1
|
DeDiego ML, Portilla Y, Daviu N, López-García D, Villamayor L, Vázquez-Utrilla P, Mulens-Arias V, Pérez-Yagüe S, Nogales A, Ovejero JG, Gallo-Cordova A, Enjuanes L, Veintemillas-Verdaguer S, Morales MP, Barber DF. Biocompatible Iron Oxide Nanoparticles Display Antiviral Activity Against Two Different Respiratory Viruses in Mice. Int J Nanomedicine 2024; 19:13763-13788. [PMID: 39723174 PMCID: PMC11669338 DOI: 10.2147/ijn.s475323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
Background Severe Acute Respiratory syndrome coronavirus 2 (SARS-CoV-2) and Influenza A viruses (IAVs) are among the most important causes of viral respiratory tract infections, causing similar symptoms. IAV and SARS-CoV-2 infections can provoke mild symptoms like fever, cough, sore throat, loss of taste or smell, or they may cause more severe consequences leading to pneumonia, acute respiratory distress syndrome or even death. While treatments for IAV and SARS-CoV-2 infection are available, IAV antivirals often target viral proteins facilitating the emergence of drug-resistant viral variants. Hence, universal treatments against coronaviruses and IAVs are hard to obtain due to genus differences (in the case of coronavirus) or subtypes (in the case of IAV), highlighting the need for novel antiviral therapies. Interestingly, iron oxide nanoparticles (IONPs) with a 10 nm core size and coated with the biocompatible dimercaptosuccinic acid (DMSA: DMSA-IONP-10) display antiviral activity against SARS-CoV-2 in vitro. Methods We analyzed the antiviral activity of DMSA-IONP-10 against SARS-CoV-2 infection in vivo, and against IAV infection in vitro and in vivo. Results DMSA-IONP-10 treatment of mice after SARS-CoV-2 infection impaired virus replication in the lungs and led to a mildly reduced pro-inflammatory cytokine induction after infection, indicating that these IONPs can serve as COVID-19 therapeutic agents. These IONPs also had a prophylactic and therapeutic effect against IAV in tissue cultured cells at non-cytotoxic doses, and a therapeutic effect in IAV-infected-mice, inhibiting viral replication and slightly dampening the inflammatory response after viral infection. As an exacerbated inflammatory response to IAVs and SARS-CoV-2 is detrimental to the host, weakening this response in mice through IONP treatment may reduce disease severity. Interestingly, our data suggest that IONP treatment affects oxidative stress and iron metabolism in cells, which may influence IAV production. Conclusion This study highlights the antiviral activity of DMSA-IONP-10 against important human respiratory viruses.
Collapse
Affiliation(s)
- Marta L DeDiego
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Yadileiny Portilla
- Department of Immunology, Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Neus Daviu
- Department of Immunology, Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Darío López-García
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Laura Villamayor
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Paula Vázquez-Utrilla
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Vladimir Mulens-Arias
- Department of Immunology, Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Sonia Pérez-Yagüe
- Department of Immunology, Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Aitor Nogales
- Center for Animal Health Research, CISA-INIA-CSIC, Madrid, Spain
| | - Jesús G Ovejero
- Department of Nanoscience and Nanotechnology, Instituto de Ciencia de Materiales de Madrid (ICMM-CSIC), Madrid, Spain
| | - Alvaro Gallo-Cordova
- Department of Nanoscience and Nanotechnology, Instituto de Ciencia de Materiales de Madrid (ICMM-CSIC), Madrid, Spain
| | - Luis Enjuanes
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Sabino Veintemillas-Verdaguer
- Department of Nanoscience and Nanotechnology, Instituto de Ciencia de Materiales de Madrid (ICMM-CSIC), Madrid, Spain
| | - M Puerto Morales
- Department of Nanoscience and Nanotechnology, Instituto de Ciencia de Materiales de Madrid (ICMM-CSIC), Madrid, Spain
| | - Domingo F Barber
- Department of Immunology, Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| |
Collapse
|
2
|
Faliti CE, Mesina M, Choi J, Bélanger S, Marshall MA, Tipton CM, Hicks S, Chappa P, Cardenas MA, Abdel-Hakeem M, Thinnes TC, Cottrell C, Scharer CD, Schief WR, Nemazee D, Woodruff MC, Lindner JM, Sanz I, Crotty S. Interleukin-2-secreting T helper cells promote extra-follicular B cell maturation via intrinsic regulation of a B cell mTOR-AKT-Blimp-1 axis. Immunity 2024; 57:2772-2789.e8. [PMID: 39612915 DOI: 10.1016/j.immuni.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/03/2024] [Accepted: 11/06/2024] [Indexed: 12/01/2024]
Abstract
During antigen-driven responses, B cells can differentiate at extra-follicular (EF) sites or initiate germinal centers (GCs) in processes that involve interactions with T cells. Here, we examined the roles of interleukin (IL)-2 secreted by T helper (Th) cells during cognate interactions with activated B cells. IL-2 boosted the expansion of EF plasma cells and the secretion of low-mutated immunoglobulin G (IgG). Conversely, genetically disrupting IL-2 expression by CD4+ T cells, or IL-2 receptor (CD25) expression by B cells, promoted B cell entry into the GC and high-affinity antibody secretion. Mechanistically, IL-2 induced early mTOR activity, expression of the transcriptional regulator IRF4, and metabolic changes in B cells required to form Blimp-1-expressing plasma cells. Thus, T cell help via IL-2 regulates an mTOR-AKT-Blimp-1 axis in activated B cells, providing insight into the mechanisms that determine EF versus GC fates and positioning IL-2 as an early switch controlling plasma cell versus GC B cell commitment.
Collapse
Affiliation(s)
- Caterina E Faliti
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Maria Mesina
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Jinyong Choi
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Microbiology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Simon Bélanger
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; VIR Biotechnology, San Francisco, CA 94158, USA
| | - Monique A Marshall
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Christopher M Tipton
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Sakeenah Hicks
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Prashanti Chappa
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | - Theresa C Thinnes
- Scripps Consortium for HIV/AIDS Vaccine Development (CHAVD), La Jolla, CA 92037, USA
| | - Christopher Cottrell
- Scripps Consortium for HIV/AIDS Vaccine Development (CHAVD), La Jolla, CA 92037, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - William R Schief
- Scripps Consortium for HIV/AIDS Vaccine Development (CHAVD), La Jolla, CA 92037, USA; Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA; The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - David Nemazee
- Scripps Consortium for HIV/AIDS Vaccine Development (CHAVD), La Jolla, CA 92037, USA
| | - Matthew C Woodruff
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | | | - Ignacio Sanz
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Scripps Consortium for HIV/AIDS Vaccine Development (CHAVD), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| |
Collapse
|
3
|
Shah SZ, Du Z, El Jack K, Pham SM, Elrefaei M. Severe SARS-CoV-2 infection is associated with increased risk of De novo HLA antibody production in lung transplant recipients: Single-center study. Transpl Immunol 2024:102161. [PMID: 39645000 DOI: 10.1016/j.trim.2024.102161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/23/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
THE PURPOSE The COVID-19 pandemic has led to significant morbidity and mortality in lung transplant recipients (LTR). Respiratory viral infections may be associated with de-novo HLA donor-specific antibody (DSA) production and impact lung transplant outcomes. Since one of the immunomodulation strategies post-SARS-CoV-2 infection in LTR include decreasing or holding anti-metabolites, concerns have been raised for higher incidence of de-novo DSA production in LTR. METHODS We performed a retrospective chart review of 63 consecutive LTR diagnosed with COVID-19 to investigate this concern. COVID-19 disease severity was divided into 3 groups: mild, moderate, and severe. Mild disease was defined as patients with COVID-19 diagnosis who were stable enough to be treated as out-patients. Moderate disease was defined as patients who required admission to the hospital and were on less than 10 l of oxygen at rest. Severe disease was identified as patients who required hospitalization and were on more than 10 l of oxygen with or without mechanical ventilation or extra corporal membrane oxygenation (ECMO). Groups were compared using the Kruskal-Wallis test. A total of 11, 43, and 9 LTR were diagnosed with mild, moderate, and severe COVID-19 respectively. RESULTS We observed no significant differences in the CPRA pre-COVID-19 compared to 1 and 6 months post-COVID-19 diagnosis in 6/11 (54.5 %), 18/43 (41.8 %), and 6/9 (66.9 %) LTR with mild (p = 0.66), moderate (p = 0.74), and severe (p = 0.22) COVID-19 respectively. HLA class I and II DSA were detected pre-COVID-19 diagnosis and persisted with no significant differences in the median MFI levels at 1 and 6 months post-COVID-19 diagnosis in 2/11 (p = 0.93), 7/43 (p = 0.71), and 0/9 LTR with mild, moderate, and severe COVID-19 respectively. De-novo HLA DSA were detected within 6 months post-COVID-19 diagnosis in 0/11 (0 %), 1/43 (2.3 %), and 3/9 (33.3 %%) LTR with mild, moderate, and severe COVID-19 respectively (p = 0.001). CONCLUSION Severe COVID-19 may be associated with increased risk of de novo HLA DSA production resulting in allograft dysfunction.
Collapse
Affiliation(s)
- Sadia Z Shah
- Department of Transplantation, Mayo Clinic, Jacksonville, FL, United States of America
| | - Zeying Du
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, United States of America
| | - Kamal El Jack
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, United States of America
| | - Si M Pham
- Department of Cardiothoracic Surgery, Mayo Clinic, Jacksonville, FL, United States of America
| | - Mohamed Elrefaei
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, United States of America.
| |
Collapse
|
4
|
Li X, Edén A, Malwade S, Cunningham JL, Bergquist J, Weidenfors JA, Sellgren CM, Engberg G, Piehl F, Gisslen M, Kumlien E, Virhammar J, Orhan F, Rostami E, Schwieler L, Erhardt S. Central and peripheral kynurenine pathway metabolites in COVID-19: Implications for neurological and immunological responses. Brain Behav Immun 2024; 124:163-176. [PMID: 39615604 DOI: 10.1016/j.bbi.2024.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/31/2024] [Accepted: 11/27/2024] [Indexed: 12/09/2024] Open
Abstract
Long-term symptoms such as pain, fatigue, and cognitive impairments are commonly observed in individuals affected by coronavirus disease 2019 (COVID-19). Metabolites of the kynurenine pathway have been proposed to account for cognitive impairment in COVID-19 patients. Here, cerebrospinal fluid (CSF) and plasma levels of kynurenine pathway metabolites in 53 COVID-19 patients and 12 non-inflammatory neurological disease controls in Sweden were measured with an ultra-performance liquid chromatography-tandem mass spectrometry system (UPLC-MS/MS) and correlated with immunological markers and neurological markers. Single cell transcriptomic data from a previous study of 130 COVID-19 patients was used to investigate the expression of key genes in the kynurenine pathway. The present study reveals that the neuroactive kynurenine pathway metabolites quinolinic acid (QUIN) and kynurenic acid (KYNA) are increased in CSF in patients with acute COVID-19. In addition, CSF levels of kynurenine, ratio of kynurenine/tryptophan (rKT) and QUIN correlate with neurodegenerative markers. Furthermore, tryptophan is significantly decreased in plasma but not in the CSF. In addition, the kynurenine pathway is strongly activated in the plasma and correlates with the peripheral immunological marker neopterin. Single-cell transcriptomics revealed upregulated gene expressions of the rate-limiting enzyme indoleamine 2,3- dioxygenase1 (IDO1) in CD14+ and CD16+ monocytes that correlated with type II-interferon response exclusively in COVID-19 patients. In summary, our study confirms significant activation of the peripheral kynurenine pathway in patients with acute COVID-19 and, notably, this is the first study to identify elevated levels of kynurenine metabolites in the central nervous system associated with the disease. Our findings suggest that peripheral inflammation, potentially linked to overexpression of IDO1 in monocytes, activates the kynurenine pathway. Increased plasma kynurenine, crossing the blood-brain barrier, serves as a source for elevated brain KYNA and neurotoxic QUIN. We conclude that blocking peripheral-to-central kynurenine transport could be a promising strategy to protect against neurotoxic effects of QUIN in COVID-19 patients.
Collapse
Affiliation(s)
- Xueqi Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Arvid Edén
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41685, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Infectious Disease, Gothenburg, 41685, Sweden
| | - Susmita Malwade
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Janet L Cunningham
- Department of Medical Science, Psychiatry, Uppsala University, Uppsala 75185, Sweden; Department of Neuroscience, Karolinska Institute, Stockholm 17177, Sweden
| | - Jonas Bergquist
- Analytical Chemistry and Neurochemistry, Department of Chemistry─BMC, Uppsala University, Box 599, 751 24 Uppsala, Sweden; The ME/CFS Collaborative Research Centre at Uppsala University, 751 24 Uppsala, Sweden
| | | | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden; Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, and Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Göran Engberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden; Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - Fredrik Piehl
- Unit of Neuroimmunology, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm 17177, Sweden; Division of Neurology, Karolinska University Hospital, Stockholm 17176, Sweden
| | - Magnus Gisslen
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41685, Sweden; Region Västra Götaland, Sahlgrenska University Hospital, Department of Infectious Disease, Gothenburg, 41685, Sweden; Public Health Agency of Sweden, Solna, Sweden
| | - Eva Kumlien
- Department of Medical Sciences, Neurology, Uppsala University, Uppsala 75185, Sweden
| | - Johan Virhammar
- Department of Medical Sciences, Neurology, Uppsala University, Uppsala 75185, Sweden
| | - Funda Orhan
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Elham Rostami
- Department of Neuroscience, Karolinska Institute, Stockholm 17177, Sweden; Department of Medical Sciences, Neurology, Uppsala University, Uppsala 75185, Sweden
| | - Lilly Schwieler
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Sophie Erhardt
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm 17177, Sweden.
| |
Collapse
|
5
|
Zhang J, Kim MH, Lee S, Park S. Integration of nanobiosensors into organ-on-chip systems for monitoring viral infections. NANO CONVERGENCE 2024; 11:47. [PMID: 39589620 PMCID: PMC11599699 DOI: 10.1186/s40580-024-00455-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/17/2024] [Indexed: 11/27/2024]
Abstract
The integration of nanobiosensors into organ-on-chip (OoC) models offers a promising advancement in the study of viral infections and therapeutic development. Conventional research methods for studying viral infection, such as two-dimensional cell cultures and animal models, face challenges in replicating the complex and dynamic nature of human tissues. In contrast, OoC systems provide more accurate, physiologically relevant models for investigating viral infections, disease mechanisms, and host responses. Nanobiosensors, with their miniaturized designs and enhanced sensitivity, enable real-time, continuous, in situ monitoring of key biomarkers, such as cytokines and proteins within these systems. This review highlights the need for integrating nanobiosensors into OoC systems to advance virological research and improve therapeutic outcomes. Although there is extensive literature on biosensors for viral infection detection and OoC models for replicating infections, real integration of biosensors into OoCs for continuous monitoring remains unachieved. We discuss the advantages of nanobiosensor integration for real-time tracking of critical biomarkers within OoC models, key biosensor technologies, and current OoC systems relevant to viral infection studies. Additionally, we address the main technical challenges and propose solutions for successful integration. This review aims to guide the development of biosensor-integrated OoCs, paving the way for precise diagnostics and personalized treatments in virological research.
Collapse
Affiliation(s)
- Jiande Zhang
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| | - Min-Hyeok Kim
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| | - Seulgi Lee
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
- Department of Metabiohealth, Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
- Department of Biophysics, Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
| |
Collapse
|
6
|
Anzurez A, Runtuwene L, Dang TTT, Nakayama-Hosoya K, Koga M, Yoshimura Y, Sasaki H, Miyata N, Miyazaki K, Takahashi Y, Suzuki T, Yotsuyanagi H, Tachikawa N, Matano T, Kawana-Tachikawa A. Characterization of the Proinflammatory Cytokine Profile during Acute SARS-CoV-2 Infection in People with Human Immunodeficiency Virus. Jpn J Infect Dis 2024; 77:301-310. [PMID: 38945856 DOI: 10.7883/yoken.jjid.2024.184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Persistent inflammation during chronic human immunodeficiency virus (HIV) infection may affect the immune response against severe acute respiratory syndrome-coronavirus 2 (SARS- CoV-2) infection. Plasma levels of multiple proinflammatory cytokines during acute SARS-CoV-2 infection were measured in people with HIV (PWH) with effective combination antiretroviral therapy. There were no significant differences in any of the measured cytokines between severity levels of coronavirus disease 2019 (COVID-19) in PWH, while most were significantly higher in HIV-uninfected individuals with severe COVID-19, suggesting that excess cytokines release by hyperinflammatory responses do not occur in individuals with severe COVID-19 with HIV infection. The strong associations between the cytokines observed in HIV-uninfected individuals, particularly between IFN-α/TNF-α and other cytokines, were lost in PWH. The steady-state plasma levels of IP-10, ICAM-1, and CD62E were significantly higher in PWH, indicating that they were in an enhanced inflammatory state. The absence of several inter-cytokine correlations was observed in in vitro lipopolysaccharide stimulus-driven cytokine production in PWH. These data suggest that inflammatory responses during SARS-CoV-2 infection in PWH are distinct from those in HIV-uninfected individuals, partially because of the underlying inflammatory state and/or impairment of innate immune cells.
Collapse
Affiliation(s)
- Alitzel Anzurez
- AIDS Research Center, National Institute of Infectious Diseases, Japan
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Japan
| | - Lucky Runtuwene
- AIDS Research Center, National Institute of Infectious Diseases, Japan
| | - Thi Thu Thao Dang
- AIDS Research Center, National Institute of Infectious Diseases, Japan
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Japan
| | | | - Michiko Koga
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Japan
| | - Yukihiro Yoshimura
- Department of Infectious Diseases, Yokohama Municipal Citizens' Hospital, Japan
| | - Hiroaki Sasaki
- Department of Infectious Diseases, Yokohama Municipal Citizens' Hospital, Japan
| | - Nobuyuki Miyata
- Department of Infectious Diseases, Yokohama Municipal Citizens' Hospital, Japan
| | - Kazuhito Miyazaki
- Department of Infectious Diseases, Yokohama Municipal Citizens' Hospital, Japan
- Department of Respiratory Medicine, Yokohama Municipal Citizens' Hospital, Japan
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Japan
| | - Hiroshi Yotsuyanagi
- Division of Infectious Diseases, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Japan
| | - Natsuo Tachikawa
- Department of Infectious Diseases, Yokohama Municipal Citizens' Hospital, Japan
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious Diseases, Japan
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Japan
- Department of AIDS Vaccine Development, Institute of Medical Science, University of Tokyo, Japan
| | - Ai Kawana-Tachikawa
- AIDS Research Center, National Institute of Infectious Diseases, Japan
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Japan
- Department of AIDS Vaccine Development, Institute of Medical Science, University of Tokyo, Japan
| |
Collapse
|
7
|
Gajate-Arenas M, García-Pérez O, Domínguez-De-Barros A, Sirvent-Blanco C, Dorta-Guerra R, García-Ramos A, Piñero JE, Lorenzo-Morales J, Córdoba-Lanús E. Differential Inflammatory and Immune Response to Viral Infection in the Upper-Airway and Peripheral Blood of Mild COVID-19 Cases. J Pers Med 2024; 14:1099. [PMID: 39590591 PMCID: PMC11595938 DOI: 10.3390/jpm14111099] [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: 10/24/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES COVID-19 is characterised by a wide variety of clinical manifestations, and clinical tests and genetic analysis might help to predict patient outcomes. METHODS In the current study, the expression of genes related to immune response (CCL5, IFI6, OAS1, IRF9, IL1B, and TGFB1) was analysed in the upper airway and paired-blood samples from 25 subjects infected with SARS-CoV-2. Relative gene expression was determined by RT-qPCR. RESULTS CCL5 expression was higher in the blood than in the upper airway (p < 0.001). In addition, a negative correlation was found between IFI6 and viral load (p = 0.033) in the upper airway, suggesting that the IFI6 expression inhibits the viral infection. Concerning sex, women expressed IL1B and IRF9 in a higher proportion than men at a systemic level (p = 0.008 and p = 0.049, respectively). However, an increased expression of IRF9 was found in men compared to women in the upper airway (p = 0.046), which could be due to the protective effect of IRF9, especially in men. CONCLUSIONS The higher expression of CCL5 in blood might be due to the key role of this gene in the migration and recruitment of immune cells from the systemic circulation to the lungs. Our findings confirm the existence of sex differences in the immune response to early stages of the infection. Further studies in a larger cohort are necessary to corroborate the current findings.
Collapse
Affiliation(s)
- Malena Gajate-Arenas
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (M.G.-A.); (O.G.-P.); (A.D.-D.-B.); (C.S.-B.); (R.D.-G.); (A.G.-R.); (J.E.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Omar García-Pérez
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (M.G.-A.); (O.G.-P.); (A.D.-D.-B.); (C.S.-B.); (R.D.-G.); (A.G.-R.); (J.E.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Angélica Domínguez-De-Barros
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (M.G.-A.); (O.G.-P.); (A.D.-D.-B.); (C.S.-B.); (R.D.-G.); (A.G.-R.); (J.E.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Candela Sirvent-Blanco
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (M.G.-A.); (O.G.-P.); (A.D.-D.-B.); (C.S.-B.); (R.D.-G.); (A.G.-R.); (J.E.P.)
| | - Roberto Dorta-Guerra
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (M.G.-A.); (O.G.-P.); (A.D.-D.-B.); (C.S.-B.); (R.D.-G.); (A.G.-R.); (J.E.P.)
- Departamento de Matemáticas, Estadística e Investigación Operativa, Facultad de Ciencias, Sección de Matemáticas, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| | - Alma García-Ramos
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (M.G.-A.); (O.G.-P.); (A.D.-D.-B.); (C.S.-B.); (R.D.-G.); (A.G.-R.); (J.E.P.)
| | - José E. Piñero
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (M.G.-A.); (O.G.-P.); (A.D.-D.-B.); (C.S.-B.); (R.D.-G.); (A.G.-R.); (J.E.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Facultad de Ciencias de la Salud, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| | - Jacob Lorenzo-Morales
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (M.G.-A.); (O.G.-P.); (A.D.-D.-B.); (C.S.-B.); (R.D.-G.); (A.G.-R.); (J.E.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Obstetricia y Ginecología, Pediatría, Medicina Preventiva y Salud Pública, Toxicología, Medicina Legal y Forense y Parasitología, Facultad de Ciencias de la Salud, Universidad de La Laguna, 38200 La Laguna, Tenerife, Spain
| | - Elizabeth Córdoba-Lanús
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, 38029 La Laguna, Tenerife, Spain; (M.G.-A.); (O.G.-P.); (A.D.-D.-B.); (C.S.-B.); (R.D.-G.); (A.G.-R.); (J.E.P.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
8
|
Huang Y, Fang Y, Jie H, Yang H, Zhou W, Chen Y, Zhong B. Network pharmacology and molecular docking to scientifically validate the potential mechanism of Lonicerae japonicae flos in the clinical treatment of COVID-19. Nat Prod Res 2024; 38:3699-3706. [PMID: 37732603 DOI: 10.1080/14786419.2023.2260070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/10/2023] [Indexed: 09/22/2023]
Abstract
Using network pharmacology and molecular docking, we predicted the potential mechanisms of Lonicerae japonicae flos (LJF) therapy for COVID-19. A total of 493 component-related targets and 6,233 COVID-19-related genes were identified, and 267 core genes with overlapping of the two types of genes were identified. The target AKT1, CASP3, IL1B, IL6, PTGS2, TNF and JUN were the hub genes in PPI network according to MCODE score. Component-Target analysis showed the close relationship between targets and components. The results of functional enrichment analyses revealed that LJF exerted pharmacological effects on COVID-19 by regulating IL-17 signalling pathway, TNF signalling pathway, AGE-RAGE signalling pathway in diabetic complications, and Toll-like receptor signalling pathway. Finally, molecular docking confirmed a strong binding affinity between the 7 main active components with the hub genes. The findings suggested that beta-sitosterol, kaempferol and luteolin might be the promising leading components due to their good molecular docking scores.
Collapse
Affiliation(s)
- Yisheng Huang
- Department of Anesthesiology, Ganzhou People's Hospital, Ganzhou, P.R. China
- Department of Anesthesiology, Nanfang Hospital Affiliated to Southern Medical University, Guangzhou, P.R. China
| | - Yan Fang
- Department of Anesthesiology, Ganzhou People's Hospital, Ganzhou, P.R. China
| | - Huanhuan Jie
- Department of Anesthesiology, Ganzhou People's Hospital, Ganzhou, P.R. China
| | - Hongbiao Yang
- Department of Anesthesiology, Ganzhou People's Hospital, Ganzhou, P.R. China
| | - Wen Zhou
- Department of Anesthesiology, Ganzhou People's Hospital, Ganzhou, P.R. China
| | - Yijian Chen
- Department of Anesthesiology, Ganzhou People's Hospital, Ganzhou, P.R. China
| | - Baolin Zhong
- Department of Anesthesiology, Ganzhou People's Hospital, Ganzhou, P.R. China
| |
Collapse
|
9
|
Paranga TG, Mitu I, Pavel-Tanasa M, Rosu MF, Miftode IL, Constantinescu D, Obreja M, Plesca CE, Miftode E. Cytokine Storm in COVID-19: Exploring IL-6 Signaling and Cytokine-Microbiome Interactions as Emerging Therapeutic Approaches. Int J Mol Sci 2024; 25:11411. [PMID: 39518964 PMCID: PMC11547016 DOI: 10.3390/ijms252111411] [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: 09/24/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
IL-6 remains a key molecule of the cytokine storms characterizing COVID-19, exerting both proinflammatory and anti-inflammatory effects. Emerging research underscores the significance of IL-6 trans-signaling over classical signaling pathways, which has shifted the focus of therapeutic strategies. Additionally, the synergistic action of TNF-α and IFN-γ has been found to induce inflammatory cell death through PANoptosis, further amplifying the severity of cytokine storms. Long COVID-19 patients, as well as those with cytokine storms triggered by other conditions, exhibit distinct laboratory profiles, indicating the need for targeted approaches to diagnosis and management. Growing evidence also highlights the gut microbiota's crucial role in modulating the immune response during COVID-19 by affecting cytokine production, adding further complexity to the disease's immunological landscape. Targeted intervention strategies should focus on specific cytokine cutoffs, though accurate cytokine quantification remains a clinical challenge. Current treatment strategies are increasingly focused on inhibiting IL-6 trans-signaling, which offers promise for more precise therapeutic approaches to manage hyperinflammatory responses in COVID-19. In light of recent discoveries, this review summarizes key research findings on cytokine storms, particularly their role in COVID-19 and other inflammatory conditions. It explores emerging therapeutic strategies targeting cytokines like IL-6, TNF-α, and IFN-γ, while also addressing open questions, such as the need for better biomarkers to detect and manage cytokine storms. Additionally, the review highlights ongoing challenges in developing targeted treatments that mitigate hyperinflammation without compromising immune function, emphasizing the importance of continued research in this field.
Collapse
Affiliation(s)
- Tudorita Gabriela Paranga
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.G.P.); (I.-L.M.); (M.O.); (C.E.P.); (E.M.)
- St. Parascheva Clinical Hospital for Infectious Diseases, 700116 Iasi, Romania
| | - Ivona Mitu
- Department of Morpho-Functional Sciences II, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Mariana Pavel-Tanasa
- Department of Immunology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Laboratory of Immunology, St. Spiridon County Clinical Emergency Hospital, 700101 Iasi, Romania
| | - Manuel Florin Rosu
- St. Parascheva Clinical Hospital for Infectious Diseases, 700116 Iasi, Romania
- Department of Preventive Medicine and Interdisciplinarity, Faculty of Medicine, University of Medicine and Pharmacy Grigore. T. Popa, 700115 Iasi, Romania
| | - Ionela-Larisa Miftode
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.G.P.); (I.-L.M.); (M.O.); (C.E.P.); (E.M.)
- St. Parascheva Clinical Hospital for Infectious Diseases, 700116 Iasi, Romania
| | - Daniela Constantinescu
- Department of Immunology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Laboratory of Immunology, St. Spiridon County Clinical Emergency Hospital, 700101 Iasi, Romania
| | - Maria Obreja
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.G.P.); (I.-L.M.); (M.O.); (C.E.P.); (E.M.)
- St. Parascheva Clinical Hospital for Infectious Diseases, 700116 Iasi, Romania
| | - Claudia Elena Plesca
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.G.P.); (I.-L.M.); (M.O.); (C.E.P.); (E.M.)
- St. Parascheva Clinical Hospital for Infectious Diseases, 700116 Iasi, Romania
| | - Egidia Miftode
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.G.P.); (I.-L.M.); (M.O.); (C.E.P.); (E.M.)
- St. Parascheva Clinical Hospital for Infectious Diseases, 700116 Iasi, Romania
| |
Collapse
|
10
|
Han E, Youn S, Kwon KT, Kim SC, Jo HY, Jung I. Disease progression associated cytokines in COVID-19 patients with deteriorating and recovering health conditions. Sci Rep 2024; 14:24712. [PMID: 39433797 PMCID: PMC11494080 DOI: 10.1038/s41598-024-75924-x] [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: 04/11/2024] [Accepted: 10/09/2024] [Indexed: 10/23/2024] Open
Abstract
Understanding the immune response to COVID-19 is challenging due to its high variability among individuals. To identify differentially expressed cytokines between the deteriorating and recovering phases, we analyzed the Electronic Health Records (EHR) and cytokine profile data in a COVID-19 cohort of 444 infected patients and 145 non-infected healthy individuals. We categorized each patient's progression into Deterioration Phase (DP) and Recovery Phase (RP) using longitudinal neutrophil, lymphocyte and lactate dehydrogenase levels. A random forest model was built using healthy and severe patients to compute the contribution of each cytokine toward disease progression using Shapley Additive Explanations (SHAP). SHAP values were used for supervised clustering to identify DP and RP-related samples and their associated cytokines. The identified clusters effectively discriminated DP and RP samples, suggesting that the cytokine profiles differed between deteriorating and recovering health conditions. Especially, CXCL10, GDF15, PTX3, and TNFSF10 were differentially expressed between the DP and RP samples, which are involved in the JAK-STAT, NF- κ B, and MAPK signaling pathways contributing to the inflammatory response. Collectively, we characterized the immune response in terms of disease progression of COVID-19 with deteriorating and recovering health conditions.
Collapse
Affiliation(s)
- Eonyong Han
- School of Computer Science and Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Sohyun Youn
- School of Computer Science and Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ki Tae Kwon
- Kyungpook National University Chilgok Hospital, 807 Hoguk-ro, Buk-gu, Daegu, 41404, Republic of Korea
| | - Sang Cheol Kim
- Division of Healthcare and Artificial Intelligence, Department of Precision Medicine, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju-si, 28159, Republic of Korea
| | - Hye-Yeong Jo
- Division of Healthcare and Artificial Intelligence, Department of Precision Medicine, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Osong, Cheongju-si, 28159, Republic of Korea.
| | - Inuk Jung
- School of Computer Science and Engineering, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
11
|
Umsumarng S, Dissook S, Arjsri P, Srisawad K, Thippraphan P, Sangphukieo A, Thongkumkoon P, Dejkriengkraikul P. Inhibitory Effect of Luteolin on Spike S1 Glycoprotein-Induced Inflammation in THP-1 Cells via the ER Stress-Inducing Calcium/CHOP/MAPK Pathway. Pharmaceuticals (Basel) 2024; 17:1402. [PMID: 39459041 PMCID: PMC11509993 DOI: 10.3390/ph17101402] [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: 08/27/2024] [Revised: 09/13/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES The global SARS-CoV-2 outbreak has escalated into a critical public health emergency, with the spike glycoprotein S1 subunit of SARS-CoV-2 (spike-S1) linked to inflammation in lung tissue and immune cells. Luteolin, a flavone with anti-inflammatory properties, shows promise, but research on its effectiveness against long-COVID-related inflammation and spike protein-induced responses remains limited. This study aims to elucidate the underlying mechanisms of inflammation in THP-1 cells induced by the spike-S1. Additionally, it seeks to assess the potential of luteolin in mitigating inflammatory responses induced by the spike-S1 in a THP-1 macrophage model. METHODS The gene expression profiles of spike-S1 in THP-1 cells were analyzed by transcriptome sequencing. The inhibitory effect of luteolin on ER stress and inflammation in spike-S1-induced THP-1 cells was investigated using Western blotting, RT-PCR, and ELISA. RESULTS The candidate genes (CAMK2A, SIGLEC7, PPARGC1B, SEC22B, USP28, IER2, and TIRAP) were upregulated in the spike-S1-induced THP-1 group compared to the control group. Among these, calcium/calmodulin-dependent protein kinase II alpha (CAMK2A) was identified as the most promising molecule in spike-S1-induced THP-1 cells. Our results indicate that the spike S1 significantly increased the expression of ER-stress markers at both gene and protein levels. Luteolin significantly reduced ER stress by decreasing the expression of ER-stress marker genes and ER-stress marker proteins (p < 0.01). Additionally, luteolin exhibited anti-inflammatory properties upon spike S1-induction in THP-1 cells by significantly suppressing IL-6, IL-8, and IL-1β cytokine secretion in a dose-dependent manner (p < 0.05). Furthermore, our results revealed that luteolin exhibited the downregulation of the MAPK pathway, as evidenced by modulating the phosphorylation of p-ERK1/2, p-JNK and p-p38 proteins (p < 0.05). CONCLUSIONS The results from this study elucidate the mechanisms by which the spike S1 induces inflammation in THP-1 cells and supports the use of naturally occurring bioactive compounds, like luteolin, against inflammation-related SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Sonthaya Umsumarng
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand;
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Sivamoke Dissook
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand;
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.A.); (K.S.); (P.T.)
| | - Punnida Arjsri
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.A.); (K.S.); (P.T.)
| | - Kamonwan Srisawad
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.A.); (K.S.); (P.T.)
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pilaiporn Thippraphan
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.A.); (K.S.); (P.T.)
| | - Apiwat Sangphukieo
- Center of Multidisciplinary Technology for Advanced Medicine (CMU-TEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (A.S.); (P.T.)
| | - Patcharawadee Thongkumkoon
- Center of Multidisciplinary Technology for Advanced Medicine (CMU-TEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (A.S.); (P.T.)
| | - Pornngarm Dejkriengkraikul
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand;
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (P.A.); (K.S.); (P.T.)
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
12
|
Deus MDC, Gadotti AC, Dias ES, Monte Alegre JB, Van Spitzenbergen BAK, Andrade GB, Tozoni SS, Stocco RB, Olandoski M, Tuon FFB, Pinho RA, de Noronha L, Baena CP, Moreno-Amaral AN. Prospective Variation of Cytokine Trends during COVID-19: A Progressive Approach from Disease Onset until Outcome. Int J Mol Sci 2024; 25:10578. [PMID: 39408907 PMCID: PMC11477561 DOI: 10.3390/ijms251910578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/19/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
COVID-19 is characterized by pronounced hypercytokinemia. The cytokine switch, marked by an imbalance between pro-inflammatory and anti-inflammatory cytokines, emerged as a focal point of investigation throughout the COVID-19 pandemic. However, the kinetics and temporal dynamics of cytokine release remain contradictory, making the development of new therapeutics difficult, especially in severe cases. This study collected serum samples from SARS-CoV-2 infected patients at 72 h intervals and monitored them for various cytokines at each timepoint until hospital discharge or death. Cytokine levels were analyzed based on time since symptom onset and patient outcomes. All cytokines studied prospectively were strong predictors of mortality, particularly IL-4 (AUC = 0.98) and IL-1β (AUC = 0.96). First-timepoint evaluations showed elevated cytokine levels in the mortality group (p < 0.001). Interestingly, IFN-γ levels decreased over time in the death group but increased in the survival group. Patients who died exhibited sustained levels of IL-1β and IL-4 and increased IL-6 levels over time. These findings suggest cytokine elevation is crucial in predicting COVID-19 mortality. The dynamic interplay between IFN-γ and IL-4 highlights the balance between Th1/Th2 immune responses and underscores IFN-γ as a powerful indicator of immune dysregulation throughout the infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Andrea Novais Moreno-Amaral
- Programa de Pós-Graduação em Ciências da Saúde (PPGCS), Escola de Medicina, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, PR, Brazil; (M.d.C.D.); (A.C.G.); (E.S.D.); (J.B.M.A.); (B.A.K.V.S.); (G.B.A.); (S.S.T.); (R.B.S.); (M.O.); (F.F.B.T.); (R.A.P.); (L.d.N.); (C.P.B.)
| |
Collapse
|
13
|
AbdelAziz RA, Abd-Allah ST, Moness HM, Anwar AM, Mohamed ZH. Role of interleukin 6 polymorphism and inflammatory markers in outcome of pediatric Covid- 19 patients. BMC Pediatr 2024; 24:625. [PMID: 39354444 PMCID: PMC11443869 DOI: 10.1186/s12887-024-05071-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 09/10/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND IL-6 polymorphisms were associated to viral infection outcomes through affection of IL-6 production and it is an early indicator of tissue injury and systemic inflammatory response. The study aimed to determine whether genetic IL-6 polymorphisms, serum interleukin-6 level and inflammatory markers (Presepsin, CXCL-10, C3, and C4) are associated with the prediction of disease severity in pediatric COVID-19 patients and its possible use as a prognostic tool in pediatric patients admitted to hospital. METHODS This prospective cohort study was conducted on 150 children with COVID-19. Patients were divided according to the severity of infection into four groups: group I (mild) 67 cases; group II (moderate) 53 cases, group III (severe) 17 cases and group IV (critical) 14 cases. Serum Interleukin 6, CXCL-10, Presepsin, renal and liver functions, electrolytes, C3, C4, ferritin, and D dimer serum levels were assessed in all patients. The Kruskal Wallis test used to compare parametric quantitative data between studied groups and Mann Whitney test for each pair of groups. Non-parametric quantitative data was compared between studied groups using a one-way ANOVA test and post-hoc Bonferroni analysis for each pair of groups. RESULTS Group I: 35 males and 32 females with a median age of 16 months. Group II: 17 males and 35 females with a median age of 13 months. Group III: 6 males and 11 females with a median age of 12 months and group IV: 3 males and 11 females with a median age of 12 months. There was no statistical difference between the studied groups regarding gender and age. Serum levels of IL- 6, serum ferritin; D-dimer, Presepsin and CXCL 10 were significantly higher in both severe and critical groups than the other 2 groups (mild and moderate). ROC curve analysis showed that interleukin-6 and Presepsin were good markers for prediction of severity of COVID-19 among the diseased children. For severe cases, the sensitivity of interleukin-6 was 76.47% and specificity was 92.31%. For critical cases, the sensitivity of interleukin-6 was 71.43% and specificity was 82.35%. The sensitivity of Presepsin was 76.47% and specificity was 88.46% in severe cases. For critical cases, the sensitivity of Presepsin was 78.57% and specificity of 91.2%. There was significant difference in IL-6 572 allelic among moderate cases with the most frequent 42.3% for genotype (GC) and allelic among severe cases with the most frequent 47.1% for genotype (GC). Significant difference in IL-6 174 allelic among critical cases with the most frequent 78.6% for genotype (CC). CONCLUSIONS Children whom expressed GC genotypes of IL6 (-572G > C) polymorphism are at a considerably higher risk of developing a severe disease. This risk is significantly larger in the severe group of children than in children in critical condition who have GC genotypes of IL6 (-174 G > C) polymorphism. While IL6 (-597G > A) polymorphism has no role in COVID 19 severity in children.
Collapse
Affiliation(s)
- Reem A AbdelAziz
- Pediatric Department, Faculty of Medicine, Minia University, Minia, Egypt
| | | | - Hend M Moness
- Clinical Pathology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | - Ahmed M Anwar
- Pediatric Department, Faculty of Medicine, Minia University, Minia, Egypt
| | | |
Collapse
|
14
|
Iqbal K, Banga A, Arif TB, Rathore SS, Bhurwal A, Naqvi SKB, Mehdi M, Kumar P, Salklan MM, Iqbal A, Ahmed J, Sharma N, Lal A, Kashyap R, Bansal V, Domecq JP. Anticoagulant use before COVID-19 diagnosis prevent COVID-19 associated acute venous thromboembolism or not: A systematic review and meta-analysis. World J Methodol 2024; 14:92983. [PMID: 39310244 PMCID: PMC11230074 DOI: 10.5662/wjm.v14.i3.92983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/24/2024] [Accepted: 05/11/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Coagulopathy and thromboembolic events are associated with poor outcomes in coronavirus disease 2019 (COVID-19) patients. There is conflicting evidence on the effects of chronic anticoagulation on mortality and severity of COVID-19 disease. AIM To summarize the body of evidence on the effects of pre-hospital anticoagulation on outcomes in COVID-19 patients. METHODS A Literature search was performed on LitCovid PubMed, WHO, and Scopus databases from inception (December 2019) till June 2023 for original studies reporting an association between prior use of anticoagulants and patient outcomes in adults with COVID-19. The primary outcome was the risk of thromboembolic events in COVID-19 patients taking anticoagulants. Secondary outcomes included COVID-19 disease severity, in terms of intensive care unit admission or invasive mechanical ventilation/intubation requirement in patients hospitalized with COVID-19 infection, and mortality. The random effects models were used to calculate crude and adjusted odds ratios (aORs) with 95% confidence intervals (95%CIs). RESULTS Forty-six observational studies met our inclusion criteria. The unadjusted analysis found no association between prior anticoagulation and thromboembolic event risk [n = 43851, 9 studies, odds ratio (OR)= 0.67 (0.22, 2.07); P = 0.49; I 2 = 95%]. The association between prior anticoagulation and disease severity was non-significant [n = 186782; 22 studies, OR = 1.08 (0.78, 1.49); P = 0.64; I 2 = 89%]. However, pre-hospital anticoagulation significantly increased all-cause mortality risk [n = 207292; 35 studies, OR = 1.72 (1.37, 2.17); P < 0.00001; I 2 = 93%]. Pooling adjusted estimates revealed a statistically non-significant association between pre-hospital anticoagulation and thromboembolic event risk [aOR = 0.87 (0.42, 1.80); P = 0.71], mortality [aOR = 0.94 (0.84, 1.05); P = 0.31], and disease severity [aOR = 0.96 (0.72, 1.26); P = 0.76]. CONCLUSION Prehospital anticoagulation was not significantly associated with reduced risk of thromboembolic events, improved survival, and lower disease severity in COVID-19 patients.
Collapse
Affiliation(s)
- Kinza Iqbal
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Akshat Banga
- Department of Internal Medicine, Sawai Man Singh Medical College, Jaipur 302004, India
| | - Taha Bin Arif
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Sawai Singh Rathore
- Department of Internal Medicine, Dr. Sampurnanand Medical College, Jodhpur 342003, Rajasthan, India
| | - Abhishek Bhurwal
- Department of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson School of Medicine, New Brunswick, NJ 08901, United States
| | | | - Muhammad Mehdi
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Pankaj Kumar
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Mitali Madhu Salklan
- Department of Internal Medicine, Pandit Bhagwat Dayal Sharma Post Graduate Institute of Medical Sciences, Rohtak 124001, Haryana, India
| | - Ayman Iqbal
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Jawad Ahmed
- Department of Internal Medicine, Dow Medical College, Karachi 74200, Pakistan
| | - Nikhil Sharma
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, United States
| | - Amos Lal
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905, United States
| | - Rahul Kashyap
- Department of Research, Wellspan Health, York, PA 17403, United States
| | - Vikas Bansal
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, United States
| | - Juan Pablo Domecq
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, United States
| |
Collapse
|
15
|
Gunasekara S, Tamil Selvan M, Murphy CL, Shatnawi S, Cowan S, More S, Ritchey J, Miller CA, Rudd JM. Characterization of Neutrophil Functional Responses to SARS-CoV-2 Infection in a Translational Feline Model for COVID-19. Int J Mol Sci 2024; 25:10054. [PMID: 39337543 PMCID: PMC11432149 DOI: 10.3390/ijms251810054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
There is a complex interplay between viral infection and host innate immune response regarding disease severity and outcomes. Neutrophil hyperactivation, including excessive release of neutrophil extracellular traps (NETs), is linked to exacerbated disease in acute COVID-19, notably in hospitalized patients. Delineating protective versus detrimental neutrophil responses is essential to developing targeted COVID-19 therapies and relies on high-quality translational animal models. In this study, we utilize a previously established feline model for COVID-19 to investigate neutrophil dysfunction in which experimentally infected cats develop clinical disease that mimics acute COVID-19. Specific pathogen-free cats were inoculated with SARS-CoV-2 (B.1.617.2; Delta variant) (n = 24) or vehicle (n = 6). Plasma, bronchoalveolar lavage fluid, and lung tissues were collected at various time points over 12 days post-inoculation. Systematic and temporal evaluation of the kinetics of neutrophil activation was conducted by measuring markers of activation including myeloperoxidase (MPO), neutrophil elastase (NE), and citrullinated histone H3 (citH3) in SARS-CoV-2-infected cats at 4 and 12 days post-inoculation (dpi) and compared to vehicle-inoculated controls. Cytokine profiling supported elevated innate inflammatory responses with specific upregulation of neutrophil activation and NET formation-related markers, namely IL-8, IL-18, CXCL1, and SDF-1, in infected cats. An increase in MPO-DNA complexes and cell-free dsDNA in infected cats compared to vehicle-inoculated was noted and supported by histopathologic severity in respiratory tissues. Immunofluorescence analyses further supported correlation of NET markers with tissue damage, especially 4 dpi. Differential gene expression analyses indicated an upregulation of genes associated with innate immune and neutrophil activation pathways. Transcripts involved in activation and NETosis pathways were upregulated by 4 dpi and downregulated by 12 dpi, suggesting peak activation of neutrophils and NET-associated markers in the early acute stages of infection. Correlation analyses conducted between NET-specific markers and clinical scores as well as histopathologic scores support association between neutrophil activation and disease severity during SARS-CoV-2 infection in this model. Overall, this study emphasizes the effect of neutrophil activation and NET release in SARS-CoV-2 infection in a feline model, prompting further investigation into therapeutic strategies aimed at mitigating excessive innate inflammatory responses in COVID-19.
Collapse
Affiliation(s)
- Sachithra Gunasekara
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Miruthula Tamil Selvan
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Chelsea L Murphy
- Department of Mathematical Sciences, College of Arts and Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| | - Shoroq Shatnawi
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Shannon Cowan
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Sunil More
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Jerry Ritchey
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Craig A Miller
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Jennifer M Rudd
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
16
|
González-Acedo A, Manzano-Moreno FJ, García-Recio E, Ruiz C, de Luna-Bertos E, Costela-Ruiz VJ. Assessment of Supplementation with Different Biomolecules in the Prevention and Treatment of COVID-19. Nutrients 2024; 16:3070. [PMID: 39339670 PMCID: PMC11434975 DOI: 10.3390/nu16183070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/30/2024] Open
Abstract
Consequences of the disease produced by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have led to an urgent search for preventive and therapeutic strategies. Besides drug treatments, proposals have been made for supplementation with biomolecules possessing immunomodulatory and antioxidant properties. The objective of this study was to review published evidence on the clinical usefulness of supplementation with vitamin D, antioxidant vitamins (vitamin A, vitamin E, and vitamin C), melatonin, lactoferrin and natural products found in food (curcumin, luteolin, ginger, allicin, magnesium and zinc) as supplements in SARS-CoV-2 infection. In general, supplementation of conventional treatments with these biomolecules has been found to improve the clinical symptoms and severity of the coronavirus disease (COVID-19), with some indications of a preventive effect. In conclusion, these compounds may assist in preventing and/or improving the symptoms of COVID-19. Nevertheless, only limited evidence is available, and findings have been inconsistent. Further investigations are needed to verify the therapeutic potential of these supplements.
Collapse
Affiliation(s)
- Anabel González-Acedo
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain; (A.G.-A.); (E.G.-R.); (C.R.); (V.J.C.-R.)
| | - Francisco Javier Manzano-Moreno
- Biomedical Group (BIO277), Department of Stomatology, School of Dentistry, University of Granada, 18016 Granada, Spain;
- Institute of Biosanitary Research, ibs.Granada, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2ª Planta, 18012 Granada, Spain
| | - Enrique García-Recio
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain; (A.G.-A.); (E.G.-R.); (C.R.); (V.J.C.-R.)
- Institute of Biosanitary Research, ibs.Granada, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2ª Planta, 18012 Granada, Spain
| | - Concepción Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain; (A.G.-A.); (E.G.-R.); (C.R.); (V.J.C.-R.)
- Institute of Biosanitary Research, ibs.Granada, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2ª Planta, 18012 Granada, Spain
- Institute of Neuroscience, University of Granada, 18016 Granada, Spain
| | - Elvira de Luna-Bertos
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain; (A.G.-A.); (E.G.-R.); (C.R.); (V.J.C.-R.)
- Institute of Biosanitary Research, ibs.Granada, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2ª Planta, 18012 Granada, Spain
| | - Víctor Javier Costela-Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Avda. Ilustración 60, 18016 Granada, Spain; (A.G.-A.); (E.G.-R.); (C.R.); (V.J.C.-R.)
- Institute of Biosanitary Research, ibs.Granada, Avda. de Madrid, 15 Pabellón de Consultas Externas, 2ª Planta, 18012 Granada, Spain
| |
Collapse
|
17
|
Mińko A, Turoń-Skrzypińska A, Rył A, Mańkowska K, Cymbaluk-Płoska A, Rotter I. The Importance of the Concentration of Selected Cytokines (IL-6, IL-10, IL-12, IL-15, TNF-α) and Inflammatory Markers (CRP, NLR, PLR, LMR, SII) in Predicting the Course of Rehabilitation for Patients after COVID-19 Infection. Biomedicines 2024; 12:2055. [PMID: 39335569 PMCID: PMC11429050 DOI: 10.3390/biomedicines12092055] [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: 08/08/2024] [Revised: 08/31/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND/OBJECTIVES In the course of COVID-19, there may be an excessive immune response of the body with the release of large amounts of pro-inflammatory cytokines, causing a "cytokine storm", also known as cytokine release syndrome (CRS). The COVID-19 pandemic has shown how important an integrated approach to health care is, with physiotherapy being one of its fundamental aspects. The aim of this study was to analyze the potential relationship between the level of selected cytokines (IL-6, IL-10, IL-12, IL-15, TNF-α) and inflammatory markers (CRP, NLR, PLR, LMR, SII) and the duration of rehabilitation in patients after COVID-19. METHODS The examined patients participated in a comprehensive rehabilitation program, which included breathing exercises, aerobic training, and strength and endurance training. Peripheral venous blood samples were also collected from each patient. RESULTS Factors such as gender, smoking status, IL-10, and the presence of pneumonia during infection were significantly associated with the length of rehabilitation after COVID-19. CONCLUSIONS The course of rehabilitation after COVID-19 may depend on many factors, including smoking, the presence of pneumonia due to infection, and some parameters of inflammation.
Collapse
Affiliation(s)
- Alicja Mińko
- Department of Medical Rehabilitation and Clinical Physiotherapy, Pomeranian Medical University, 71-210 Szczecin, Poland; (A.T.-S.); (A.R.); (I.R.)
| | - Agnieszka Turoń-Skrzypińska
- Department of Medical Rehabilitation and Clinical Physiotherapy, Pomeranian Medical University, 71-210 Szczecin, Poland; (A.T.-S.); (A.R.); (I.R.)
| | - Aleksandra Rył
- Department of Medical Rehabilitation and Clinical Physiotherapy, Pomeranian Medical University, 71-210 Szczecin, Poland; (A.T.-S.); (A.R.); (I.R.)
| | - Katarzyna Mańkowska
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Aneta Cymbaluk-Płoska
- Department of Reconstructive Surgery and Gynecological Oncology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Iwona Rotter
- Department of Medical Rehabilitation and Clinical Physiotherapy, Pomeranian Medical University, 71-210 Szczecin, Poland; (A.T.-S.); (A.R.); (I.R.)
| |
Collapse
|
18
|
Frent S, Popovici AF, Balan A, Cerbu B, Marincu I, Mihaicuta S, Bikov A. Religiosity and Health Outcomes in a Cohort of Romanian Patients Hospitalized for COVID-19. JOURNAL OF RELIGION AND HEALTH 2024:10.1007/s10943-024-02120-6. [PMID: 39254783 DOI: 10.1007/s10943-024-02120-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 09/11/2024]
Abstract
There is a growing body of evidence for the interrelation between health status and religious beliefs. Our aim was to evaluate the level of religiosity in patients hospitalized for COVID-19 and to assess the link between religiosity and measurable health outcomes. This was an observational, single-center study which included patients with moderate-to-severe forms of COVID-19. A total of 112 patients were enrolled in the study, of whom 77 were highly religious (CRS-15 score ≥ 4) and 35 non-highly religious (CRS-15 score < 4). There was no difference in demographics or prevalence of comorbidities between the two groups. Furthermore, we found no difference between groups in radiological extension of lung lesions, length of hospital stays, or ICU need; however, in-hospital mortality rate was significantly lower in highly religious group (1% vs. 14%, p = 0.005). Serum ferritin level at admission was significantly lower (p = 0.03) and prevalence of post-COVID-19 pulmonary sequelae significantly higher in highly religious group (p = 0.02).
Collapse
Affiliation(s)
- Stefan Frent
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases, Department of Pulmonology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041, Timisoara, Romania
| | - Alexandru-Filip Popovici
- Faculty of Psychology and Educational Sciences, University of Bucharest, Panduri Street No. 90, 050657, Bucharest, Romania.
| | - Adrian Balan
- Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041, Timisoara, Romania
| | - Bianca Cerbu
- Infectious Diseases Department, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041, Timisoara, Romania
| | - Iosif Marincu
- Infectious Diseases Department, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041, Timisoara, Romania
| | - Stefan Mihaicuta
- Center for Research and Innovation in Precision Medicine of Respiratory Diseases, Department of Pulmonology, "Victor Babes" University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041, Timisoara, Romania
| | - Andras Bikov
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Oxford Road, Manchester, M13 9WL, UK
- Division of Infection, Immunity & Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|
19
|
Valle N, Eapen MS, Pillai K, Morris R, Akhter J, Mekkawy AH, Morris DL, Valle SJ. Impact of Nebulized BromAc ® on Mucus Plug Clearance in a Mechanically Ventilated Ex Vivo Ovine Lung Model of Obstructive Respiratory Conditions. Life (Basel) 2024; 14:1111. [PMID: 39337895 PMCID: PMC11433166 DOI: 10.3390/life14091111] [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: 06/17/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Mucus plugging of the respiratory tract occurs in airway diseases, including asthma, chronic obstructive pulmonary disease, and cystic fibrosis. It can cause blockage of the airways, leading to breathlessness and lung failure. Here, we used a ventilatory setup to demonstrate the effect of BromAc® in dissolving mucus plugs in a novel ex vivo ovine obstructive lung model. Mucus simulant was filled into the trachea of freshly slaughtered ovine lungs and ventilated via an endotracheal tube (ETT) using Continuous Mandatory Ventilation. Predetermined single or repeated doses of Bromelain, Acetylcysteine (Ac), BromAc®, and saline control were administered via an Aerogen® vibrating nebulizer and ventilated for 30 or 60 min. Ventilatory recording of resistance, compliance, and tidal volume was conducted, and rheology pre- and post-treatment were measured. A significant decline in airway resistance (p < 0.0001) compared to the saline control was observed when treated with Bromelain, Ac, and BromAc®, with the latter showing a stronger mucolytic effect than single agents. The decline in resistance was also effective in shorter time points (p < 0.05) at lower doses of the drugs. Changes in compliance, peak pressure, and tidal volume were not observed after administration of the drugs. Rheology measurements revealed that BromAc®TM significantly reduced the viscosity of the mucin at the end of 30 min and 60 min time points (p < 0.001) compared to the saline control. BromAc® showed complete dissolution of the respiratory mucus simulant and improved ventilatory airflow parameters in the ex vivo ovine model.
Collapse
Affiliation(s)
- Nicole Valle
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
| | - Mathew Suji Eapen
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
| | - Krishna Pillai
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
| | - Richard Morris
- Intensive Care Unit, Shoalhaven District Memorial Hospital, Nowra, NSW 2541, Australia;
| | - Javed Akhter
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
| | - Ahmed H. Mekkawy
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
| | - David L. Morris
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
- Department of Surgery, St George Hospital, Sydney, NSW 2217, Australia
- St George and Sutherland Clinical School of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Sarah J. Valle
- Mucpharm Pty Ltd., Sydney, NSW 2217, Australia; (N.V.); (K.P.); (J.A.); (A.H.M.); (D.L.M.)
- St George and Sutherland Clinical School of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
- Intensive Care Unit, St George Hospital, Sydney, NSW 2217, Australia
| |
Collapse
|
20
|
Xu Y, Biby S, Guo C, Liu Z, Cai J, Wang XY, Zhang S. Characterization of a small molecule inhibitor of the NLRP3 inflammasome and its potential use for acute lung injury. Bioorg Chem 2024; 150:107562. [PMID: 38901282 PMCID: PMC11270536 DOI: 10.1016/j.bioorg.2024.107562] [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: 04/24/2024] [Revised: 05/23/2024] [Accepted: 06/13/2024] [Indexed: 06/22/2024]
Abstract
Accumulating data support the key roles of the NLRP3 inflammasome, an essential component of the innate immune system, in human pathophysiology. As an emerging drug target and a potential biomarker for human diseases, small molecule inhibitors of the NLRP3 inflammasome have been actively pursued. Our recent studies identified a small molecule, MS-II-124, as a potent NLRP3 inhibitor and potential imaging probe. In this report, MS-II-124 was further characterized by an unbiased and comprehensive analysis through Eurofins BioMAP Diversity PLUS panel that contains 12 human primary cell-based systems. The analysis revealed promising activities of MS-II-124 on inflammation and immune functions, further supporting the roles of the NLRP3 inflammasome in these model systems. Further studies of MS-II-124 in mouse model of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) and NLRP3 knockout mice demonstrated its target engagement, efficacy to suppress inflammatory cytokines and infiltration of immune cells in the lung tissues. In summary, the results support the therapeutic potential of MS-II-124 as a NLRP3 inhibitor and warrant future studies of this compound and its analogs to develop therapeutics for ALI/ARDS.
Collapse
Affiliation(s)
- Yiming Xu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Savannah Biby
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Chunqing Guo
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Zheng Liu
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jinyang Cai
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
21
|
Moeed A, Larik MO, Fahim MAA, Rahman HAU, Najmi L, Changez MIK, Javed MM, Hasibuzzaman MA. Impact of non-alcoholic fatty liver disease on coronavirus disease 2019: A systematic review. World J Hepatol 2024; 16:1185-1198. [PMID: 39221098 PMCID: PMC11362910 DOI: 10.4254/wjh.v16.i8.1185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/17/2024] [Accepted: 07/26/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Many studies have revealed a link between non-alcoholic fatty liver disease (NAFLD) and coronavirus disease 2019 (COVID-19), making understanding the relationship between these two conditions an absolute requirement. AIM To provide a qualitative synthesis on the currently present data evaluating COVID-19 and NAFLD. METHODS This systematic review was conducted in accordance with the guidelines provided by preferred reporting items for systematic reviews and meta-analyses and the questionnaire utilized the population, intervention, comparison, and outcome framework. The search strategy was run on three separate databases, PubMed/MEDLINE, Scopus, and Cochrane Central, which were systematically searched from inception until March 2024 to select all relevant studies. In addition, ClinicalTrials.gov, Medrxiv.org, and Google Scholar were searched to identify grey literature. RESULTS After retrieval of 11 studies, a total of 39282 patients data were pooled. Mortality was found in 11.5% and 9.4% of people in NAFLD and non-NAFLD groups. In all, 23.2% of NAFLD patients and 22% of non-NAFLD admissions diagnosed with COVID-19 were admitted to the intensive care unit, with days of stay varying. Ventilatory support ranged from 5% to 40.5% in the NAFLD cohort and from 3.1% to 20% in the non-NAFLD cohort. The incidence of acute liver injury showed significance. Clinical improvement on days 7 and 14 between the two classifications was significant. Hospitalization stay ranged from 9.6 days to 18.8 days and 7.3 days to 16.4 days in the aforementioned cohorts respectively, with 73.3% and 76.3% of patients being discharged. Readmission rates varied. CONCLUSION Clinical outcomes except mortality consistently showed a worsening trend in patients with NAFLD and concomitant COVID-19. Further research in conducting prospective longitudinal studies is essential for a more powerful conclusion.
Collapse
Affiliation(s)
- Abdul Moeed
- Department of Internal Medicine, Dow University of Health Sciences, Karachi 74200, Sindh, Pakistan
| | - Muhammad Omar Larik
- Department of Internal Medicine, Dow International Medical College, Karachi 74200, Sindh, Pakistan
| | - Muhammad Ahmed Ali Fahim
- Department of Internal Medicine, Dow University of Health Sciences, Karachi 74200, Sindh, Pakistan
| | - Hafsah Alim Ur Rahman
- Department of Internal Medicine, Dow University of Health Sciences, Karachi 74200, Sindh, Pakistan
| | - Lubna Najmi
- Department of Internal Medicine, Ziauddin University, Karachi 75000, Sindh, Pakistan
| | - Mah I Kan Changez
- Department of Research, Yale University, New Haven 06520, CT, United States
| | - Muhammad Moiz Javed
- Department of Internal Medicine, King Edward Medical University, Lahore 54000, Punjab, Pakistan
| | - Md Al Hasibuzzaman
- Institute of Nutrition and Food Science, University of Dhaka, Dhaka 1000, Bangladesh
- Department of Medicine, The Affiliated Hospital of Medical School of Ningbo University, Ningbo 315211, Zhejiang Province, China.
| |
Collapse
|
22
|
Kurata H, Harun-Or-Roshid M, Tsukiyama S, Maeda K. PredIL13: Stacking a variety of machine and deep learning methods with ESM-2 language model for identifying IL13-inducing peptides. PLoS One 2024; 19:e0309078. [PMID: 39172871 PMCID: PMC11340954 DOI: 10.1371/journal.pone.0309078] [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: 05/23/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024] Open
Abstract
Interleukin (IL)-13 has emerged as one of the recently identified cytokine. Since IL-13 causes the severity of COVID-19 and alters crucial biological processes, it is urgent to explore novel molecules or peptides capable of including IL-13. Computational prediction has received attention as a complementary method to in-vivo and in-vitro experimental identification of IL-13 inducing peptides, because experimental identification is time-consuming, laborious, and expensive. A few computational tools have been presented, including the IL13Pred and iIL13Pred. To increase prediction capability, we have developed PredIL13, a cutting-edge ensemble learning method with the latest ESM-2 protein language model. This method stacked the probability scores outputted by 168 single-feature machine/deep learning models, and then trained a logistic regression-based meta-classifier with the stacked probability score vectors. The key technology was to implement ESM-2 and to select the optimal single-feature models according to their absolute weight coefficient for logistic regression (AWCLR), an indicator of the importance of each single-feature model. Especially, the sequential deletion of single-feature models based on the iterative AWCLR ranking (SDIWC) method constructed the meta-classifier consisting of the top 16 single-feature models, named PredIL13, while considering the model's accuracy. The PredIL13 greatly outperformed the-state-of-the-art predictors, thus is an invaluable tool for accelerating the detection of IL13-inducing peptide within the human genome.
Collapse
Affiliation(s)
- Hiroyuki Kurata
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Kawazu, Iizuka, Fukuoka, Japan
| | - Md. Harun-Or-Roshid
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Kawazu, Iizuka, Fukuoka, Japan
| | - Sho Tsukiyama
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Kawazu, Iizuka, Fukuoka, Japan
| | - Kazuhiro Maeda
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Kawazu, Iizuka, Fukuoka, Japan
| |
Collapse
|
23
|
Jang S, Hong W, Moon Y. Obesity-compromised immunity in post-COVID-19 condition: a critical control point of chronicity. Front Immunol 2024; 15:1433531. [PMID: 39188722 PMCID: PMC11345197 DOI: 10.3389/fimmu.2024.1433531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Post-COVID-19 condition is recognized as a multifactorial disorder, with persistent presence of viral antigens, discordant immunity, delayed viral clearance, and chronic inflammation. Obesity has emerged as an independent risk factor for both SARS-CoV-2 infection and its subsequent sequelae. In this study, we aimed to predict the molecular mechanisms linking obesity and post-COVID-19 distress. Viral antigen-exposed adipose tissues display remarkable levels of viral receptors, facilitating viral entry, deposition, and chronic release of inflammatory mediators and cells in patients. Subsequently, obesity-associated inflammatory insults are predicted to disturb cellular and humoral immunity by triggering abnormal cell differentiation and lymphocyte exhaustion. In particular, the decline in SARS-CoV-2 antibody titers and T-cell exhaustion due to chronic inflammation may account for delayed virus clearance and persistent activation of inflammatory responses. Taken together, obesity-associated defective immunity is a critical control point of intervention against post-COVID-19 progression, particularly in subjects with chronic metabolic distress.
Collapse
Affiliation(s)
- Soonwoo Jang
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Republic of Korea
- Department of Medicine, Pusan National University, Yangsan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Yangsan, Republic of Korea
| | - Wooyoung Hong
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, United States
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Republic of Korea
- Department of Medicine, Pusan National University, Yangsan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Yangsan, Republic of Korea
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan, Republic of Korea
| |
Collapse
|
24
|
Al-Saigh NN, Harb AA, Abdalla S. Receptors Involved in COVID-19-Related Anosmia: An Update on the Pathophysiology and the Mechanistic Aspects. Int J Mol Sci 2024; 25:8527. [PMID: 39126095 PMCID: PMC11313362 DOI: 10.3390/ijms25158527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Olfactory perception is an important physiological function for human well-being and health. Loss of olfaction, or anosmia, caused by viral infections such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has received considerable attention, especially in persistent cases that take a long time to recover. This review discusses the integration of different components of the olfactory epithelium to serve as a structural and functional unit and explores how they are affected during viral infections, leading to the development of olfactory dysfunction. The review mainly focused on the role of receptors mediating the disruption of olfactory signal transduction pathways such as angiotensin converting enzyme 2 (ACE2), transmembrane protease serine type 2 (TMPRSS2), neuropilin 1 (NRP1), basigin (CD147), olfactory, transient receptor potential vanilloid 1 (TRPV1), purinergic, and interferon gamma receptors. Furthermore, the compromised function of the epithelial sodium channel (ENaC) induced by SARS-CoV-2 infection and its contribution to olfactory dysfunction are also discussed. Collectively, this review provides fundamental information about the many types of receptors that may modulate olfaction and participate in olfactory dysfunction. It will help to understand the underlying pathophysiology of virus-induced anosmia, which may help in finding and designing effective therapies targeting molecules involved in viral invasion and olfaction. To the best of our knowledge, this is the only review that covered all the receptors potentially involved in, or mediating, the disruption of olfactory signal transduction pathways during COVID-19 infection. This wide and complex spectrum of receptors that mediates the pathophysiology of olfactory dysfunction reflects the many ways in which anosmia can be therapeutically managed.
Collapse
Affiliation(s)
- Noor N. Al-Saigh
- Department of Basic Medical Sciences, Faculty of Medicine, Ibn Sina University for Medical Sciences, Amman 16197, Jordan;
| | - Amani A. Harb
- Department of Basic Sciences, Faculty of Arts and Sciences, Al-Ahliyya Amman University, Amman 19111, Jordan;
| | - Shtaywy Abdalla
- Department of Biological Sciences, School of Science, The University of Jordan, Amman 11942, Jordan
| |
Collapse
|
25
|
Gazeau S, Deng X, Brunet-Ratnasingham E, Kaufmann DE, Larochelle C, Morel PA, Heffernan JM, Davis CL, Smith AM, Jenner AL, Craig M. Using virtual patient cohorts to uncover immune response differences in cancer and immunosuppressed COVID-19 patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.01.605860. [PMID: 39131351 PMCID: PMC11312602 DOI: 10.1101/2024.08.01.605860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The COVID-19 pandemic caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) resulted in millions of deaths globally. Adults with immunosuppression (e.g., solid organ transplant recipients) and those undergoing active cancer treatments experience worse infections and more severe COVID-19. It is difficult to conduct clinical studies in these populations, resulting in a restricted amount of data that can be used to relate mechanisms of immune dysfunction to COVID-19 outcomes in these vulnerable groups. To study immune dynamics after infection with SARS-CoV-2 and to investigate drivers of COVID-19 severity in individuals with cancer and immunosuppression, we adapted our mathematical model of the immune response during COVID-19 and generated virtual patient cohorts of cancer and immunosuppressed patients. The cohorts of plausible patients recapitulated available longitudinal clinical data collected from patients in Montréal, Canada area hospitals. Our model predicted that both cancer and immunosuppressed virtual patients with severe COVID-19 had decreased CD8+ T cells, elevated interleukin-6 concentrations, and delayed type I interferon peaks compared to those with mild COVID-19 outcomes. Additionally, our results suggest that cancer patients experience higher viral loads (however, with no direct relation with severity), likely because of decreased initial neutrophil counts (i.e., neutropenia), a frequent toxic side effect of anti-cancer therapy. Furthermore, severe cancer and immunosuppressed virtual patients suffered a high degree of tissue damage associated with elevated neutrophils. Lastly, parameter values associated with monocyte recruitment by infected cells were found to be elevated in severe cancer and immunosuppressed patients with respect to the COVID-19 reference group. Together, our study highlights that dysfunction in type I interferon and CD8+ T cells are key drivers of immune dysregulation in COVID-19, particularly in cancer patients and immunosuppressed individuals.
Collapse
Affiliation(s)
- Sonia Gazeau
- Sainte-Justine University Hospital Research Centre, Montréal, Québec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Québec, Canada
| | - Xiaoyan Deng
- Sainte-Justine University Hospital Research Centre, Montréal, Québec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Québec, Canada
| | | | - Daniel E. Kaufmann
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Division of Infectious Diseases, Department of Medicine, Lausanne University Hospital (CHUV) and Université de Lausanne, Lausanne, Switzerland
| | - Catherine Larochelle
- Research Centre of the Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Québec, Canada
| | - Penelope A. Morel
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jane M. Heffernan
- Centre for Disease Modelling, Department of Mathematics & Statistics, York University, Toronto, Ontario, Canada
| | - Courtney L. Davis
- Natural Science Division, Pepperdine University, Malibu, California, USA
| | - Amber M. Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Adrianne L. Jenner
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Morgan Craig
- Sainte-Justine University Hospital Research Centre, Montréal, Québec, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
26
|
Schaler L, Ghanim M, Guardiola J, Kaulsay J, Ibrahim A, Brady G, McCormack W, Conlon N, Kelly VP, Wingfield M, Glover L. Impact of COVID-19 vaccination on seminal and systemic inflammation in men. J Reprod Immunol 2024; 164:104287. [PMID: 38964132 DOI: 10.1016/j.jri.2024.104287] [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: 06/07/2023] [Revised: 05/08/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024]
Abstract
Expedited development of SARS-CoV-2 vaccines led to public concerns regarding impacts of the novel vaccine on gametes in patients seeking assisted reproduction. In cases of an acute intermittent illness or fever in men, it is often advised to postpone ART treatments so that efforts can be made to enhance wellbeing and improve sperm parameters. However, it is unknown whether sperm parameters are altered in the acute (24-72 hour) phase following COVID-19 vaccination. We performed a longitudinal cohort study of 17 normospermic male patients attending a fertility clinic for semen analysis. Semen and matched peripheral blood samples were collected prior to vaccination, within 46 + 18.9 hours of vaccine course completion (acute) and at 88.4 + 12 days (3 months) post-vaccination. No overall change from baseline was seen in symptoms, mean volume, pH, sperm concentration, motility, morphology or DNA damage in the acute or long phase. Seminal plasma was found to be negative for anti-SARS-CoV2 Spike antibody detection, and MCP-1 levels showed an acute but transient elevation post-vaccine, while IL-8 was marginally increased 3 months after completion of vaccination. A modest, positive correlation was noted between serum levels of the anti-inflammatory cytokine IL-10 and self-reported symptoms post-vaccine. Our findings are reassuring in that no significant adverse effect of vaccination was noted and provide evidence to support the current recommendations of reproductive medicine organisations regarding timing of vaccination during fertility treatment.
Collapse
Affiliation(s)
- Laurentina Schaler
- Merrion Fertility Clinic, 60 Lower Mount Street, Dublin 2, Ireland; National Maternity Hospital, Holles Street, Dublin 2, Ireland; School of Medicine, University College Dublin, Dublin 4, Ireland.
| | - Magda Ghanim
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Jordi Guardiola
- Merrion Fertility Clinic, 60 Lower Mount Street, Dublin 2, Ireland
| | - Julia Kaulsay
- Merrion Fertility Clinic, 60 Lower Mount Street, Dublin 2, Ireland
| | - Aya Ibrahim
- Trinity Health Kidney Centre, Trinity Translational Medicine Institute, Trinity College Dublin, St. James' Hospital Campus, Dublin, Ireland; Trinity Translational Medicine Institute, Trinity College Dublin, St. James' Hospital Campus, Dublin 8, Ireland
| | - Gareth Brady
- Trinity Health Kidney Centre, Trinity Translational Medicine Institute, Trinity College Dublin, St. James' Hospital Campus, Dublin, Ireland; Trinity Translational Medicine Institute, Trinity College Dublin, St. James' Hospital Campus, Dublin 8, Ireland
| | - William McCormack
- Trinity Translational Medicine Institute, Trinity College Dublin, St. James' Hospital Campus, Dublin 8, Ireland
| | - Niall Conlon
- Department of Clinical Medicine, Trinity Translational Medicine Institute, School of Medicine, Trinity College Dublin, Dublin 8, Ireland; Department of Immunology, St. James's Hospital, Dublin 8, Ireland
| | - Vincent P Kelly
- School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Mary Wingfield
- Merrion Fertility Clinic, 60 Lower Mount Street, Dublin 2, Ireland; National Maternity Hospital, Holles Street, Dublin 2, Ireland; School of Medicine, University College Dublin, Dublin 4, Ireland; School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland
| | - Louise Glover
- Merrion Fertility Clinic, 60 Lower Mount Street, Dublin 2, Ireland; School of Medicine, University College Dublin, Dublin 4, Ireland; School of Biochemistry & Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, Ireland.
| |
Collapse
|
27
|
Martins YA, Guerra-Gomes IC, Rodrigues TS, Tapparel C, Lopez RFV. Enhancing pulmonary delivery and immunomodulation of respiratory diseases through virus-mimicking nanoparticles. J Control Release 2024; 372:417-432. [PMID: 38908758 DOI: 10.1016/j.jconrel.2024.06.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
This study introduces the nanobromhexine lipid particle (NBL) platform designed for effective pulmonary drug delivery. Inspired by respiratory virus transport mechanisms, NBL address challenges associated with mucus permeation and inflammation in pulmonary diseases. Composed of low molecular weight polyethylene glycol-coated lipid nanoparticles with bromhexine hydrochloride, NBL exhibit a size of 118 ± 24 nm, a neutral zeta potential, osmolarity of 358 ± 28 mOsmol/kg, and a pH of 6.5. Nebulizing without leakage and showing no toxicity to epithelial cells, NBL display mucoadhesive properties with a 60% mucin-binding efficiency. They effectively traverse the dense mucus layer of Calu-3 cultures in an air-liquid interface, as supported by a 55% decrease in MUC5AC density and a 29% increase in nanoparticles internalization compared to non-exposed cells. In assessing immunomodulatory effects, NBL treatment in SARS-CoV-2-infected lung cells leads to a 40-fold increase in anti-inflammatory MUC1 gene expression, a proportional reduction in pro-inflammatory IL-6 expression, and elevated anti-inflammatory IL-10 expression. These findings suggest a potential mechanism to regulate the excessive IL-6 expression triggered by virus infection. Therefore, the NBL platform demonstrates promising potential for efficient pulmonary drug delivery and immunomodulation, offering a novel approach to addressing mucus permeation and inflammation in pulmonary diseases.
Collapse
Affiliation(s)
- Yugo Araújo Martins
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-900, Brazil
| | - Isabel Cristina Guerra-Gomes
- Fundação Oswaldo Cruz - FIOCRUZ, Bi-Institutional Translational Medicine Plataform, Ribeirão Preto, SP 14040-030, Brazil
| | - Tamara Silva Rodrigues
- Department of Biochemistry and Imumunology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Caroline Tapparel
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva 1211, Switzerland
| | - Renata Fonseca Vianna Lopez
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-900, Brazil.
| |
Collapse
|
28
|
Balachandran H, Kroll K, Terry K, Manickam C, Jones R, Woolley G, Hayes T, Martinot AJ, Sharma A, Lewis M, Jost S, Reeves RK. NK cells modulate in vivo control of SARS-CoV-2 replication and suppression of lung damage. PLoS Pathog 2024; 20:e1012439. [PMID: 39133756 PMCID: PMC11341101 DOI: 10.1371/journal.ppat.1012439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/22/2024] [Accepted: 07/22/2024] [Indexed: 08/18/2024] Open
Abstract
Natural killer (NK) cells play a critical role in virus control. However, it has remained largely unclear whether NK cell mobilization in SARS-CoV-2 infections is beneficial or pathologic. To address this deficit, we employed a validated experimental NK cell depletion non-human primate (NHP) model with SARS-CoV-2 Delta variant B.1.617.2 challenge. Viral loads (VL), NK cell numbers, activation, proliferation, and functional measures were evaluated in blood and tissues. In non-depleted (control) animals, infection rapidly induced NK cell expansion, activation, and increased tissue trafficking associated with VL. Strikingly, we report that experimental NK cell depletion leads to higher VL, longer duration of viral shedding, significantly increased levels of pro-inflammatory cytokines in the lungs, and overt lung damage. Overall, we find the first significant and conclusive evidence for NK cell-mediated control of SARS-CoV-2 virus replication and disease pathology. These data indicate that adjunct therapies for infection could largely benefit from NK cell-targeted approaches.
Collapse
Affiliation(s)
- Harikrishnan Balachandran
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Kyle Kroll
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Karen Terry
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Cordelia Manickam
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Rhianna Jones
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Griffin Woolley
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Tammy Hayes
- Department of Infectious Diseases and Global Health, Cummings School of Veterinary Medicine at Tufts University, North Grafton, Massachusetts, United States of America
| | - Amanda J. Martinot
- Department of Infectious Diseases and Global Health, Cummings School of Veterinary Medicine at Tufts University, North Grafton, Massachusetts, United States of America
| | - Ankur Sharma
- BIOQUAL, Inc., Rockville, Maryland, United States of America
| | - Mark Lewis
- BIOQUAL, Inc., Rockville, Maryland, United States of America
| | - Stephanie Jost
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - R. Keith Reeves
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Department of Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| |
Collapse
|
29
|
Maimunah U, Kholili U, Vidyani A, Sugihartono T, Tanaya WM, Wessels FI, Alshawsh MA, Miftahussurur M. Association between COVID-19 severity with liver abnormalities: A retrospective study in a referral hospital in Indonesia. NARRA J 2024; 4:e816. [PMID: 39280282 PMCID: PMC11391993 DOI: 10.52225/narra.v4i2.816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 06/18/2024] [Indexed: 09/18/2024]
Abstract
Coronavirus disease 2019 (COVID-19) is characterized by an acute respiratory infection with multisystem involvement and the association of its severity to liver function abnormalities is not well characterized. The aim of this study was to assess the association between the severity of COVID-19 patients and liver function abnormalities. This retrospective study included adult patients with confirmed COVID-19, which were classified as non-severe or severe according to World Health Organization guidelines. Liver function test results were compared between the severity groups. A total of 339 patients were included of which 150 (44.25%) were severe cases. The male-to-female ratio was 0.9:1 and 3:2 in the non-severe and severe groups, respectively (p=0.031). Aspartate aminotransferase (AST), alanine transaminase (ALT), and total bilirubin levels and acute liver injury (ALI) incidence were significantly higher in the severe group compared to non-severe group (p<0.001, p<0.001, p=0.025, p=0.014, respectively). In contrast, albumin levels were significantly lower (p=0.001). Multivariate analysis showed that ALI was significantly associated with human immunodeficiency virus (HIV) infection (odds ratio (OR): 5.275; 95% confidence interval (CI): 1.165-23.890, p=0.031), hemoglobin level (OR: 1.214; 95%CI: 1.083-1.361, p=0.001), and hypoalbuminemia (OR: 2.627; 95%CI: 1.283-5.379, p=0.008). Pre-existing liver diseases were present in 6.5% of patients. No significant differences were observed between the groups based on COVID-19 severity and ALI presence. Liver function test abnormalities, including ALI, are more prevalent in patients with severe COVID-19 infection. HIV infection, high hemoglobin levels, and hypoalbuminemia may be potential risk factors for ALI.
Collapse
Affiliation(s)
- Ummi Maimunah
- Division of Gastroenterohepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Division of Gastroenterohepatology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Ulfa Kholili
- Division of Gastroenterohepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Division of Gastroenterohepatology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Amie Vidyani
- Division of Gastroenterohepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Division of Gastroenterohepatology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Titong Sugihartono
- Division of Gastroenterohepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Division of Gastroenterohepatology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Willa M Tanaya
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Firda I Wessels
- Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Mohammed A Alshawsh
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Muhammad Miftahussurur
- Division of Gastroenterohepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Division of Gastroenterohepatology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
- Helicobacter pylori and Microbiota Study Group, Institute Tropical Disease, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
30
|
Zhang J. Immune responses in COVID-19 patients: Insights into cytokine storms and adaptive immunity kinetics. Heliyon 2024; 10:e34577. [PMID: 39149061 PMCID: PMC11325674 DOI: 10.1016/j.heliyon.2024.e34577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 08/17/2024] Open
Abstract
SARS-CoV-2 infection can trigger cytokine storm in some patients, which characterized by an excessive production of cytokines and chemical mediators. This hyperactive immune response may cause significant tissue damage and multiple organ failure (MOF). The severity of COVID-19 correlates with the intensity of cytokine storm, involving elements such as IFN, NF-κB, IL-6, HMGB1, etc. It is imperative to rapidly engage adaptive immunity to effectively control the disease progression. CD4+ T cells facilitate an immune response by improving B cells in the production of neutralizing antibodies and activating CD8+ T cells, which are instrumental in eradicating virus-infected cells. Meanwhile, antibodies from B cells can neutralize virus, obstructing further infection of host cells. In individuals who have recovered from the disease, virus-specific antibodies and memory T cells were observed, which could confer a level of protection, reducing the likelihood of re-infection or attenuating severity. This paper discussed the roles of macrophages, IFN, IL-6 and HMGB1 in cytokine release syndrome (CRS), the intricacies of adaptive immunity, and the persistence of immune memory, all of which are critical for the prevention and therapeutic strategies against COVID-19.
Collapse
Affiliation(s)
- Junguo Zhang
- Pulmonology Department, Fengdu General Hospital, Chongqing, 408200, China
| |
Collapse
|
31
|
Gürcü S, Irmak Kaya Z, Uncu A, Yorulmaz G, Ilgin S. Evaluation of serum neopterin levels in severe COVID-19 patients: An observational study. Medicine (Baltimore) 2024; 103:e38996. [PMID: 39058886 PMCID: PMC11272344 DOI: 10.1097/md.0000000000038996] [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: 11/22/2023] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
In patients with coronavirus disease (COVID-19), a massive inflammatory response is a significant cause of morbidity and mortality. Inflammatory markers are prognostic indicators of disease severity and the ultimate clinical outcome. Several studies have demonstrated a correlation between serum levels of neopterin, which can be an immune system marker, disease severity, and poor outcomes in COVID-19 patients. Our study aimed to determine the diagnostic significance of neopterin in conjunction with routinely measured inflammatory markers in patients with severe COVID-19. Serum neopterin, C-reactive protein (CRP), albumin levels, and complete blood count were determined in 39 patients with severe COVID-19 and 30 healthy individuals. Demographic characteristics, serum neopterin levels, and other laboratory data were compared between patients and healthy volunteers and statistically analyzed. High neopterin levels were observed in patients with severe COVID-19 compared to healthy volunteers. Furthermore, albumin levels were decreased, while CRP levels were increased in patients, statistically significantly. Also, positive correlations were shown between serum neopterin levels and serum CRP levels, while negative correlations were shown between serum neopterin levels and serum albumin levels. Systemic inflammation markers, CRP/albumin ratio, neutrophil/lymphocyte ratio, and platelet/lymphocyte ratio were significantly higher, while lymphocyte/monocyte ratio was also significantly lower in patients with severe COVID-19 than in healthy volunteers. However, serum neopterin levels were not linked to the CRP/albumin ratio, the neutrophil/lymphocyte ratio, or the platelet/lymphocyte ratio. On the other hand, they were linked negatively to the lymphocyte/monocyte ratio. Our findings highlight the association between high neopterin levels and patients with severe COVID-19. Neopterin is correlated with traditional inflammatory biomarkers and may indicate general immune and inflammatory activation in patients with severe COVID-19.
Collapse
Affiliation(s)
- Sinem Gürcü
- Department of Pharmacy, Eskişehir City Hospital, Eskişehir, Turkey
| | - Zeynep Irmak Kaya
- Department of Internal Medicine, University of Health Sciences, Eskişehir Health Application and Research Center, Eskişehir, Turkey
| | - Ali Uncu
- Department of Medical Biochemistry, Department of Basic Medical Sciences, University of Health Sciences Eskişehir Health Application and Research Center, Eskişehir, Turkey
| | - Göknur Yorulmaz
- Department of Endocrinology, Faculty of Medicine, Eskisehir Osmangazi University, Eskişehir, Turkey
| | - Sinem Ilgin
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| |
Collapse
|
32
|
Sasidharan A, Grosche A, Xu X, Kinane TB, Angoli D, Vidyasagar S. Select amino acids recover cytokine-altered ENaC function in human bronchial epithelial cells. PLoS One 2024; 19:e0307809. [PMID: 39052685 PMCID: PMC11271875 DOI: 10.1371/journal.pone.0307809] [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: 10/01/2023] [Accepted: 07/11/2024] [Indexed: 07/27/2024] Open
Abstract
The airway epithelium plays a pivotal role in regulating mucosal immunity and inflammation. Epithelial barrier function, homeostasis of luminal fluid, and mucociliary clearance are major components of mucosal defense mechanisms. The epithelial sodium channel (ENaC) is one of the key players in controlling airway fluid volume and composition, and characteristic cytokines cause ENaC and barrier dysfunctions following pulmonary infections or allergic reactions. Given the limited understanding of the requisite duration and magnitude of cytokines to affect ENaC and barrier function, available treatment options for restoring normal ENaC activity are limited. Previous studies have demonstrated that distinct amino acids can modulate epithelial ion channel activities and barrier function in intestines and airways. Here, we have investigated the time- and concentration-dependent effect of representative cytokines for Th1- (IFN-γ and TNF-α), Th2- (IL-4 and IL-13), and Treg-mediated (TGF-β1) immune responses on ENaC activity and barrier function in human bronchial epithelial cells. When cells were exposed to Th1 and Treg cytokines, ENaC activity decreased gradually while barrier function remained largely unaffected. In contrast, Th2 cytokines had an immediate and profound inhibitory effect on ENaC activity that was subsequently followed by epithelial barrier disruption. These functional changes were associated with decreased membrane protein expression of α-, β-, and γ-ENaC, and decreased mRNA levels of β- and γ-ENaC. A proprietary blend of amino acids was developed based on their ability to prevent Th2 cytokine-induced ENaC dysfunction. Exposure to the select amino acids reversed the inhibitory effect of IL-13 on ENaC activity by increasing mRNA levels of β- and γ-ENaC, and protein expression of γ-ENaC. This study indicates the beneficial effect of select amino acids on ENaC activity in an in vitro setting of Th2-mediated inflammation suggesting these amino acids as a novel therapeutic approach for correcting this condition.
Collapse
Affiliation(s)
- Anusree Sasidharan
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - Astrid Grosche
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - Xiaodong Xu
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| | - T. Bernard Kinane
- Pediatric Pulmonary Division, Massachusetts General Hospital for Children, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
| | - Damiano Angoli
- Pediatric Pulmonary Division, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Sadasivan Vidyasagar
- Department of Radiation Oncology, Shands Cancer Center, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
33
|
Singh M, Verma H, Gera N, Baddipadige R, Choudhary S, Bhandu P, Silakari O. Evaluation of Cordyceps militaris steroids as anti-inflammatory agents to combat the Covid-19 cytokine storm: a bioinformatics and structure-based drug designing approach. J Biomol Struct Dyn 2024; 42:5159-5177. [PMID: 37551029 DOI: 10.1080/07391102.2023.2245039] [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: 11/29/2022] [Accepted: 06/08/2023] [Indexed: 08/09/2023]
Abstract
Since the SARS-CoV-2 epidemic, researchers have been working on figuring out ways to tackle multi-organ failure and hyperinflation, which are brought on by a cytokine storm. Angiotensin-converting enzyme 2 (ACE2), a SARS-CoV-2 spike glycoprotein's cellular receptor, is involved in complicated molecular processes that result in hyperinflammation. Cordyceps militaris is one of the traditional Chinese medicines that is used as an immune booster, and it has exhibited efficacy in lowering blood glucose levels, seminal emissions, and infertility. In the current study, we explored the potential of Cordyceps militaris steroids as key agents in managing the anger of cytokine storm in Covid-19 using network ethnopharmacological techniques and structure-based drug designing approaches. The steroids present in Cordyceps militaris were initially screened against the targets involved in inflammatory pathways. The results revealed that out of 16 steroids, 5 may be effective against 17 inflammatory pathways by targeting 11 pathological proteins. Among the five steroids, beta-sitosterol, Cholest-5-en-3β-ol, 3β, and 7α-Dihydroxycholest-5-ene were found to interact with thrombin (F2), an important protein reported to reduce the severity of inflammatory mediators and Cholest-4-en-3-one was found to target Glucocorticoid receptor (NR3C1). The top docked steroid displayed key interactions with both targets, which retained key interactions throughout the 100 ns simulation period. These compounds were also shown high binding free energy scores in water swap studies. Based on obtained results the current study suggests the use of Cordyceps militaris as an add-on therapy that may reduce the progression of inflammatory co-morbidities among patients infected with SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Manmeet Singh
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Himanshu Verma
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Narendra Gera
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Raju Baddipadige
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Shalki Choudhary
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Priyanka Bhandu
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Om Silakari
- Molecular Modeling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| |
Collapse
|
34
|
Sun D, Shi Z, Chen H, Du Q, Zhang Y, Wang R, Kong L, Luo W, Lang Y, Wang X, Zhou H. COVID-19 susceptibility, hospitalization and severity and the risk of brain cortical structure: a Mendelian randomization study. QJM 2024; 117:413-421. [PMID: 38195890 DOI: 10.1093/qjmed/hcad291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/18/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Observational studies have reported structural changes in the brains of patients with coronavirus disease 2019 (COVID-19); it remains unclear whether these associations are causal. AIM We evaluated the causal effects of COVID-19 susceptibility, hospitalization and severity on cortical structures. DESIGN Mendelian randomization (MR) study. METHODS Data on the different COVID-19 phenotypes were obtained from the latest large-scale genome-wide association study (R7) of the COVID-19 Host Genetics Initiative. Brain structure data, including cortical thickness (TH) and surface area (SA), were obtained from the ENIGMA Consortium. Additionally, we employed the round 5 dataset released in January 2021 as the validation cohort. The inverse-variance weighted (IVW) method was used as the primary analysis in MR. Sensitivity analyses were conducted to evaluate heterogeneity and pleiotropy. We performed enrichment analysis on the MR analyses that passed the sensitivity analysis filtering. RESULTS After IVW and sensitivity analyses, we observed causal associations between COVID-19 susceptibility and rostral middle frontal SAw (P = 0.0308, β = -39.1236), cuneus THw (P = 0.0170, β = -0.0121), medial orbitofrontal THw (P = 0.0002, β = 0.0225), postcentral THw (P = 0.0217, β = -0.0106), temporal pole THw (P = 0.0077, β = 0.0359), medial orbitofrontal SAnw (P = 0.0106, β = -24.0397), medial orbitofrontal THnw (P = 0.0007, β = 0.0232), paracentral SAnw (P = 0.0483, β = -20.1442), rostral middle frontal SAnw (P = 0.0368, β = -81.9719) and temporal pole THnw (P = 0.0429, β = 0.0353). COVID-19 hospitalization had causal effects on medial orbitofrontal THw (P = 0.0053, β = 0.0063), postcentral THw (P = 0.0143, β = -0.0042), entorhinal THnw (P = 0.0142, β = 0.0142), medial orbitofrontal THnw (P = 0.0147, β = 0.0065) and paracentral SAnw (P = 0.0119, β = -7.9970). COVID-19 severity had causal effects on rostral middle frontal SAw (P = 0.0122, β = -11.8296), medial orbitofrontal THw (P = 0.0155, β = 0.0038), superior parietal THw (P = 0.0291, β = -0.0021), lingual SAnw (P = 0.0202, β = -11.5270), medial orbitofrontal THnw (P = 0.0290, β = 0.0039), paracentral SAnw (P = 0.0180, β = -5.7744) and pars triangularis SAnw (P = 0.0151, β = -5.4520). CONCLUSION Our MR results demonstrate a causal relationship between different COVID-19 phenotypes and cortical structures.
Collapse
Affiliation(s)
- D Sun
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Z Shi
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - H Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Q Du
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Y Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - R Wang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - L Kong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - W Luo
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Y Lang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - X Wang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - H Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
35
|
Han H, Kim JE, Lee HJ. Effect of apigetrin in pseudo-SARS-CoV-2-induced inflammatory and pulmonary fibrosis in vitro model. Sci Rep 2024; 14:14545. [PMID: 38914619 PMCID: PMC11196261 DOI: 10.1038/s41598-024-65447-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: 04/09/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024] Open
Abstract
SARS-CoV-2 has become a global public health problem. Acute respiratory distress syndrome (ARDS) is the leading cause of death due to the SARS-CoV-2 infection. Pulmonary fibrosis (PF) is a severe and frequently reported COVID-19 sequela. In this study, an in vitro model of ARDS and PF caused by SARS-CoV-2 was established in MH-S, THP-1, and MRC-5 cells using pseudo-SARS-CoV-2 (PSCV). Expression of proinflammatory cytokines (IL-6, IL-1β, and TNF-α) and HIF-1α was increased in PSCV-infected MH-S and THP-1 cells, ARDS model, consistent with other profiling data in SARS-CoV-2-infected patients have been reported. Hypoxia-inducible factor-1 alpha (HIF-1α) siRNA and cobalt chloride were tested using this in vitro model. HIF-1α knockdown reduces inflammation caused by PSCV infection in MH-S and THP-1 cells and lowers elevated levels of CTGF, COLA1, and α-SMA in MRC-5 cells exposed to CPMSCV. Furthermore, apigetrin, a glycoside bioactive dietary flavonoid derived from several plants, including Crataegus pinnatifida, which is reported to be a HIF-1α inhibitor, was tested in this in vitro model. Apigetrin significantly reduced the increased inflammatory cytokine (IL-6, IL-1β, and TNF-α) expression and secretion by PSCV in MH-S and THP-1 cells. Apigetrin inhibited the binding of the SARS-CoV-2 spike protein RBD to the ACE2 protein. An in vitro model of PF induced by SARS-CoV-2 was produced using a conditioned medium of THP-1 and MH-S cells that were PSCV-infected (CMPSCV) into MRC-5 cells. In a PF model, CMPSCV treatment of THP-1 and MH-S cells increased cell growth, migration, and collagen synthesis in MRC-5 cells. In contrast, apigetrin suppressed the increase in cell growth, migration, and collagen synthesis induced by CMPSCV in THP-1 and MH-S MRC-5 cells. Also, compared to control, fibrosis-related proteins (CTGF, COLA1, α-SMA, and HIF-1α) levels were over two-fold higher in CMPSV-treated MRC-5 cells. Apigetrin decreased protein levels in CMPSCV-treated MRC-5 cells. Thus, our data suggest that hypoxia-inducible factor-1 alpha (HIF-1α) might be a novel target for SARS-CoV-2 sequela therapies and apigetrin, representative of HIF-1alpha inhibitor, exerts anti-inflammatory and PF effects in PSCV-treated MH-S, THP-1, and CMPVSC-treated MRC-5 cells. These findings indicate that HIF-1α inhibition and apigetrin would have a potential value in controlling SARS-CoV-2-related diseases.
Collapse
Affiliation(s)
- Hengmin Han
- Department of Cancer Preventive Material Development, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - Jung-Eun Kim
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
| | - Hyo-Jeong Lee
- Department of Cancer Preventive Material Development, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea.
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea.
| |
Collapse
|
36
|
Wu B, Li R, Hao J, Qi Y, Liu B, Wei H, Li Z, Zhang Y, Liu Y. CT semi-quantitative score used as risk factor for hyponatremia in patients with COVID-19: a cross-sectional study. Front Endocrinol (Lausanne) 2024; 15:1342204. [PMID: 38948513 PMCID: PMC11211362 DOI: 10.3389/fendo.2024.1342204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
Purpose Chest computed tomography (CT) is used to determine the severity of COVID-19 pneumonia, and pneumonia is associated with hyponatremia. This study aims to explore the predictive value of the semi-quantitative CT visual score for hyponatremia in patients with COVID-19 to provide a reference for clinical practice. Methods In this cross-sectional study, 343 patients with RT-PCR confirmed COVID-19, all patients underwent CT, and the severity of lung lesions was scored by radiologists using the semi-quantitative CT visual score. The risk factors of hyponatremia in COVID-19 patients were analyzed and combined with laboratory tests. The thyroid function changes caused by SARS-CoV-2 infection and their interaction with hyponatremia were also analyzed. Results In patients with SARS-CoV-2 infection, the total severity score (TSS) of hyponatremia was higher [M(range), 3.5(2.5-5.5) vs 3.0(2.0-4.5) scores, P=0.001], implying that patients with hyponatremia had more severe lung lesions. The risk factors of hyponatremia in the multivariate regression model included age, vomiting, neutrophils, platelet, and total severity score. SARS-CoV-2 infection impacted thyroid function, and patients with hyponatremia showed a lower free triiodothyronine (3.1 ± 0.9 vs 3.7 ± 0.9, P=0.001) and thyroid stimulating hormone level [1.4(0.8-2.4) vs 2.2(1.2-3.4), P=0.038]. Conclusion Semi-quantitative CT score can be used as a risk factor for hyponatremia in patients with COVID-19. There is a weak positive correlation between serum sodium and free triiodothyronine in patients with SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Baofeng Wu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Ru Li
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Jinxuan Hao
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yijie Qi
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Botao Liu
- Department of Medical Imaging, Shanxi Medical University, Taiyuan, China
| | - Hongxia Wei
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Zhe Li
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
- First Clinical Medical College, Shanxi Medical University, Taiyuan, China
| | - Yi Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, China
| | - Yunfeng Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
37
|
Lu W, Yan L, Tang X, Wang X, Du J, Zou Z, Li L, Ye J, Zhou L. Efficacy and safety of mesenchymal stem cells therapy in COVID-19 patients: a systematic review and meta-analysis of randomized controlled trials. J Transl Med 2024; 22:550. [PMID: 38851730 PMCID: PMC11162060 DOI: 10.1186/s12967-024-05358-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) has become a serious public health issue. In COVID-19 patients, the elevated levels of inflammatory cytokines lead to the manifestation of COVID-19 symptoms, such as lung tissue edema, lung diffusion dysfunction, acute respiratory distress syndrome (ARDS), secondary infection, and ultimately mortality. Mesenchymal stem cells (MSCs) exhibit anti-inflammatory and immunomodulatory properties, thus providing a potential treatment option for COVID-19. The number of clinical trials of MSCs for COVID-19 has been rising. However, the treatment protocols and therapeutic effects of MSCs for COVID-19 patients are inconsistent. This meta-analysis was performed to systematically determine the safety and efficacy of MSC infusion in COVID-19 patients. METHODS We conducted a comprehensive literature search from PubMed/Medline, Web of Science, EMBASE, and Cochrane Library up to 22 November 2023 to screen for eligible randomized controlled trials. Inclusion and exclusion criteria for searched literature were formulated according to the PICOS principle, followed by the use of literature quality assessment tools to assess the risk of bias. Finally, outcome measurements including therapeutic efficacy, clinical symptoms, and adverse events of each study were extracted for statistical analysis. RESULTS A total of 14 randomized controlled trials were collected. The results of enrolled studies demonstrated that patients with COVID-19 pneumonia who received MSC inoculation showed a decreased mortality compared with counterparts who received conventional treatment (RR: 0.76; 95% CI [0.60, 0.96]; p = 0.02). Reciprocally, MSC inoculation improved the clinical symptoms in patients (RR: 1.28; 95% CI [1.06, 1.55]; p = 0.009). In terms of immune biomarkers, MSC treatment inhibited inflammation responses in COVID-19 patients, as was indicated by the decreased levels of CRP and IL-6. Importantly, our results showed that no significant differences in the incidence of adverse reactions or serious adverse events were monitored in patients after MSC inoculation. CONCLUSION This meta-analysis demonstrated that MSC inoculation is effective and safe in the treatment of patients with COVID-19 pneumonia. Without increasing the incidence of adverse events or serious adverse events, MSC treatment decreased patient mortality and inflammatory levels and improved the clinical symptoms in COVID-19 patients. However, large-cohort randomized controlled trials with expanded numbers of patients are required to further confirm our results.
Collapse
Affiliation(s)
- Wenming Lu
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- School of Rehabilitation Medicine, Gannan Medical University, GanZhou City, 341000, Jiangxi, People's Republic of China
- The First Clinical College of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Longxiang Yan
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- School of Rehabilitation Medicine, Gannan Medical University, GanZhou City, 341000, Jiangxi, People's Republic of China
- The First Clinical College of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Xingkun Tang
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- School of Rehabilitation Medicine, Gannan Medical University, GanZhou City, 341000, Jiangxi, People's Republic of China
| | - Xuesong Wang
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- School of Rehabilitation Medicine, Gannan Medical University, GanZhou City, 341000, Jiangxi, People's Republic of China
| | - Jing Du
- School of Rehabilitation Medicine, Gannan Medical University, GanZhou City, 341000, Jiangxi, People's Republic of China
| | - Zhengwei Zou
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- Ganzhou Key Laboratory of Stem Cell and Regenerative Medicine, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Lincai Li
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- Ganzhou Key Laboratory of Stem Cell and Regenerative Medicine, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Junsong Ye
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- Ganzhou Key Laboratory of Stem Cell and Regenerative Medicine, Ganzhou, 341000, Jiangxi, People's Republic of China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
- Jiangxi Provincal Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Lin Zhou
- Subcenter for Stem Cell Clinical Translation, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China.
- Ganzhou Key Laboratory of Stem Cell and Regenerative Medicine, Ganzhou, 341000, Jiangxi, People's Republic of China.
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China.
- Jiangxi Provincal Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China.
| |
Collapse
|
38
|
Saito M, Iijima T, Kurai H, Aso Y. Thyroid storm after coronavirus disease 2019 mRNA vaccination in a patient with a history of Graves' disease after coronavirus disease 2019 infection. BMJ Case Rep 2024; 17:e259796. [PMID: 38834307 DOI: 10.1136/bcr-2024-259796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024] Open
Abstract
We report a case where the patient may have developed Graves' disease after COVID-19 infection, and where the COVID-19 vaccination may have exacerbated the condition, inducing the onset of a thyroid storm. Although any association between the vaccine and the onset of thyroid disease is impossible to demonstrate through a single case, the antecedent COVID-19 infection and COVID-19 messenger ribonucleic acid vaccination may have synergistically contributed to the development of Graves' disease followed by thyroid storm.
Collapse
Affiliation(s)
- Masahiro Saito
- Endocrinology and Metabolism, Dokkyo Medical University, Shimotsuga-gun, Japan
| | - Toshie Iijima
- Endocrinology and Metabolism, Dokkyo Medical University, Shimotsuga-gun, Japan
| | - Hidetaka Kurai
- Endocrinology and Metabolism, Dokkyo Medical University, Shimotsuga-gun, Japan
| | - Yosimasa Aso
- Endocrinology and Metabolism, Dokkyo Medical University, Shimotsuga-gun, Japan
| |
Collapse
|
39
|
Chittasupho C, Umsumarng S, Srisawad K, Arjsri P, Phongpradist R, Samee W, Tingya W, Ampasavate C, Dejkriengkraikul P. Inhibition of SARS-CoV-2-Induced NLRP3 Inflammasome-Mediated Lung Cell Inflammation by Triphala-Loaded Nanoparticle Targeting Spike Glycoprotein S1. Pharmaceutics 2024; 16:751. [PMID: 38931873 PMCID: PMC11206841 DOI: 10.3390/pharmaceutics16060751] [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: 04/18/2024] [Revised: 05/24/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, poses a significant global health threat. The spike glycoprotein S1 of the SARS-CoV-2 virus is known to induce the production of pro-inflammatory mediators, contributing to hyperinflammation in COVID-19 patients. Triphala, an ancient Ayurvedic remedy composed of dried fruits from three plant species-Emblica officinalis (Family Euphorbiaceae), Terminalia bellerica (Family Combretaceae), and Terminalia chebula (Family Combretaceae)-shows promise in addressing inflammation. However, the limited water solubility of its ethanolic extract impedes its bioavailability. In this study, we aimed to develop nanoparticles loaded with Triphala extract, termed "nanotriphala", as a drug delivery system. Additionally, we investigated the in vitro anti-inflammatory properties of nanotriphala and its major compounds, namely gallic acid, chebulagic acid, and chebulinic acid, in lung epithelial cells (A549) induced by CoV2-SP. The nanotriphala formulation was prepared using the solvent displacement method. The encapsulation efficiency of Triphala in nanotriphala was determined to be 87.96 ± 2.60% based on total phenolic content. In terms of in vitro release, nanotriphala exhibited a biphasic release profile with zero-order kinetics over 0-8 h. A549 cells were treated with nanotriphala or its active compounds and then induced with 100 ng/mL of spike S1 subunit (CoV2-SP). The results demonstrate that chebulagic acid and chebulinic acid are the active compounds in nanotriphala, which significantly reduced cytokine release (IL-6, IL-1β, and IL-18) and suppressed the expression of inflammatory genes (IL-6, IL-1β, IL-18, and NLRP3) (p < 0.05). Mechanistically, nanotriphala and its active compounds notably attenuated the expression of inflammasome machinery proteins (NLRP3, ASC, and Caspase-1) (p < 0.05). In conclusion, the nanoparticle formulation of Triphala enhances its stability and exhibits anti-inflammatory properties against CoV2-SP-induction. This was achieved by suppressing inflammatory mediators and the NLRP3 inflammasome machinery. Thus, nanotriphala holds promise as a supportive preventive anti-inflammatory therapy for COVID-19-related chronic inflammation.
Collapse
Affiliation(s)
- Chuda Chittasupho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (R.P.); (W.T.); (C.A.)
| | - Sonthaya Umsumarng
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand;
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kamonwan Srisawad
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (P.A.)
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Punnida Arjsri
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (P.A.)
| | - Rungsinee Phongpradist
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (R.P.); (W.T.); (C.A.)
| | - Weerasak Samee
- Department of Pharmaceutical Chemistry, Srinakharinwirot University, Ongkharak, Nakhon Nayok 26120, Thailand;
| | - Wipawan Tingya
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (R.P.); (W.T.); (C.A.)
| | - Chadarat Ampasavate
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand; (C.C.); (R.P.); (W.T.); (C.A.)
| | - Pornngarm Dejkriengkraikul
- Center for Research and Development of Natural Products for Health, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (K.S.); (P.A.)
- Anticarcinogenesis and Apoptosis Research Cluster, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
40
|
Kozłowski P, Leszczyńska A, Ciepiela O. Long COVID Definition, Symptoms, Risk Factors, Epidemiology and Autoimmunity: A Narrative Review. AMERICAN JOURNAL OF MEDICINE OPEN 2024; 11:100068. [PMID: 39034937 PMCID: PMC11256271 DOI: 10.1016/j.ajmo.2024.100068] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/29/2024] [Accepted: 02/01/2024] [Indexed: 07/23/2024]
Abstract
The virus called SARS-CoV-2 emerged in 2019 and quickly spread worldwide, causing COVID-19. It has greatly impacted on everyday life, healthcare systems, and the global economy. In order to save as many lives as possible, precautions such as social distancing, quarantine, and testing policies were implemented, and effective vaccines were developed. A growing amount of data collected worldwide allowed the characterization of this new disease, which turned out to be more complex than other common respiratory tract infections. An increasing number of convalescents presented with a variety of nonspecific symptoms emerging after the acute infection. This possible new global health problem was identified and labelled as long COVID. Since then, a great effort has been made by clinicians and the scientific community to understand the underlying mechanisms and to develop preventive measures and effective treatment. The role of autoimmunity induced by SARS-CoV-2 infection in the development of long COVID is discussed in this review. We aim to deliver a description of several conditions with an autoimmune background observed in COVID-19 convalescents, including Guillain-Barré syndrome, antiphospholipid syndrome and related thrombosis, and Kawasaki disease highlighting a relationship between SARS-CoV-2 infection and the development of autoimmunity. However, further studies are required to determine its true clinical significance.
Collapse
Affiliation(s)
- Paweł Kozłowski
- Central Laboratory, University Clinical Centre of the Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Leszczyńska
- Central Laboratory, University Clinical Centre of the Medical University of Warsaw, Warsaw, Poland
| | - Olga Ciepiela
- Central Laboratory, University Clinical Centre of the Medical University of Warsaw, Warsaw, Poland
- Department of Laboratory Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
41
|
Ahmad S, Singh AP, Bano N, Raza K, Singh J, Medigeshi GR, Pandey R, Gautam HK. Integrative analysis discovers Imidurea as dual multitargeted inhibitor of CD69, CD40, SHP2, lysozyme, GATA3, cCBL, and S-cysteinase from SARS-CoV-2 and M. tuberculosis. Int J Biol Macromol 2024; 270:132332. [PMID: 38768914 DOI: 10.1016/j.ijbiomac.2024.132332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024]
Abstract
Two of the deadliest infectious diseases, COVID-19 and tuberculosis (TB), have combined to establish a worldwide pandemic, wreaking havoc on economies and claiming countless lives. The optimised, multitargeted medications may diminish resistance and counter them together. Based on computational expression studies, 183 genes were co-expressed in COVID-19 and TB blood samples. We used the multisampling screening algorithms on the top ten co-expressed genes (CD40, SHP2, Lysozyme, GATA3, cCBL, SIVmac239 Nef, CD69, S-adenosylhomocysteinase, Chemokine Receptor-7, and Membrane Protein). Imidurea is a multitargeted inhibitor for COVID-19 and TB, as confirmed by extensive screening and post-filtering utilising MM\GBSA algorithms. Imidurea has shown docking and MM\GBSA scores of -8.21 to -4.75 Kcal/mol and -64.16 to -29.38 Kcal/mol, respectively. The DFT, pharmacokinetics, and interaction patterns suggest that Imidurea may be a drug candidate, and all ten complexes were tested for stability and bond strength using 100 ns for all MD atoms. The modelling findings showed the complex's repurposing potential, with a cumulative deviation and fluctuation of <2 Å and significant intermolecular interaction, which validated the possibilities. Finally, an inhibition test was performed to confirm our in-silico findings on SARS-CoV-2 Delta variant infection, which was suppressed by adding imidurea to Vero E6 cells after infection.
Collapse
Affiliation(s)
- Shaban Ahmad
- Computational Intelligence and Bioinformatics Lab, Department of Computer Science, Jamia Millia Islamia, New Delhi 110025, India.
| | - Akash Pratap Singh
- Division of Immunology and Infectious Disease Biology, Institute of Genomics and Integrative Biology (IGIB), Mathura Road, New Delhi 110025, India; Academy of Innovative and Scientific Research (AcSIR), Ghaziabad 201002, India; Department of Botany, Maitreyi College, University of Delhi, New Delhi 110021, India.
| | - Nagmi Bano
- Computational Intelligence and Bioinformatics Lab, Department of Computer Science, Jamia Millia Islamia, New Delhi 110025, India.
| | - Khalid Raza
- Computational Intelligence and Bioinformatics Lab, Department of Computer Science, Jamia Millia Islamia, New Delhi 110025, India.
| | - Janmejay Singh
- Bioassay Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India.
| | - Guruprasad R Medigeshi
- Bioassay Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana 121001, India.
| | - Rajesh Pandey
- Academy of Innovative and Scientific Research (AcSIR), Ghaziabad 201002, India; Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE), Institute of Genomics and Integrative Biology (IGIB), Mall Road, New Delhi 110007, India.
| | - Hemant K Gautam
- Division of Immunology and Infectious Disease Biology, Institute of Genomics and Integrative Biology (IGIB), Mathura Road, New Delhi 110025, India; Academy of Innovative and Scientific Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
42
|
Sumichika Y, Temmoku J, Saito K, Yoshida S, Matsumoto H, Watanabe G, Utsumi A, Fujita Y, Matsuoka N, Asano T, Sato S, Migita K. New-onset Systemic Lupus Erythematosus Manifestation Following COVID-19: A Case Report and Literature Review. Intern Med 2024; 63:1491-1498. [PMID: 38369349 PMCID: PMC11157318 DOI: 10.2169/internalmedicine.3211-23] [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: 11/14/2023] [Accepted: 12/24/2023] [Indexed: 02/20/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a respiratory viral disease, and several cases of autoimmune diseases have been reported after infection. This report presents the case of a 38-year-old Japanese woman who developed systemic lupus erythematosus (SLE) following COVID-19. Clinical manifestations included dermatological complications, joint pain, and positive autoantibodies. The patient met the SLE classification criteria, and renal involvement was observed. Her symptoms improved with immunosuppressive therapy. A literature review identified 10 similar cases, those with lymphopenia and renal involvement. SLE should be considered in patients with persistent nonspecific symptoms after COVID-19 infection, particularly when hematologic and renal involvement are present.
Collapse
Affiliation(s)
- Yuya Sumichika
- Department of Rheumatology, Fukushima Medical University School of Medicine, Japan
| | - Jumpei Temmoku
- Department of Rheumatology, Fukushima Medical University School of Medicine, Japan
| | - Kenji Saito
- Department of Rheumatology, Fukushima Medical University School of Medicine, Japan
| | - Shuhei Yoshida
- Department of Rheumatology, Fukushima Medical University School of Medicine, Japan
| | - Haruki Matsumoto
- Department of Rheumatology, Fukushima Medical University School of Medicine, Japan
| | - Guy Watanabe
- Department of Nephrology and Hypertension, Fukushima Medical University School of Medicine, Japan
| | - Akihito Utsumi
- Department of Diagnostic Pathology, Fukushima Medical University, School of Medicine, Japan
| | - Yuya Fujita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Japan
| | - Naoki Matsuoka
- Department of Rheumatology, Fukushima Medical University School of Medicine, Japan
| | - Tomoyuki Asano
- Department of Rheumatology, Fukushima Medical University School of Medicine, Japan
| | - Shuzo Sato
- Department of Rheumatology, Fukushima Medical University School of Medicine, Japan
| | - Kiyoshi Migita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Japan
| |
Collapse
|
43
|
Ramos A, Martins S, Marinho AS, Norton P, Cardoso MJ, Guimarães JT. Evaluation of SARS-CoV-2 interferon gamma release assay in BNT162b2 vaccinated healthcare workers. PLoS One 2024; 19:e0303244. [PMID: 38728294 PMCID: PMC11086832 DOI: 10.1371/journal.pone.0303244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
To predict protective immunity to SARS-CoV-2, cellular immunity seems to be more sensitive than humoral immunity. Through an Interferon-Gamma (IFN-γ) Release Assay (IGRA), we show that, despite a marked decrease in total antibodies, 94.3% of 123 healthcare workers have a positive cellular response 6 months after inoculation with the 2nd dose of BNT162b2 vaccine. Despite the qualitative relationship found, we did not observe a quantitative correlation between IFN-γ and IgG levels against SARS-CoV-2. Using stimulated whole blood from a subset of participants, we confirmed the specific T-cell response to SARS-CoV-2 by dosing elevated levels of the IL-6, IL-10 and TNF-α. Through a 20-month follow-up, we found that none of the infected participants had severe COVID-19 and that the first positive cases were only 12 months after the 2nd dose inoculation. Future studies are needed to understand if IGRA-SARS-CoV-2 can be a powerful diagnostic tool to predict future COVID-19 severe disease, guiding vaccination policies.
Collapse
Affiliation(s)
- Angélica Ramos
- Serviço de Patologia Clínica, Centro Hospitalar Universitário de São João, Porto, Portugal
- EPIUnit–Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
| | - Sandra Martins
- Serviço de Patologia Clínica, Centro Hospitalar Universitário de São João, Porto, Portugal
- EPIUnit–Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
| | - Ana Sofia Marinho
- Serviço de Patologia Clínica, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Pedro Norton
- EPIUnit–Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
- Serviço de Saúde Ocupacional, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Maria João Cardoso
- Serviço de Patologia Clínica, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - João Tiago Guimarães
- Serviço de Patologia Clínica, Centro Hospitalar Universitário de São João, Porto, Portugal
- EPIUnit–Instituto de Saúde Pública, Universidade do Porto, Porto, Portugal
- Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Universidade do Porto, Porto, Portugal
- Departamento de Biomedicina, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| |
Collapse
|
44
|
Choudhary S, Nehul S, Singh A, Panda PK, Kumar P, Sharma GK, Tomar S. Unraveling antiviral efficacy of multifunctional immunomodulatory triterpenoids against SARS-COV-2 targeting main protease and papain-like protease. IUBMB Life 2024; 76:228-241. [PMID: 38059400 DOI: 10.1002/iub.2793] [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: 03/15/2023] [Accepted: 10/20/2023] [Indexed: 12/08/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may be over, but its variants continue to emerge, and patients with mild symptoms having long COVID is still under investigation. SARS-CoV-2 infection leading to elevated cytokine levels and suppressed immune responses set off cytokine storm, fatal systemic inflammation, tissue damage, and multi-organ failure. Thus, drug molecules targeting the SARS-CoV-2 virus-specific proteins or capable of suppressing the host inflammatory responses to viral infection would provide an effective antiviral therapy against emerging variants of concern. Evolutionarily conserved papain-like protease (PLpro) and main protease (Mpro) play an indispensable role in the virus life cycle and immune evasion. Direct-acting antivirals targeting both these viral proteases represent an attractive antiviral strategy that is also expected to reduce viral inflammation. The present study has evaluated the antiviral and anti-inflammatory potential of natural triterpenoids: azadirachtin, withanolide_A, and isoginkgetin. These molecules inhibit the Mpro and PLpro proteolytic activities with half-maximal inhibitory concentrations (IC50) values ranging from 1.42 to 32.7 μM. Isothermal titration calorimetry (ITC) analysis validated the binding of these compounds to Mpro and PLpro. As expected, the two compounds, withanolide_A and azadirachtin, exhibit potent anti-SARS-CoV-2 activity in cell-based assays, with half-maximum effective concentration (EC50) values of 21.73 and 31.19 μM, respectively. The anti-inflammatory roles of azadirachtin and withanolide_A when assessed using HEK293T cells, were found to significantly reduce the levels of CXCL10, TNFα, IL6, and IL8 cytokines, which are elevated in severe cases of COVID-19. Interestingly, azadirachtin and withanolide_A were also found to rescue the decreased type-I interferon response (IFN-α1). The results of this study clearly highlight the role of triterpenoids as effective antiviral molecules that target SARS-CoV-2-specific enzymes and also host immune pathways involved in virus-mediated inflammation.
Collapse
Affiliation(s)
- Shweta Choudhary
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Sanketkumar Nehul
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Ankur Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Prasan Kumar Panda
- Department of Internal Medicine (Division of Infectious diseases), All India Institute of Medical Sciences (AIIMS), Rishikesh, India
| | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Gaurav Kumar Sharma
- Centre for Animal Disease Research and Diagnosis (CADRAD), Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, India
| |
Collapse
|
45
|
Abdeltawab MSA, Fateen M, Saad El-Din S, Elmessiery RM, Mohammady Mohamed O, Marzouk Sadek K, Medhat E, Hamed AMR. Effect of SARS-CoV-2 and Toxoplasma gondii co-infection on IFN-γ and TNF-α expression and its impact on disease severity. Cytokine 2024; 177:156545. [PMID: 38368695 DOI: 10.1016/j.cyto.2024.156545] [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: 12/06/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/20/2024]
Abstract
The symptomatology of COVID-19 is dependent on the immune status and the cytokine response of the host. The cytokine level of the host is influenced by the presence of chronic persistent or latent infections with co-pathogens. Parasitic diseases are known to induce host immune-modulation which may impact the response to co-infection. Toxoplasmosis is a widespread protozoal infection that remains quiescent in its latent form to be re-activated during states of immune depression. Clinical data on the relation between toxoplasmosis and COVID-19 cytokine profile and symptomatology are still insufficient. Seventy-nine subjects were included in this study. Patients were diagnosed with COVID-19 by PCR. Serological testing for toxoplasmosis was performed by the detection of anti-Toxoplasma IgG antibodies, in addition to IgG avidity testing. IFN-γ and TNF-α levels were determined by RT-PCR. Among patients diagnosed with COVID-19, 67.1% were seronegative for anti-Toxoplasma IgG, while 32.9% were seropositive. High avidity was found in 10 cases (40% of seropositive cases), 4 of whom required ICU administration, while low avidity was found in 15 cases (60%), 7 of which were administered to the ICU. TNF-α and INF-γ levels were significantly higher in COVID-19 patients than in healthy control subjects. No significant association was found between the seroprevalence of toxoplasmosis and the presence of COVID-19 and its severity. Cytokines were significantly higher in both seropositive and seronegative COVID-19 patients than in their control counterparts. The high prevalence of toxoplasmosis merits further exploration of its relation to COVID-19 by mass studies.
Collapse
Affiliation(s)
| | - Mohamed Fateen
- Clinical and Chemical Pathology Department, Faculty of Medicine, Cairo University, Egypt
| | - Shimaa Saad El-Din
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Egypt
| | - Riem M Elmessiery
- Internal Medicine Department, Faculty of Medicine, Cairo University, Egypt
| | | | | | - Engy Medhat
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Egypt
| | - Alshaimaa M R Hamed
- Medical Parasitology Department, Faculty of Medicine, Cairo University, Egypt
| |
Collapse
|
46
|
Altay Benetti A, Tan EYZ, Chang ZW, Bae KH, Thwin MT, Muthuramalingam RPK, Liao KC, Wan Y, Ng LFP, Renia L, Liu J, Chen X, Yang YY, White KP, Pastorin G. Design and Characterization of a New Formulation for the Delivery of COVID-19-mRNA Vaccine to the Nasal Mucosa. Vaccines (Basel) 2024; 12:409. [PMID: 38675792 PMCID: PMC11054997 DOI: 10.3390/vaccines12040409] [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: 03/14/2024] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Chitosan, a natural polysaccharide derived from chitin, possesses biocompatibility, biodegradability, and mucoadhesive characteristics, making it an attractive material for the delivery of mRNA payloads to the nasal mucosa and promoting their uptake by target cells such as epithelial and immune cells (e.g., dendritic cells and macrophages). In this project, we aimed at developing novel lipid-based nanoformulations for mRNA delivery to counteract the pandemic caused by SARS-CoV-2 virus. The formulations achieved a mRNA encapsulation efficiency of ~80.2% with chitosan-lipid nanoparticles, as measured by the RiboGreen assay. Furthermore, the evaluation of SARS-CoV-2 Spike (S) receptor-binding domain (RBD) expression via ELISA for our vaccine formulations showed transfection levels in human embryonic kidney cells (HEK 293), lung carcinoma cells (A549), and dendritic cells (DC 2.4) equal to 9.9 ± 0.1 ng/mL (174.7 ± 1.1 fold change from untreated cells (UT)), 7.0 ± 0.2 ng/mL (128.1 ± 4.9 fold change from UT), and 0.9 ± 0.0 ng/mL (18.0 ± 0.1 fold change from UT), respectively. Our most promising vaccine formulation was also demonstrated to be amenable to lyophilization with minimal degradation of loaded mRNA, paving the way towards a more accessible and stable vaccine. Preliminary in vivo studies in mice were performed to assess the systemic and local immune responses. Nasal bronchoalveolar lavage fluid (BALF) wash showed that utilizing the optimized formulation resulted in local antibody concentrations and did not trigger any systemic antibody response. However, if further improved and developed, it could potentially contribute to the management of COVID-19 through nasopharyngeal immunization strategies.
Collapse
Affiliation(s)
- Ayça Altay Benetti
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117544, Singapore; (A.A.B.); (E.Y.Z.T.); (M.T.T.); (R.P.K.M.)
| | - Eugene Yang Zhi Tan
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117544, Singapore; (A.A.B.); (E.Y.Z.T.); (M.T.T.); (R.P.K.M.)
| | - Zi Wei Chang
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore 138632, Singapore
| | - Ki Hyun Bae
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore; (K.H.B.); (Y.Y.Y.)
| | - Ma Thinzar Thwin
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117544, Singapore; (A.A.B.); (E.Y.Z.T.); (M.T.T.); (R.P.K.M.)
| | - Ram Pravin Kumar Muthuramalingam
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117544, Singapore; (A.A.B.); (E.Y.Z.T.); (M.T.T.); (R.P.K.M.)
| | - Kuo-Chieh Liao
- Genome Institute of Singapore, Singapore 138672, Singapore; (K.-C.L.)
| | - Yue Wan
- Genome Institute of Singapore, Singapore 138672, Singapore; (K.-C.L.)
| | - Lisa F. P. Ng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore 138632, Singapore
| | - Laurent Renia
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore 138632, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore 639798, Singapore
| | - Jianping Liu
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore; (J.L.); (X.C.)
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117544, Singapore
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore; (J.L.); (X.C.)
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117544, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138632, Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore; (K.H.B.); (Y.Y.Y.)
| | - Kevin P. White
- Precision Medicine Translational Research Program and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| | - Giorgia Pastorin
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, Singapore 117544, Singapore; (A.A.B.); (E.Y.Z.T.); (M.T.T.); (R.P.K.M.)
| |
Collapse
|
47
|
Igwe JK, Alaribe U. Cannabis use associated with lower mortality among hospitalized Covid-19 patients using the national inpatient sample: an epidemiological study. J Cannabis Res 2024; 6:18. [PMID: 38582889 PMCID: PMC10998318 DOI: 10.1186/s42238-024-00228-w] [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: 11/09/2023] [Accepted: 03/20/2024] [Indexed: 04/08/2024] Open
Abstract
BACKGROUND Prior reports indicate that modulation of the endocannabinoid system (ECS) may have a protective benefit for Covid-19 patients. However, associations between cannabis use (CU) or CU not in remission (active cannabis use (ACU)), and Covid-19-related outcomes among hospitalized patients is unknown. METHODS In this multicenter retrospective observational cohort analysis of adults (≥ 18 years-old) identified from 2020 National Inpatient Sample database, we utilize multivariable regression analyses and propensity score matching analysis (PSM) to analyze trends and outcomes among Covid-19-related hospitalizations with CU and without CU (N-CU) for primary outcome of interest: Covid-19-related mortality; and secondary outcomes: Covid-19-related hospitalization, mechanical ventilation (MV), and acute pulmonary embolism (PE) compared to all-cause admissions; for CU vs N-CU; and for ACU vs N-ACU. RESULTS There were 1,698,560 Covid-19-related hospitalizations which were associated with higher mortality (13.44% vs 2.53%, p ≤ 0.001) and worse secondary outcomes generally. Among all-cause hospitalizations, 1.56% of CU and 6.29% of N-CU were hospitalized with Covid-19 (p ≤ 0.001). ACU was associated with lower odds of MV, PE, and death among the Covid-19 population. On PSM, ACU(N(unweighted) = 2,382) was associated with 83.97% lower odds of death compared to others(N(unweighted) = 282,085) (2.77% vs 3.95%, respectively; aOR:0.16, [0.10-0.25], p ≤ 0.001). CONCLUSIONS These findings suggest that the ECS may represent a viable target for modulation of Covid-19. Additional studies are needed to further explore these findings.
Collapse
Affiliation(s)
- Joseph-Kevin Igwe
- Department of Medicine, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA.
| | - Ugo Alaribe
- Caribbean Medical University School of Medicine, 5600 N River Rd Suite 800, Rosemont, IL, 60018, USA
| |
Collapse
|
48
|
Hamdorf M, Imhof T, Bailey-Elkin B, Betz J, Theobald SJ, Simonis A, Di Cristanziano V, Gieselmann L, Dewald F, Lehmann C, Augustin M, Klein F, Alejandre Alcazar MA, Rongisch R, Fabri M, Rybniker J, Goebel H, Stetefeld J, Brachvogel B, Cursiefen C, Koch M, Bock F. The unique ORF8 protein from SARS-CoV-2 binds to human dendritic cells and induces a hyper-inflammatory cytokine storm. J Mol Cell Biol 2024; 15:mjad062. [PMID: 37891014 PMCID: PMC11181941 DOI: 10.1093/jmcb/mjad062] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 02/01/2023] [Accepted: 10/26/2023] [Indexed: 10/29/2023] Open
Abstract
The novel coronavirus pandemic, first reported in December 2019, was caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). SARS-CoV-2 infection leads to a strong immune response and activation of antigen-presenting cells, which can elicit acute respiratory distress syndrome (ARDS) characterized by the rapid onset of widespread inflammation, the so-called cytokine storm. In response to viral infections, monocytes are recruited into the lung and subsequently differentiate into dendritic cells (DCs). DCs are critical players in the development of acute lung inflammation that causes ARDS. Here, we focus on the interaction of a specific SARS-CoV-2 open reading frame protein, ORF8, with DCs. We show that ORF8 binds to DCs, causes pre-maturation of differentiating DCs, and induces the secretion of multiple proinflammatory cytokines by these cells. In addition, we identified DC-SIGN as a possible interaction partner of ORF8 on DCs. Blockade of ORF8 leads to reduced production of IL-1β, IL-6, IL-12p70, TNF-α, MCP-1 (also named CCL2), and IL-10 by DCs. Therefore, a neutralizing antibody blocking the ORF8-mediated cytokine and chemokine response could be an improved therapeutic strategy against SARS-CoV-2.
Collapse
Affiliation(s)
- Matthias Hamdorf
- Cornea Lab Experimental Ophthalmology, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Thomas Imhof
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
- Institute for Experimental Dentistry and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Ben Bailey-Elkin
- Department of Microbiology, University of Manitoba, Winnipeg MB R3B 2E9 Manitoba, Canada
| | - Janina Betz
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
- Institute for Experimental Dentistry and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Sebastian J Theobald
- Department I of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Alexander Simonis
- Department I of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Veronica Di Cristanziano
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, 50935 Cologne, Germany
| | - Lutz Gieselmann
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, 50935 Cologne, Germany
| | - Felix Dewald
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, 50935 Cologne, Germany
| | - Clara Lehmann
- Department I of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Max Augustin
- Department I of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Florian Klein
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, 50935 Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Miguel A Alejandre Alcazar
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Department of Children and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster Stress Responses in Aging-associated Diseases, 50931 Cologne, Germany
- Institute for Lung Health (ILH), Universities of Gießen and Marburg Lung Centre, Member of the German Center for Lung Research, 35392 Gießen, Germany
| | - Robert Rongisch
- Dermatology, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Mario Fabri
- Dermatology, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Jan Rybniker
- Department I of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Heike Goebel
- Institute of Pathology, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Jörg Stetefeld
- Department of Microbiology, University of Manitoba, Winnipeg MB R3B 2E9 Manitoba, Canada
| | - Bent Brachvogel
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Claus Cursiefen
- Cornea Lab Experimental Ophthalmology, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Manuel Koch
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
- Institute for Experimental Dentistry and Oral Musculoskeletal Biology, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
| | - Felix Bock
- Cornea Lab Experimental Ophthalmology, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
49
|
Islam F, Habib S, Badruddza K, Rahman M, Islam MR, Sultana S, Nessa A. The Association of Cytokines IL-2, IL-6, TNF-α, IFN-γ, and IL-10 With the Disease Severity of COVID-19: A Study From Bangladesh. Cureus 2024; 16:e57610. [PMID: 38707035 PMCID: PMC11069400 DOI: 10.7759/cureus.57610] [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: 04/04/2024] [Indexed: 05/07/2024] Open
Abstract
Introduction Clinically, the early prediction of the severity of COVID-19 is often challenging, as a dramatic change in severity can occur without warning. The severity of COVID-19 disease is associated with an increased level of inflammatory mediators, including cytokines. This study aimed to evaluate the association of the levels of cytokines interleukin (IL)-2, IL-6, tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), and IL-10 with the severity of COVID-19 in Bangladesh. Materials and methods This cross-sectional study included a total of 60 confirmed cases of COVID-19, comprising 30 severe cases (Group A) and 30 non-severe cases (Group B), and 10 healthy individuals (Group C) attending Bangabandhu Sheikh Mujib Medical University (BSMMU) from March 2021 to February 2022. The cytokine assay was performed using the human Th1/Th2/Th17 cytokine kit BD cytometric bead array (CBA) on the BD Accuri C6 Plus flow cytometer. Statistical analysis was conducted using IBM SPSS Statistics for Windows, Version 23 (Released 2015; IBM Corp., Armonk, New York). Results The mean ages of the patients in Groups A, B, and C were 60.73±5.97, 57.13±7.68, and 48.10±9.13 years, respectively, with a male predominance in all groups. The mean IL-2, IL-6, IL-10, and TNF-α levels had a positive correlation with the increased age group and male gender, although it was not statistically significant. The mean IL-6 and IFN-γ levels were significantly higher among severe cases (216.95±147.78 and 0.98±0.95 pg/mL, respectively) compared to non-severe cases (94.29±128.79 and 0.41±0.61 pg/mL, respectively) and healthy individuals (1.08±1.97 and 0.15±0.28 pg/mL, respectively). Furthermore, the anti-inflammatory cytokine IL-10 was also significantly higher among severe cases (17.92±21.87 pg/mL) compared to non-severe cases (5.38±6.73 pg/mL) and healthy individuals (1.62±1.65 pg/mL). Conclusion IL-6, IFN-γ, and IL-10 have a significant association with the severity of COVID-19 disease. Clinicians treating patients with COVID-19 can consider the level of these cytokines as biomarkers of severity.
Collapse
Affiliation(s)
- Farzana Islam
- Division of Laboratory Service, Sheikh Hasina National Institute of Burn and Plastic Surgery, Dhaka, BGD
| | - Shahriar Habib
- Department of Microbiology, Sher-E-Bangla Medical College, Barishal, BGD
| | - Khaja Badruddza
- Department of Biochemistry, Sher-E-Bangla Medical College, Barishal, BGD
| | - Mahatabur Rahman
- Department of Gastroenterology, Sheikh Russel National Gastro-Liver Institute, Dhaka, BGD
| | - Mohammad R Islam
- Department of Anesthesia, Mainamoti Medical College and Hospital, Cumilla, BGD
| | - Sharmin Sultana
- Department of Virology, Bangabandhu Sheikh Mujib Medical University, Dhaka, BGD
| | - Afzalun Nessa
- Department of Virology, Bangabandhu Sheikh Mujjib Medical University, Dhaka, BGD
| |
Collapse
|
50
|
Akhmaltdinova L, Mekhantseva I, Turgunova L, Kostinov M, Zhumadilova Z, Turmukhambetova A. Association of soluble PD-L1 and NLR combination with 1-Year mortality in patients with COVID-19. Int Immunopharmacol 2024; 129:111600. [PMID: 38325048 DOI: 10.1016/j.intimp.2024.111600] [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: 12/12/2023] [Revised: 01/16/2024] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
PURPOSE Understanding the relationship between patient immune characteristics, disease severity, and mortality represents a critical step in the fight against COVID-19. Elevated levels of programmed death ligand-1 (PD-L1) and Neutrophil-lymphocyte ratio (NLR) are linked to increased severity of acute COVID-19 in patients. This study aimed to investigate the association of the combination of sPD-L1 and NLR with 1-year Mortality in patients with COVID-19. METHODS A prospective study was conducted involving patients with COVID-19 in Karaganda, Kazakhstan. The level of sPD-L1 in the blood serum was evaluated by ELISA. The effect of biomarkers on the development of mortality was analyzed with multivariate regression. RESULTS The risk of mortality within one year HR was 2.46 if the plasma sPD-L1 value of more than 277.13 pg/ml, and for NLR more than 2.46 HR was 2.87. The model of combining sPD-L1 and NLR resulted in an improvement in the predictive accuracy of the Hazard Ratio 7.6 (95 % CI: 3.02-19.11). CONCLUSION The combination of two immune-mediated markers (sPD-L1 and NLR), which reflect the systemic inflammatory balance of activation and exhaustion, can complement each other and improve the assessment of the risk of death in patients with COVID-19.
Collapse
Affiliation(s)
| | - Irina Mekhantseva
- Karaganda Medical University, Scientific and Research Center, Karaganda, Kazakhstan.
| | - Lyudmila Turgunova
- Karaganda Medical University, Scientific and Research Center, Karaganda, Kazakhstan.
| | - Mikhail Kostinov
- I.I. Mechnikov Research Institute of Vaccines and Sera, Sechenov First Moscow State Medical University, Department of Epidemiology and Modern Vaccination Technologies, Moscow, Russia.
| | - Zhibek Zhumadilova
- Karaganda Medical University, Scientific and Research Center, Karaganda, Kazakhstan.
| | - Anar Turmukhambetova
- Karaganda Medical University, Scientific and Research Center, Karaganda, Kazakhstan.
| |
Collapse
|