany
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, 90014, Finland
| | - Katharina Biehl
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Christoforos K Vaxevanis
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Kamatchi Ulagappan
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | - Wafa Wahbi
- Department of Oral and Maxillofacial Diseases, Clinicum, University of Helsinki, Helsinki, Finland
| | - Matthias Reimers
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany
| | | | | | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Barbara Seliger
- Medical Faculty, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle (Saale), Germany.
- Institute of Translational Immunology, Brandenburg an der Havel, Germany.
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany.
- Institute of Translational Medicine, Medical School Theodor Fontane, Hochstr. 29, 14770, Brandenburg an der Havel, Germany.
| |
Collapse
|
47
|
Zhu R, Zhao Y, Yin H, Shu L, Ma Y, Tao Y. Identification of immune-related hub genes and potential molecular mechanisms involved in COVID-19 via integrated bioinformatics analysis. Sci Rep 2024; 14:29964. [PMID: 39622956 PMCID: PMC11612211 DOI: 10.1038/s41598-024-81803-2] [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: 07/08/2024] [Accepted: 11/29/2024] [Indexed: 12/06/2024] Open
Abstract
COVID-19, caused by the SARS-CoV-2 virus, poses significant health challenges worldwide, particularly due to severe immune-related complications. Understanding the molecular mechanisms and identifying key immune-related genes (IRGs) involved in COVID-19 pathogenesis is critical for developing effective prevention and treatment strategies. This study employed computational tools to analyze biological data (bioinformatics) and a method for inferring causal relationships based on genetic variations, known as Mendelian randomization (MR), to explore the roles of IRGs in COVID-19. We identified differentially expressed genes (DEGs) from datasets available in the Gene Expression Omnibus (GEO), comparing COVID-19 patients with healthy controls. IRGs were sourced from the ImmPort database. We conducted functional enrichment analysis, pathway analysis, and immune infiltration assessments to determine the biological significance of the identified IRGs. A total of 360 common differential IRGs were identified. Among these genes, CD1C, IL1B, and SLP1 have emerged as key IRGs with potential protective effects against COVID-19. Pathway enrichment analysis revealed that CD1C is involved in terpenoid backbone biosynthesis and Th17 cell differentiation, while IL1B is linked to B-cell receptor signaling and the NF-kappa B signaling pathway. Significant correlations were observed between key genes and various immune cells, suggesting that they influence immune cell modulation in COVID-19. This study provides new insights into the immune mechanisms underlying COVID-19, highlighting the crucial role of IRGs in disease progression. These findings suggest that CD1C and IL1B could be potential therapeutic targets. The integrated bioinformatics and MR analysis approach offers a robust framework for further exploring immune responses in COVID-19 patients, as well as for targeted therapy and vaccine development.
Collapse
Affiliation(s)
- Rui Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yaping Zhao
- Department of Pharmacy, Shaoxing Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Shaoxing, 312000, China
| | - Hui Yin
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | - Linfeng Shu
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yuhang Ma
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yingli Tao
- Department of Reproductive Immunology, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China.
| |
Collapse
|
48
|
Chatterjee D, Kurup D, Smeyne RJ. Environmental exposures and familial background alter the induction of neuropathology and inflammation after SARS-CoV-2 infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.02.626375. [PMID: 39677638 PMCID: PMC11642758 DOI: 10.1101/2024.12.02.626375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Basal ganglia disease has been reported as a post-infection sequela of several viruses, with documentation of this phenomenon from the H1N1 Spanish flu to the recent COVID-19 (SARS-CoV-2) pandemic. SARS-CoV-2 infection leads to multisystem deficits, including those affecting the nervous system. Here, we investigated whether a SARS-CoV-2 infection alone increases the susceptibility to develop parkinsonian phenotypes in C57BL/6J mice expressing the human ACE2 receptor, or in addition to two well-known toxin exposures, MPTP and paraquat. Additionally, we examined mice carrying a G2019S mutation in the LRRK2 gene. We also examined if vaccination with either an mRNA- or protein-based vaccine can alter any observed neuropathology. We find that the infection with the WA-1/2020 (alpha) or omicron B1.1.529 strains in ACE2 and G2019S LRRK2 mice both synergize with a subtoxic exposure to the mitochondrial toxin MPTP to induce neurodegeneration and neuroinflammation in the substantia nigra. This synergy appears toxin-dependent since we do not observe this following exposure to the direct redox-inducing compound paraquat. This synergistic neurodegeneration and neuroinflammation is rescued in WT mice that were vaccinated using either mRNA- and protein- based vaccines directed against the Spike protein of the SARS-CoV-2 virus. However, in the G2019S LRRK2 mutant mice, we find that only the protein-based vaccine but not the mRNA- based vaccine resulted in a rescue of the SARS-CoV-2 mediated neuropathology. Taken together, our results highlight the role of both environmental exposures and familial background on the development of parkinsonian pathology secondary to viral infection and the benefit of vaccines in reducing these risks.
Collapse
|
49
|
Valderrábano RJ, Wipper B, Pencina KM, Migaud M, Shang YV, Latham NK, Montano M, Cunningham JM, Wilson L, Peng L, Memish‐Beleva Y, Bhargava A, Swain PM, Lehman P, Lavu S, Livingston DJ, Bhasin S. Dysregulated nicotinamide adenine dinucleotide metabolome in patients hospitalized with COVID-19. Aging Cell 2024; 23:e14326. [PMID: 39354697 PMCID: PMC11634700 DOI: 10.1111/acel.14326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 10/03/2024] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) depletion has been postulated as a contributor to the severity of COVID-19; however, no study has prospectively characterized NAD+ and its metabolites in relation to disease severity in patients with COVID-19. We measured NAD+ and its metabolites in 56 hospitalized patients with COVID-19 and in two control groups without COVID-19: (1) 31 age- and sex-matched adults with comorbidities, and (2) 30 adults without comorbidities. Blood NAD+ concentrations in COVID-19 group were only slightly lower than in the control groups (p < 0.05); however, plasma 1-methylnicotinamide concentrations were significantly higher in patients with COVID-19 (439.7 ng/mL, 95% CI: 234.0, 645.4 ng/mL) than in age- and sex-matched controls (44.5 ng/mL, 95% CI: 15.6, 73.4) and in healthy controls (18.1 ng/mL, 95% CI 15.4, 20.8; p < 0.001 for each comparison). Plasma nicotinamide concentrations were also higher in COVID-19 group and in controls with comorbidities than in healthy control group. Plasma concentrations of 2-methyl-2-pyridone-5-carboxamide (2-PY), but not NAD+, were significantly associated with increased risk of death (HR = 3.65; 95% CI 1.09, 12.2; p = 0.036) and escalation in level of care (HR = 2.90, 95% CI 1.01, 8.38, p = 0.049). RNAseq and RTqPCR analyses of PBMC mRNA found upregulation of multiple genes involved in NAD+ synthesis as well as degradation, and dysregulation of NAD+-dependent processes including immune response, DNA repair, metabolism, apoptosis/autophagy, redox reactions, and mitochondrial function. Blood NAD+ concentrations are modestly reduced in COVID-19; however, NAD+ turnover is substantially increased with upregulation of genes involved in both NAD+ biosynthesis and degradation, supporting the rationale for NAD+ augmentation to attenuate disease severity.
Collapse
Affiliation(s)
- Rodrigo J. Valderrábano
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Benjamin Wipper
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Karol Mateusz Pencina
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Marie Migaud
- Department of Pharmacology, Mitchell Cancer InstituteUniversity of South AlabamaMobileAlabamaUSA
| | - Yili Valentine Shang
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Nancy K. Latham
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Monty Montano
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - James M. Cunningham
- Division of Hematology, Brigham and Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Lauren Wilson
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Liming Peng
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Yusnie Memish‐Beleva
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Avantika Bhargava
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | | | - Phoebe Lehman
- Metro International BiotechWorcesterMassachusettsUSA
| | - Siva Lavu
- Metro International BiotechWorcesterMassachusettsUSA
| | | | - Shalender Bhasin
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence CenterBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
50
|
Yu LCH. Gastrointestinal pathophysiology in long COVID: Exploring roles of microbiota dysbiosis and serotonin dysregulation in post-infectious bowel symptoms. Life Sci 2024; 358:123153. [PMID: 39454992 DOI: 10.1016/j.lfs.2024.123153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/01/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) triggered an unprecedented public health crisis known as the coronavirus disease 2019 (COVID-19) pandemic. Gastrointestinal (GI) symptoms develop in patients during acute infection and persist after recovery from airway distress in a chronic form of the disease (long COVID). A high incidence of irritable bowel syndrome (IBS) manifested by severe abdominal pain and defecation pattern changes is reported in COVID patients. Although COVID is primarily considered a respiratory disease, fecal shedding of SARS-CoV-2 antigens positively correlates with bowel symptoms. Active viral infection in the GI tract was identified by human intestinal organoid studies showing SARS-CoV-2 replication in gut epithelial cells. In this review, we highlight the key findings in post-COVID bowel symptoms and explore possible mechanisms underlying the pathophysiology of the illness. These mechanisms include mucosal inflammation, gut barrier dysfunction, and microbiota dysbiosis during viral infection. Viral shedding through the GI route may be the primary factor causing the alteration of the microbiome ecosystem, particularly the virome. Recent evidence in experimental models suggested that microbiome dysbiosis could be further aggravated by epithelial barrier damage and immune activation. Moreover, altered microbiota composition has been associated with dysregulated serotonin pathways, resulting in intestinal nerve hypersensitivity. These mechanisms may explain the development of post-infectious IBS-like symptoms in long COVID. Understanding how coronavirus infection affects gut pathophysiology, including microbiome changes, would benefit the therapeutic advancement for managing post-infectious bowel symptoms.
Collapse
Affiliation(s)
- Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|