1
|
Nawa H, Murakami M. Neurobiology of COVID-19-Associated Psychosis/Schizophrenia: Implication of Epidermal Growth Factor Receptor Signaling. Neuropsychopharmacol Rep 2025; 45:e12520. [PMID: 39754403 PMCID: PMC11702486 DOI: 10.1002/npr2.12520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/06/2025] Open
Abstract
COVID-19 exhibits not only respiratory symptoms but also neurological/psychiatric symptoms rarely including delirium/psychosis. Pathological studies on COVID-19 provide evidence that the cytokine storm, in particular (epidermal growth factor) EGF receptor (EGFR, ErbB1, Her1) activation, plays a central role in the progression of viral replication and lung fibrosis. Of note, SARS-CoV-2 virus (specifically, S1 spike domain) mimics EGF and directly transactivates EGFR, preceding the inflammatory process. In agreement, the anticancer drugs targeting EGFR such as Nimotuzumab and tyrosine kinase inhibitors are markedly effective on COVID-19. However, these data might raise a provisional caution regarding implication of psychiatric disorder such as schizophrenia. The author's group has been investigating the etiologic and neuropathologic associations of EGFR signaling with schizophrenia. There are significant molecular associations between schizophrenia and EGFR ligand levels in blood as well as in the brain. In addition, perinatal challenges of EGFR ligands and intraventricular administration of EGF to rodents and monkeys both resulted in severe behavioral and/or electroencephalographic endophenotypes relevant to this disorder. These animal models also display postpubertal abnormality in soliloquy-like self-vocalization as well as in intercortical functional connectivity. Here, we discuss neuropsychiatric implication of coronavirus infection and its interaction with the EGFR system, by searching related literatures in PubMed database as of the end of 2023.
Collapse
Affiliation(s)
- Hiroyuki Nawa
- Department of Physiological Sciences, School of Pharmaceutical SciencesWakayama Medical UniversityWakayamaJapan
| | - Masaaki Murakami
- Molecular Psychoneuroimmunology, Institute for Genetic MedicineHokkaido UniversitySapporoHokkaidoJapan
| |
Collapse
|
2
|
Costa MP, Abdu JOC, Machado Resende Guedes MC, Sarcinelli MA, Fabri RL, Pittella F, Macedo GC, Vilela FMP, Rocha HVA, Tavares GD. Dexamethasone-loaded chitosan-decorated PLGA nanoparticles: A step forward in attenuating the COVID-19 cytokine storm? Colloids Surf B Biointerfaces 2025; 246:114359. [PMID: 39522287 DOI: 10.1016/j.colsurfb.2024.114359] [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: 07/18/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
This study aims to develop and characterize poly (lactic-co-glycolic acid) (PLGA) nanoparticles decorated with chitosan (CS) for the encapsulation of dexamethasone (DEX) (NP-DEX-CS), targeting improved efficacy in the treatment of severe acute respiratory syndrome (SARS) associated with COVID-19. The nanoparticles were systematically characterized for size, zeta potential (ZP), morphology, encapsulation efficiency, and in vitro drug release. Incorporation of CS resulted in significant modifications in the nanoparticles' physical properties, notably an increase in size (from 207.3 ± 6.7 nm to 264.4 ± 4.4 nm) and a shift in ZP to positive values (from -11.8 ±1.4 mV to +30.0 ± 1,6 mV). The NP-DEX-CS formulation achieved a high encapsulation efficiency (∼79 %) and a drug loading capacity of 6.53 ± 0.02 %.In addition, the in vitro release rate of DEX from NP-DEX-CS was lower compared to undecorated nanoparticles, with a reduction from approximately 64-37 % within 24 h. Microscopy analyses revealed a smoother surface on the CS-decorated nanoparticles. FTIR and XRD analyses confirmed successful chitosan coating and DEX encapsulation. The CS coating enhanced the tolerability of J774.A1 cells to the nanoparticles, particularly evident at the highest concentration (400ug/mL), resulting in a cell viability ≥70 %. Importantly, the NP-DEX-CS significantly reduced levels of nitric oxide and inflammatory cytokines (IL-1, IL-6, IL-12, and TNF-α). These findings suggest that CS-decorated PLGA nanoparticles hold promise as an effective dexamethasone delivery system for treating SARS related to COVID-19.
Collapse
Affiliation(s)
- Mirsiane Pascoal Costa
- Postgraduate Program in Pharmaceutical Science, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | | | - Maria Clara Machado Resende Guedes
- Postgraduate Program in Biological Sciences, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | | | - Rodrigo Luiz Fabri
- Postgraduate Program in Pharmaceutical Science, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | - Frederico Pittella
- Postgraduate Program in Pharmaceutical Science, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | - Gilson Costa Macedo
- Postgraduate Program in Biological Sciences, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | - Fernanda Maria Pinto Vilela
- Postgraduate Program in Pharmaceutical Science, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| | | | - Guilherme Diniz Tavares
- Postgraduate Program in Pharmaceutical Science, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil; Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
| |
Collapse
|
3
|
Galipeau Y, Cooper C, Langlois MA. Autoantibodies in COVID-19: implications for disease severity and clinical outcomes. Front Immunol 2025; 15:1509289. [PMID: 39835117 PMCID: PMC11743527 DOI: 10.3389/fimmu.2024.1509289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Few pathogens have historically been subjected to as intense scientific and clinical scrutiny as SARS-CoV-2. The genetic, immunological, and environmental factors influencing disease severity and post-infection clinical outcomes, known as correlates of immunity, remain largely undefined. Clinical outcomes of SARS-CoV-2 infection vary widely, ranging from asymptomatic cases to those with life-threatening COVID-19 symptoms. While most infected individuals return to their former health and fitness within a few weeks, some develop debilitating chronic symptoms, referred to as long-COVID. Autoimmune responses have been proposed as one of the factors influencing long-COVID and the severity of SARS-CoV-2 infection. The association between viral infections and autoimmune pathologies is not new. Viruses such as Epstein-Barr virus and cytomegalovirus, among others, have been shown to induce the production of autoantibodies and the onset of autoimmune conditions. Given the extensive literature on SARS-CoV-2, here we review current evidence on SARS-CoV-2-induced autoimmune pathologies, with a focus on autoantibodies. We closely examine mechanisms driving autoantibody production, particularly their connection with disease severity and long-COVID.
Collapse
Affiliation(s)
- Yannick Galipeau
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Curtis Cooper
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation (CI3), University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
4
|
Vakani K, Norbury R, Vanova M, Ratto M, Parton A, Antonova E, Kumari V. Cognitive function and brain structure in COVID-19 survivors: The role of persistent symptoms. Behav Brain Res 2025; 476:115283. [PMID: 39368712 DOI: 10.1016/j.bbr.2024.115283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
Persistent COVID-19 symptoms post-acute state have been shown to have a significant negative impact on brain structure and function. In this study, we conducted magnetic resonance imaging (MRI) of the whole brain in 43 working-age adults (mean age: 44.79±10.80; range: 24-65 years) with a history of COVID-19 (731.17±312.41 days post-diagnosis), and also assessed their cognitive function (processing speed, attention, working memory, executive function, and recognition memory), mental health, and sleep quality. MRI data were processed using FSL to derive regional volumes for bilateral nucleus accumbens, caudate, pallidum, putamen, thalamus, amygdala, and hippocampus, and total grey matter, white matter, and cerebral spinal fluid volume, and analysed in relation to persistent COVID-19 symptom load, mental health, and sleep quality. Higher persistent COVID-19 symptom load was significantly associated with smaller putamen volume, lower response accuracy on working memory, executive function, and recognition memory tasks, as well as a longer time to complete the executive function task, and poorer mental health and sleep quality. Smaller putamen fully mediated the relationship between persistent COVID-19 symptom load and lower executive function. Further research is required to confirm whether reduced putamen volume and its association with poor executive function persists in COVID-19 survivors in the long term.
Collapse
Affiliation(s)
- Krupa Vakani
- Division of Psychology, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom; Centre for Cognitive and Clinical Neuroscience, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom.
| | - Ray Norbury
- Division of Psychology, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom; Centre for Cognitive and Clinical Neuroscience, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Martina Vanova
- Royal Holloway, University of London, London, United Kingdom
| | | | - Andrew Parton
- Division of Psychology, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom; Centre for Cognitive and Clinical Neuroscience, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Elena Antonova
- Division of Psychology, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom; Centre for Cognitive and Clinical Neuroscience, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Veena Kumari
- Division of Psychology, Department of Life Sciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom; Centre for Cognitive and Clinical Neuroscience, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom.
| |
Collapse
|
5
|
Fonseca TAH, Von Rekowski CP, Araújo R, Oliveira MC, Justino GC, Bento L, Calado CRC. Comparison of two metabolomics-platforms to discover biomarkers in critically ill patients from serum analysis. Comput Biol Med 2025; 184:109393. [PMID: 39549530 DOI: 10.1016/j.compbiomed.2024.109393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/08/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024]
Abstract
Serum metabolome analysis is essential for identifying disease biomarkers and predicting patient outcomes in precision medicine. Thus, this study aims to compare Ultra-High Performance Liquid Chromatography-High-Resolution Mass Spectrometry (UHPLC-HRMS) with Fourier Transform Infrared (FTIR) spectroscopy in acquiring the serum metabolome of critically ill patients, associated with invasive mechanical ventilation (IMV), and predicting death. Three groups of 8 patients were considered. Group A did not require IMV and survived hospitalization, while Groups B and C required IMV. Group C patients died a median of 5 days after sample harvest. Good prediction models were achieved when comparing groups A to B and B to C using both platforms' data, with UHPLC-HRMS showing 8-17 % higher accuracies (≥83 %). However, developing predictive models using metabolite sets was not feasible when comparing unbalanced populations, i.e., Groups A and B combined to Group C. Alternatively, FTIR-spectroscopy enabled the development of a model with 83 % accuracy. Overall, UHPLC-HRMS data yields more robust prediction models when comparing homogenous populations, potentially enhancing understanding of metabolic mechanisms and improving patient therapy adjustments. FTIR-spectroscopy is more suitable for unbalanced populations. Its simplicity, speed, cost-effectiveness, and high-throughput operation make it ideal for large-scale studies and clinical translation in complex populations.
Collapse
Affiliation(s)
- Tiago A H Fonseca
- NMS - NOVA Medical School, FCM - Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Dos Mártires da Pátria 130, 1169-056, Lisbon, Portugal; ISEL - Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Rua Conselheiro Emídio Navarro 1, 1959-007, Lisbon, Portugal; CHRC - Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082, Lisbon, Portugal.
| | - Cristiana P Von Rekowski
- NMS - NOVA Medical School, FCM - Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Dos Mártires da Pátria 130, 1169-056, Lisbon, Portugal; ISEL - Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Rua Conselheiro Emídio Navarro 1, 1959-007, Lisbon, Portugal; CHRC - Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082, Lisbon, Portugal.
| | - Rúben Araújo
- NMS - NOVA Medical School, FCM - Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Dos Mártires da Pátria 130, 1169-056, Lisbon, Portugal; ISEL - Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Rua Conselheiro Emídio Navarro 1, 1959-007, Lisbon, Portugal; CHRC - Comprehensive Health Research Centre, Universidade NOVA de Lisboa, 1150-082, Lisbon, Portugal.
| | - M Conceição Oliveira
- Centro de Química Estrutural - Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001, Lisbon, Portugal.
| | - Gonçalo C Justino
- Centro de Química Estrutural - Institute of Molecular Sciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001, Lisbon, Portugal.
| | - Luís Bento
- Intensive Care Department, ULSSJ - Unidade Local de Saúde de São José, Rua José António Serrano, 1150-199, Lisbon, Portugal; Integrated Pathophysiological Mechanisms, CHRC - Comprehensive Health Research Centre, NMS - NOVA Medical School, FCM - Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria, 1169-056, Lisbon, Portugal.
| | - Cecília R C Calado
- ISEL - Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, Rua Conselheiro Emídio Navarro 1, 1959-007, Lisbon, Portugal; IBB-Institute for Bioengineering and Biosciences, The Associate Laboratory Institute for Health and Bioeconomy (i4HB), Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisbon, Portugal.
| |
Collapse
|
6
|
Adilović M, Hromić-Jahjefendić A, Mahmutović L, Šutković J, Rubio-Casillas A, Redwan EM, Uversky VN. Intrinsic Factors Behind the Long-COVID: V. Immunometabolic Disorders. J Cell Biochem 2025; 126:e30683. [PMID: 39639607 DOI: 10.1002/jcb.30683] [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: 07/07/2024] [Revised: 11/02/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
The complex link between COVID-19 and immunometabolic diseases demonstrates the important interaction between metabolic dysfunction and immunological response during viral infections. Severe COVID-19, defined by a hyperinflammatory state, is greatly impacted by underlying chronic illnesses aggravating the cytokine storm caused by increased levels of Pro-inflammatory cytokines. Metabolic reprogramming, including increased glycolysis and altered mitochondrial function, promotes viral replication and stimulates inflammatory cytokine production, contributing to illness severity. Mitochondrial metabolism abnormalities, strongly linked to various systemic illnesses, worsen metabolic dysfunction during and after the pandemic, increasing cardiovascular consequences. Long COVID-19, defined by chronic inflammation and immune dysregulation, poses continuous problems, highlighting the need for comprehensive therapy solutions that address both immunological and metabolic aspects. Understanding these relationships shows promise for effectively managing COVID-19 and its long-term repercussions, which is the focus of this review paper.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Lejla Mahmutović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Jasmin Šutković
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan, Mexico
- Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Mexico
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria, Egypt
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
7
|
Lin MW, Lin CH, Chang JR, Chiang HH, Wu TH, Lin CS. The influence of PM2.5 exposure on SARS-CoV-2 infection via modulating the expression of angiotensin converting enzyme II. JOURNAL OF HAZARDOUS MATERIALS 2024; 485:136887. [PMID: 39700942 DOI: 10.1016/j.jhazmat.2024.136887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
Particulate matter 2.5 (PM2.5) pollution and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic are the greatest environmental health issues worldwide. Several statistics revealed the significant positive correlation between the morbidity of coronavirus disease-19 (COVID-19) and the levels of air pollution. Nevertheless, there is no direct experimental evidence to indicate the effect of PM2.5 exposure on SARS-CoV-2 infection. The objective of this study was to evaluate whether the infection of SARS-CoV-2 affected by PM2.5 through angiotensin-converting enzyme II (ACE2) expression enhances and investigate the function of ACE2 in lung injury induced by PM2.5. An animal model of PM2.5-induced lung injury was established using wild-type (WT, C57BL/6), human ACE2 transgenic (K18-hACE2 TG), and murine ACE2 gene knockout (mACE2 KO) mice. The results indicate that PM2.5 exposure facilitates SARS-CoV-2 infection through inducing ACE2 expression in vitro (10 μg/mL) and in vivo (6.25 mg/kg/day in 50 μL saline). The levels of ACE, inflammatory cytokines, and mitogen-activated protein kinase (MAPK) proteins in WT, K18-hACE TG and mACE2 KO mice were significantly increased after PM2.5 instillation. The severest PM2.5-induced lung damage was observed in mACE2 KO mice. In summary, ACE2 plays a double-edged sword role in lung injury, PM2.5 exposure contributed to SARS-CoV-2 infection through inducing ACE2 expression, but ACE2 also protected pulmonary inflammation from PM2.5 challenge.
Collapse
Affiliation(s)
- Meng-Wei Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Cheng-Han Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Jia-Rong Chang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Hua-Hsin Chiang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Ting-Hsuan Wu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| | - Chih-Sheng Lin
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 30068, Taiwan.
| |
Collapse
|
8
|
Joveini S, Yarmohammadi F, Iranshahi M, Nikpoor AR, Askari VR, Attaranzadeh A, Etemad L, Taherzadeh Z. Distinct therapeutic effects of auraptene and umbelliprenin on TNF-α and IL-17 levels in a murine model of chronic inflammation. Heliyon 2024; 10:e40731. [PMID: 39687160 PMCID: PMC11648749 DOI: 10.1016/j.heliyon.2024.e40731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Objective To compare the anti-arthritic potential of orally administered auraptene (AUR) and umbellliprenin (UMB) in chronic inflammation by exploring the differential effect on regulating TNF-α and IL-17. Methods & materials Sixty male rats were divided into ten groups, and after confirming chronic inflammation, the treatment groups received AUR or UMB orally for 9 days. On day 16, histopathological changes were evaluated. Altered serum levels of the inflammatory cytokines TNF-α and IL-17 were examined as the underlying mechanisms. Results Administering AUR orally at 16 mM/kg caused a significant increase in body weight gain compared to the baseline (p < 0.05), while UMB at a dose of 64 mM/kg significantly reduced edema size (p < 0.01). TNF-α levels were significantly lower in all doses of AUR and UMB treatments compared to the arthritis control group (p < 0.05). Treatment with AUR at all relative doses resulted in a significant decrease in IL-17 levels compared to the arthritis control group (p < 0.05), whereas UMB treatment did not show a significant effect on IL-17 levels. Conclusion AUR and UMB regulate TNF-α and IL-17 differently; AUR inhibits both, showing broad therapeutic potential, while UMB specifically targets TNF-α, showing a specialized role.
Collapse
Affiliation(s)
- Saeid Joveini
- Department of Pharmacodynamy and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Yarmohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehrdad Iranshahi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amin Reza Nikpoor
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Armin Attaranzadeh
- Department of Medical Genetics, Faculty of Medicines, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Etemad
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zhila Taherzadeh
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Targeted Drug Delivery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Thakur N, Chakraborty P, Tufariello JM, Basler CF. SARS-CoV-2 Nsp14 binds Tollip and activates pro-inflammatory pathways while downregulating interferon-α and interferon-γ receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.628214. [PMID: 39713296 PMCID: PMC11661139 DOI: 10.1101/2024.12.12.628214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
SARS coronavirus 2 (SARS-CoV-2) non-structural protein 14 (Nsp14) possesses an N-terminal exonuclease (ExoN) domain that provides a proofreading function for the viral RNA-dependent RNA polymerase and a C-terminal N7-methyltransferase (N7-MTase) domain that methylates viral mRNA caps. Nsp14 also modulates host functions. This includes the activation of NF-κB and downregulation of interferon alpha/beta receptor 1 (IFNAR1). Here we demonstrate that Nsp14 exerts broader effects, activating not only NF-κB responses but also ERK, p38 and JNK MAP kinase (MAPK) signaling, promoting cytokine production. Further, Nsp14 downregulates not only IFNAR1 but also IFN-γ receptor 1 (IFNGR1), impairing cellular responses to both IFNα and IFNγ. IFNAR1 and IFNGR1 downregulation is via a lysosomal pathway and also occurs in SARS-CoV-2 infected cells. Analysis of a panel of Nsp14 mutants reveals a consistent pattern. Mutants that disable ExoN function remain active, whereas N7-MTase mutations impair both pro-inflammatory pathway activation and IFN receptor downregulation. Innate immune modulating functions also require the presence of both the ExoN and N7-MTase domains likely reflecting the need for the ExoN domain for N7-MTase activity. We further identify multi-functional host protein Tollip as an Nsp14 interactor. Interaction requires the phosphoinositide-binding C2 domain of Tollip and sequences C-terminal to the C2 domain. Full length Tollip or regions encompassing the Nsp14 interaction domain are sufficient to counteract both Nsp14-mediated and Nsp14-independent activation of NF-κB. Knockdown of Tollip partially reverses IFNAR1 and IFNGR1 downregulation in SARS-CoV-2 infected cells, suggesting relevance of Nsp14-Tollip interaction for Nsp14 innate immune evasion functions.
Collapse
Affiliation(s)
- Naveen Thakur
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Poushali Chakraborty
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - JoAnn M. Tufariello
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Christopher F. Basler
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
10
|
Ushigome E, Imai D, Hamaguchi M, Hashimoto S, Fukui M. Maximum insulin dose in patients admitted to the intensive care units with severe COVID-19 in the "Cross ICU Searchable Information System" study: A multicenter retrospective cohort study. J Diabetes Investig 2024. [PMID: 39658883 DOI: 10.1111/jdi.14380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 11/07/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
AIMS This study aimed to determine the maximum daily insulin dose (MDI) and associated factors in critically ill patients with coronavirus disease 2019 (COVID-19) receiving insulin therapy, under ventilator and/or extracorporeal membrane oxygenation (ECMO) management. MATERIALS AND METHODS This cross-sectional analysis used the Cross ICU Searchable Information System data from a Japanese multicenter retrospective observational cohort study of critically ill patients with COVID-19 receiving ventilation and/or ECMO, from February 2020 to March 2022. Maximum daily insulin dose was determined, and factors associated with it and maximum daily insulin dose per body weight were assessed using linear regression analysis. RESULTS The analysis included 788 patients. Their mean age, glycated hemoglobin level, maximum daily insulin dose, and time from admission to the maximum daily insulin dose were 65.2 ± 13.0 years, 7.0 ± 1.5% (53.0 ± 7.1 mmol/mol), 46.0 ± 43.6 U/day, and 7.3 ± 7.0 days, respectively. Male sex (β = 6.902, P = 0.034), body mass index (β = 1.020, P = 0.001), glycated hemoglobin (β = 12.272, P < 0.001), and having renal failure (β = 20.637, P = 0.003) were independent determinants of maximum daily insulin dose. Age (β = 0.004, P = 0.035), glycated hemoglobin (β = 0.154, P < 0.001), and having renal failure (β = 0.282, P = 0.004) were independent determinants of maximum daily insulin dose per body weight. CONCLUSIONS In patients with COVID-19 on ventilator and/or ECMO management, the maximum daily insulin dose reached after about 1 week of hospitalization was approximately 46.0 U/day. Glycated hemoglobin and renal failure were both associated with the maximum daily insulin dose and maximum daily insulin dose per body weight.
Collapse
Affiliation(s)
- Emi Ushigome
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Dan Imai
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
11
|
Pang Z, Tang A, He Y, Fan J, Yang Q, Tong Y, Fan H. Neurological complications caused by SARS-CoV-2. Clin Microbiol Rev 2024; 37:e0013124. [PMID: 39291997 PMCID: PMC11629622 DOI: 10.1128/cmr.00131-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
SUMMARYSARS-CoV-2 can not only cause respiratory symptoms but also lead to neurological complications. Research has shown that more than 30% of SARS-CoV-2 patients present neurologic symptoms during COVID-19 (A. Pezzini and A. Padovani, Nat Rev Neurol 16:636-644, 2020, https://doi.org/10.1038/s41582-020-0398-3). Increasing evidence suggests that SARS-CoV-2 can invade both the central nervous system (CNS) (M.S. Xydakis, M.W. Albers, E.H. Holbrook, et al. Lancet Neurol 20: 753-761, 2021 https://doi.org/10.1016/S1474-4422(21)00182-4 ) and the peripheral nervous system (PNS) (M.N. Soares, M. Eggelbusch, E. Naddaf, et al. J Cachexia Sarcopenia Muscle 13:11-22, 2022, https://doi.org/10.1002/jcsm.12896), resulting in a variety of neurological disorders. This review summarized the CNS complications caused by SARS-CoV-2 infection, including encephalopathy, neurodegenerative diseases, and delirium. Additionally, some PNS disorders such as skeletal muscle damage and inflammation, anosmia, smell or taste impairment, myasthenia gravis, Guillain-Barré syndrome, ICU-acquired weakness, and post-acute sequelae of COVID-19 were described. Furthermore, the mechanisms underlying SARS-CoV-2-induced neurological disorders were also discussed, including entering the brain through retrograde neuronal or hematogenous routes, disrupting the normal function of the CNS through cytokine storms, inducing cerebral ischemia or hypoxia, thus leading to neurological complications. Moreover, an overview of long-COVID-19 symptoms is provided, along with some recommendations for care and therapeutic approaches of COVID-19 patients experiencing neurological complications.
Collapse
Affiliation(s)
- Zehan Pang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Ao Tang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yujie He
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Junfen Fan
- Department of Neurology, Institute of Cerebrovascular Diseases Research, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Qingmao Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Huahao Fan
- School of Life Sciences, Tianjin University, Tianjin, China
| |
Collapse
|
12
|
Smit K, van Uum RT, Rijks S, van de Pol AC, Ahmad A, Venekamp RP, Rutten FH, Zwart DLM. Acute remote home monitoring of acutely ill patients with COVID-19: how Dutch home monitoring initiatives were organized during the pandemic. BMC Health Serv Res 2024; 24:1521. [PMID: 39623361 PMCID: PMC11610280 DOI: 10.1186/s12913-024-11910-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 11/08/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Acute remote home monitoring of acutely ill patients with COVID-19 holds potential for early detection of deterioration and thus subsequentearly intervention that may prevent or mitigate progression to severe illness and need for respiratory support. Our aim was to describe common features of acute remote home monitoring programs for acutely ill patients with COVID-19 in the Netherlands. METHODS We performed literature searches (both grey and academic) between 1st March 2020 and 1st March 2023 to identify Dutch acute remote home monitoring initiatives, excluding studies on early hospital discharge. From the available protocols, we extracted relevant information on patient eligibility, organization of acute remote home monitoring and home management. RESULTS We identified and approached ten acute remote home monitoring initiatives for information regarding their used protocols. Seven out of ten protocols were retrieved and assessed. All initiatives focused on adult patients with COVID-19 who where at risk of developing severe COVID-19, and all initiatives provided close follow-up through remote home monitoring using medically certified pulse oximeters. Daily measurements included peripheral oxygen saturation (all initiatives, n = 7), body temperature (n = 6), heart frequency per minute (n = 4) and breathing rate per minute (n = 4). For follow-up and review of measured values, in most initiatives (n = 6) the physician (general practitioner or hospital physician) in charge was supported by a dedicated monitoring center. In 5 out of 7 initiatives, the general practitioner (GP) was responsible for supervising the patients and monitoring staff. CONCLUSION The acute remote home monitoring initiatives that emerged in the Netherlands during the first wave of the COVID-19 pandemic were similarly organized. Common building blocks for home monitoring include daily check of peripheral oxygen saturation, monitoring through a dedicated remote monitoring center alongside healthcare personnel and a supervising physician.
Collapse
Affiliation(s)
- Karin Smit
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| | - Rick T van Uum
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Stella Rijks
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Alma C van de Pol
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Abeer Ahmad
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of General Practice, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Roderick P Venekamp
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Dorien L M Zwart
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
13
|
Bahrami S, Babaei N, Esmaeili Gouvarchin Ghaleh H, Mohajeri Borazjani J, Farzanehpour M. Anti-inflammatory activity of Ziziphus jujuba hydroalcoholic extract in acetic acid-induced ulcerative colitis model. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2024; 21:481-489. [PMID: 39485260 DOI: 10.1515/jcim-2024-0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/23/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVES Ulcerative colitis (UC) is a common gastrointestinal (GI) disorder characterized by chronic inflammation. Current treatments primarily focus on symptom management, but they have inherent limitations. Global attention is increasingly directed towards exploring herbal remedies as complementary approaches. This study aims to investigate the effects of the hydroalcoholic extract of jujuba on an experimental model of ulcerative colitis. METHODS In this study, 15 male BALB/c mice were divided into three experimental groups. The first group served as the untreated UC model, acting as the positive control (PC). The second group received treatment with the hydroalcoholic extract of Ziziphus jujuba, while the third group was treated with mesalamine. UC was induced by injecting 100 μL of 4 % acetic acid (AA) intra-rectally several times. Treatment commenced after the onset of symptoms such as diarrhea and bloody stools. The mice were eventually euthanized ethically, and their spleen and intestinal tissues were collected for analysis. Evaluations included the Disease Activity Index (DAI), myeloperoxidase activity (MPO), nitric oxide (NO) levels, cytokine levels (IL-1β, IL-6, TNF-α), and gene expression (iNOS, COX-2, and cytokines). RESULTS The hydroalcoholic extract of the jujuba plant significantly reduced MPO, NO, the DAI, and the production and expression of inflammatory cytokines, as well as the genes iNOS and COX-2, in the group receiving this extract compared to the positive control group (p<0.05). CONCLUSIONS The study demonstrates that the hydroalcoholic extract of Ziziphus jujuba significantly reduces inflammation markers such as TNF-α, NO, MPO, IL-1β, and IL-6 in a mouse model of ulcerative colitis. Additionally, it downregulates the expression of pro-inflammatory genes, including iNOS and COX-2. These findings suggest that Z. jujuba extract has potential as an effective anti-inflammatory treatment for managing ulcerative colitis symptoms.
Collapse
Affiliation(s)
- Shabnam Bahrami
- Department of Molecular Cell Biology and Genetics, Bushehr Branch, Islamic Azad University, Bushehr, Iran
| | - Nahid Babaei
- Department of Molecular Cell Biology and Genetics, Bushehr Branch, Islamic Azad University, Bushehr, Iran
| | - Hadi Esmaeili Gouvarchin Ghaleh
- Department of Immunology, 48417 Applied Virology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences , Tehran, Iran
| | - Jaleh Mohajeri Borazjani
- Department of Fisheries and Natural Resources, Bushehr Branch, Islamic Azad University, Bushehr, Iran
| | - Mahdieh Farzanehpour
- Department of Immunology, 48417 Applied Virology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences , Tehran, Iran
| |
Collapse
|
14
|
Li Y, Qin S, Dong L, Xiao Y, Zhang Y, Hou Y, Qiao S, Zhang R, Li Y, Bao Y, Zhao X, Ma Y, Gao GF. Multi-omic characteristics of longitudinal immune profiling after breakthrough infections caused by Omicron BA.5 sublineages. EBioMedicine 2024; 110:105428. [PMID: 39536392 PMCID: PMC11605469 DOI: 10.1016/j.ebiom.2024.105428] [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: 06/13/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Omicron sub-variants breakthrough infections (BTIs) have led to millions of coronavirus disease 2019 (COVID-19) cases worldwide. The acute-phase immune status is critical for prognosis, however, the dynamic immune profiling of COVID-19 during the first month after BTIs remains unclear. METHODS In this study, we monitored the immune dynamics at various timepoints in a longitudinal cohort during the first month post-BTIs through clinical evaluation, single-cell RNA sequencing (scRNA-seq), T cell receptor (TCR)/B cell receptor (BCR) sequencing, and antibody mass spectrometry. FINDINGS Serological analysis revealed limited impairment to functions of major organs, active cellular and humoral immunity at 2 weeks post-BTI, with significant increases in cytokines (CKs) and neutralizing antibody levels. However, 1 month post-BTI, organ function parameters and CK levels reverted to pre-infection levels, whereas neutralizing antibody levels remained high. Notably, scRNA-seq showed that lymphocytes maintained strong antiviral activity and cell depletion at 2 weeks and 1 month post-BTI, with genes CD81, ABHD17A, CXCR4, DUSP1, etc. upregulated, and genes PFDN5, DYNLRB1, CD52, etc. downregulated, indicating that lymphocytes status take longer to recover to normal levels than that routine blood tests revealed. Additionally, T cell-exhaustion associated genes, including LAG3, TIGIT, PDCD1, CTLA4, HAVCR2, and TOX, were upregulated after BTI. TCRs and BCRs exhibited higher clonotypes, mainly in CD8Tem or plasmablast cells, at 2 weeks post-BTI comparing 1 month. More IgG and IgA-type BCRs were found in the groups of 1 month post-BTI, with higher somatic hypermutation, indicating greater maturity. Verification of monoclonal antibodies corresponding to amplified BCRs highlighted the antigen-specific and broad-spectrum characteristics. INTERPRETATION Our study elucidated the dynamic immune profiling of individuals after Omicron BA.5 sublineages BTI. Strong immune activation, antiviral response, antibody maturation and class transition at 2 weeks and 1 month after BTI may provide essential insights into pathogenicity, sequential immune status, recovery mechanisms of Omicron sublineage BTI. FUNDING This study was supported by the National Key R&D Program of China, the China Postdoctoral Science Foundation, Guangdong Basic and Applied Basic Research Foundation, the National Natural Science Foundation of China, CAS Project for Young Scientists in Basic Research, and the Air Force Special Medical Center Science and Technology Booster Program.
Collapse
MESH Headings
- Humans
- COVID-19/immunology
- COVID-19/virology
- SARS-CoV-2/immunology
- Male
- Female
- Middle Aged
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Longitudinal Studies
- Adult
- Cytokines/metabolism
- Aged
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/genetics
- Single-Cell Analysis
- Breakthrough Infections
- Multiomics
Collapse
Affiliation(s)
- Yanhua Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shijie Qin
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China; Shenzhen Children's Hospital, Shenzhen, 518026, China; The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Lei Dong
- Department of Clinical Laboratory, Air Force Medical Center, Beijing, 100142, China
| | - Yunfeng Xiao
- Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yanan Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China; Shenzhen Children's Hospital, Shenzhen, 518026, China
| | - Yali Hou
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shitong Qiao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Rong Zhang
- Guangxi University State Key Laboratory for Conservation and Utilization of Subtropical Agro BioResources, Nanning, 53000, China
| | - Ying Li
- Department of Clinical Laboratory, Air Force Medical Center, Beijing, 100142, China
| | - Yanmin Bao
- Shenzhen Children's Hospital, Shenzhen, 518026, China.
| | - Xin Zhao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China; Beijing Life Science Academy, Beijing, 102209, China.
| | - Yueyun Ma
- Department of Clinical Laboratory, Air Force Medical Center, Beijing, 100142, China.
| | - George Fu Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
15
|
Sabino‐Santos G, Leggio CE, Litwin SM, Waheed N, Bai S, Ulusan S, Karunathilake A, Elliott DH, Smira AR, Chandra S, Li L, Ning B, Hu T, Schieffelin JS, Gunn BM, Robinson JE, Fuloria J, Norton EB. Post-COVID immunity in patients with solid tumor or hematological malignancies treated with SARS-CoV-2 monoclonal antibodies. Immun Inflamm Dis 2024; 12:e70039. [PMID: 39659018 PMCID: PMC11632117 DOI: 10.1002/iid3.70039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/12/2024] [Accepted: 10/01/2024] [Indexed: 12/12/2024] Open
Abstract
PURPOSE SARS-CoV-2 monoclonal antibody (mAB) therapy has effectively treated severe COVID-19, although how this contributes to protective antiviral immunity in settings of malignancy is poorly defined. PATIENTS AND METHODS We evaluated the development of post-infection immunity in five patients with malignancies who received mAB therapy targeting spike protein for their PCR-confirmed SARS-CoV-2 infection in 2021, compared with non-mAB controls. Patients were identified from a larger study on oncology with a history or documented current infection with SARS-CoV-2. Subjects include two patients with lymphoma and CD20-depletion therapy, one with myeloma and two with solid tumor (stage IIA rectal adenocarcinoma and metastatic breast cancer). Cancer therapies and COVID vaccination history varied by patient. Blood samples (1-4 per patient) were collected 71-635 days post-mAB therapy. We employed clinical histories with comprehensive immunoprofiling analysis, including systems serology antibody isotyping and effector function, T-cell immunophenotyping for subset and memory cells, and sensitive blood viral RNA detection up to 2 years post-mAB therapy. RESULTS B-cell deficiency was confirmed in 3/5 patients. All patients had detectable anti-spike and nucleoprotein antibody isotypes, effector functions, and neutralizing antibodies (which increased over time by subject) at similar levels to the control group. Virus-specific T-cell activation and phenotypes varied by time and patient. Spike-specific effector and memory CD8 + T-cells were significantly elevated in mAB subjects compared to the control group. SARS-CoV-2 viral RNA detection was also higher in mAB-treated patients. One patient on bortezomib therapy had unique alterations in these populations. CONCLUSION All mAB-treated patients with malignancies developed polyfunctional immunity humoral and T-cell immunity to SARS-CoV-2 even in the setting of B-cell deficiency. The evolution of this immunity, including new variant-specific antibodies, without secondary illnesses suggests that patients were protected from symptomatic re-infection, and mAB therapy did not blunt the development of host immunity. Future studies are warranted to better characterize immunologic memory over time with exposures to new viral variants, evaluate prolonged viral shedding and the continued use of appropriate mAB for infection in high-risk patients.
Collapse
Affiliation(s)
- Gilberto Sabino‐Santos
- Department of Microbiology and ImmunologyTulane University School of MedicineNew OrleansLouisianaUSA
| | | | - Sean M. Litwin
- Department of Microbiology and ImmunologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Najia Waheed
- University Medical Center New OrleansNew OrleansLouisianaUSA
| | - Shuangyi Bai
- Paul G. Allen School of Global HealthWashington State UniversityPullmanWashingtonUSA
| | - Sinem Ulusan
- Paul G. Allen School of Global HealthWashington State UniversityPullmanWashingtonUSA
| | - Anoli Karunathilake
- Paul G. Allen School of Global HealthWashington State UniversityPullmanWashingtonUSA
| | - Debra H. Elliott
- Department of PediatricsTulane University School of MedicineNew OrleansLouisianaUSA
| | - Ashley R. Smira
- Department of PediatricsTulane University School of MedicineNew OrleansLouisianaUSA
| | - Sruti Chandra
- Department of Microbiology and ImmunologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Lin Li
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Bo Ning
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - Tony Hu
- Department of Biochemistry and Molecular BiologyTulane University School of MedicineNew OrleansLouisianaUSA
| | - John S. Schieffelin
- Department of PediatricsTulane University School of MedicineNew OrleansLouisianaUSA
| | - Bronwyn M. Gunn
- Paul G. Allen School of Global HealthWashington State UniversityPullmanWashingtonUSA
| | - James E. Robinson
- Department of PediatricsTulane University School of MedicineNew OrleansLouisianaUSA
| | - Jyotsna Fuloria
- University Medical Center New OrleansNew OrleansLouisianaUSA
| | - Elizabeth B. Norton
- Department of Microbiology and ImmunologyTulane University School of MedicineNew OrleansLouisianaUSA
| |
Collapse
|
16
|
Bi Q, Zhu J, Zheng J, Xu Q, Chen J, Zhang L, Mu X. Blood Inflammatory Markers and Cytokines in COVID-19 Patients With Bacterial Coinfections. Immun Inflamm Dis 2024; 12:e70105. [PMID: 39692539 PMCID: PMC11653711 DOI: 10.1002/iid3.70105] [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: 03/05/2024] [Revised: 11/16/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Bacterial coinfection in patients with SARS-CoV-2 infection is an important risk factor for death. This study investigated whether there were differences in levels of serum inflammatory markers in COVID-19 patients with bacterial coinfections compared with those without bacterial infection. METHODS A total of 235 inpatients with SARS-CoV-2 infection admitted to Qingdao Central Hospital from December 7, 2022, to August 7, 2024, were included. Patients were divided into a bacteria-positive group (115 cases) and a bacteria-negative group (120 cases) according to whether they had bacterial coinfections. PCT, CRP, and 12 kinds of cytokines were compared between groups, and the distribution of bacterial species in the positive group was statistically analyzed. RESULTS The serum levels of CRP (Z = 8.94, p < 0.001), PCT (Z = 5.59, p < 0.001), IL-1β (t = 4.863, p < 0.001), IL-2 (t = 5.810, p < 0.001), IL-5 (t = 3.837, p < 0.001), IL-6 (t = 4.910, p < 0.001), IL-8 (t = 3.325, p < 0.001), ILIL-12p70 (t = 4.722, p < 0.001), IL-17 (t = 3.315, p = 0.001) and TNF-α (t = 4.251, p < 0.001) between the two groups were significantly different. IL-4, IL-10, IFN-α, and IFN-γ were not statistically significant (p > 0.05). Among the 115 bacteria-positive patients, 56 patients were positive for one species and 59 patients were multiple infections. Acinetobacter baumannii, Klebsiella pneumoniae, Pseudomonas aeruginosa, Staphylococcus aureus, and Haemophilus influenzae were common species. CONCLUSIONS Serum PCT and CRP levels in COVID-19 patients with bacterial coinfection are higher than those without bacterial infection. Cytokines such as IL-1β, IL-2, IL-5, IL-6, IL-8, IL-12p70, IL-17, and TNF-α may be involved in the progression of COVID-19 combined with bacterial infection. They can be used as potential markers to evaluate the disease condition and prognosis.
Collapse
Affiliation(s)
- Qingqing Bi
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
| | - Jie Zhu
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
| | - Jinju Zheng
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
| | - Qingyun Xu
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
- Department of Clinical LaboratoryPeking University First HospitalBeijingChina
| | - Juan Chen
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
| | - Lei Zhang
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
| | - Xiaofeng Mu
- Department of Laboratory MedicineQingdao Central HospitalQingdaoChina
| |
Collapse
|
17
|
Meybodi SM, Rabori VS, Salkhorde D, Jafari N, Zeinaly M, Mojodi E, Kesharwani P, Saberiyan M, Sahebkar A. Dexamethasone in COVID-19 treatment: Analyzing monotherapy and combination therapy approaches. Cytokine 2024; 184:156794. [PMID: 39489912 DOI: 10.1016/j.cyto.2024.156794] [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: 07/19/2024] [Revised: 10/12/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
The COVID-19 pandemic has prompted the exploration of effective treatment options, with dexamethasone emerging as a key corticosteroid for severe cases. This review evaluates the efficacy and safety of dexamethasone, highlighting its ability to reduce mortality rates, alleviate acute respiratory distress syndrome (ARDS), and mitigate hyperinflammation. While dexamethasone shows therapeutic promise, potential adverse effects-including cardiovascular issues, neuropsychiatric complications, lung infections, and liver damage-necessitate careful monitoring and individualized treatment strategies. The review also addresses the debate over using dexamethasone alone versus in combination with other therapies targeting SARS-CoV-2, examining potential synergistic effects and drug resistance. In summary, dexamethasone is a valuable treatment option for COVID-19 but its risks highlight the need for tailored surveillance approaches. Further research is essential to establish clear guidelines for optimizing treatment and improving patient outcomes.
Collapse
Affiliation(s)
| | | | - Darya Salkhorde
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Negar Jafari
- Department of Cardiology, Urmia University of Medical Sciences, Urmia, Iran
| | - Mahsa Zeinaly
- Department of Biology, Faculty of Science, University of Guilan
| | - Elham Mojodi
- Depatment of Biology, Faculty of Science, Yazd University, Yazd, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Mohammadreza Saberiyan
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran; Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
18
|
Kjellberg A, Zhao A, Lussier A, Hassler A, Al-Ezerjawi S, Boström E, Catrina SB, Bergman P, Rodriguez-Wallberg KA, Lindholm P. Hyperbaric oxygen therapy as an immunomodulatory intervention in COVID-19-induced ARDS: Exploring clinical outcomes and transcriptomic signatures in a randomised controlled trial. Pulm Pharmacol Ther 2024; 87:102330. [PMID: 39393522 DOI: 10.1016/j.pupt.2024.102330] [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: 08/20/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
Immunomodulatory agents with the potential to reverse critical COVID-19, targeting host-virus immune response are needed. In this exploratory sub study of a randomised controlled clinical trial, critical COVID-19 patients with moderate acute respiratory distress syndrome at one Swedish university hospital were randomly assigned (1:1) to hyperbaric oxygen therapy (HBOT) group plus best practice, or best practice (Control). Follow-up was 30 days. HBOT was administered with five treatments at 2.4 atm absolute (ATA), lasting 80 min, within the first seven days. Clinical outcome, inflammatory markers, and bulk RNA sequencing (RNAseq) on peripheral blood mononuclear cells were analysed. Between December 3rd, 2020, and May 17th, 2021, 23 patients were randomised, and 17 were analysed. RNA-sequencing revealed 791 differentially expressed genes in the HBOT group compared to 46 in the control group at Day 7 vs. baseline. Gene set enrichment analysis revealed a unique transcriptomic signature associated with endoplasmic reticulum stress (ERS) in the HBOT group. Patients in the HBOT group recovered faster and had a shorter mean hospital length of stay (HLoS), 16 vs. 26 days (95.99 % CI -16-0), p = 0.045. National early warning score (NEWS) was lower in the HBOT group (ANOVA, F [8, 120] = 3.817, p < 0.001) and PaO2/FiO2 was higher in the HBOT group (Mixed effects model, F [8, 94] = 2.900, p < 0.01). We showed a unique transcriptomic signature related to viral-induced ERS in critically ill COVID-19 patients treated with HBOT. The finding was associated with a positive clinical outcome; the HBOT patients recovered faster and had a reduced HLoS compared with controls. TRIAL REGISTRATION: NCT04327505 (March 31, 2020) and EudraCT 2020-001349-37 (April 24, 2020).
Collapse
Affiliation(s)
- Anders Kjellberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Perioperative Medicine and Intensive Care Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | - Allan Zhao
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Anna Lussier
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Department of Emergency Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Adrian Hassler
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Acute and Reparative Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Sarah Al-Ezerjawi
- Acute and Reparative Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Emil Boström
- Acute and Reparative Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Sergiu-Bogdan Catrina
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Academic Specialist Center, Center for Diabetes, Stockholm, Sweden
| | - Peter Bergman
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Kenny Alexandra Rodriguez-Wallberg
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Reproductive Medicine, Division of Gynaecology and Reproduction, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Lindholm
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Department of Emergency Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
19
|
Tjiptaningrum A, Kurniati I, Fadilah F, Susantiningsih T, Prawiningrum AF, Utari WD, Erlina L. Protein Interaction Analysis and Molecular Simulation of the Anti-Inflammatory Activities in Melaleuca cajuputi Extract Against COVID-19. Int J Inflam 2024; 2024:5568294. [PMID: 39640429 PMCID: PMC11620808 DOI: 10.1155/ijin/5568294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 07/22/2024] [Accepted: 09/12/2024] [Indexed: 12/07/2024] Open
Abstract
Coronavirus disease-19 (COVID-19) is correlated to a severe condition caused by a cytokine storm during which numerous proinflammatory cytokines, including interleukin-6 (IL-6) are released. IL-6 is a critical driver in the COVID-19 inflammatory state, and the inhibition is considered a potential treatment approach to prevent serious complications. Meanwhile, Melaleuca cajuputi is a plant with antibacterial, antiviral, anti-inflammatory, and antioxidant activities. Therefore, this aimed to investigate the anti-inflammatory potential of M. cajuputi in silico. Extraction of leaves was conducted by using 96% ethanol, followed by fractionation to obtain active compounds. Subsequently, LC/MS and GC/MS analyses were performed to obtain active compound profiling. Protein-protein interaction (PPI), as well as molecular docking and dynamic analyses, were performed to examine interaction of active compounds of M. cajuputi with IL-6. The results showed that 30 protein nodes played a significant role in COVID-19 cytokine storm and eight active compounds had interactions with IL-6. Among the active compounds, pinostrobin chalcone had the best delta G interaction with IL-6. In conclusion, M. cajuputi has potential activity as an anti-inflammatory agent against COVID-19.
Collapse
Affiliation(s)
- Agustyas Tjiptaningrum
- Doctoral Program of Biomedical Science, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
- Department of Clinical Pathology of University of Lampung, Bandar Lampung, Indonesia
| | - Intanri Kurniati
- Department of Clinical Pathology of University of Lampung, Bandar Lampung, Indonesia
| | - Fadilah Fadilah
- Department of Medical Chemistry, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Bioinformatics Core Facilities, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Tiwuk Susantiningsih
- Doctoral Program of Biomedical Science, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
| | - Aisyah Fitriannisa Prawiningrum
- Bioinformatics Core Facilities, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Wahyu Dian Utari
- Bioinformatics Core Facilities, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Linda Erlina
- Doctoral Program of Biomedical Science, Faculty of Medicine, University of Indonesia, Jakarta, Indonesia
- Bioinformatics Core Facilities, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
20
|
Dhakal S, Yin A, Escarra-Senmarti M, Demko ZO, Pisanic N, Johnston TS, Trejo-Zambrano MI, Kruczynski K, Lee JS, Hardick JP, Shea P, Shapiro JR, Park HS, Parish MA, Caputo C, Ganesan A, Mullapudi SK, Gould SJ, Betenbaugh MJ, Pekosz A, Heaney CD, Antar AAR, Manabe YC, Cox AL, Karaba AH, Andrade F, Zeger SL, Klein SL. Application of machine learning algorithms to identify serological predictors of COVID-19 severity and outcomes. COMMUNICATIONS MEDICINE 2024; 4:249. [PMID: 39592832 PMCID: PMC11599591 DOI: 10.1038/s43856-024-00658-w] [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: 10/25/2023] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Critically ill hospitalized patients with COVID-19 have greater antibody titers than those with mild to moderate illness, but their association with recovery or death from COVID-19 has not been characterized. METHODS In a cohort study of 178 COVID-19 patients, 73 non-hospitalized and 105 hospitalized patients, mucosal swabs and plasma samples were collected at hospital enrollment and up to 3 months post-enrollment (MPE) to measure virus RNA, cytokines/chemokines, binding antibodies, ACE2 binding inhibition, and Fc effector antibody responses against SARS-CoV-2. The association of demographic variables and more than 20 serological antibody measures with intubation or death due to COVID-19 was determined using machine learning algorithms. RESULTS Predictive models reveal that IgG binding and ACE2 binding inhibition responses at 1 MPE are positively and anti-Spike antibody-mediated complement activation at enrollment is negatively associated with an increased probability of intubation or death from COVID-19 within 3 MPE. CONCLUSIONS At enrollment, serological antibody measures are more predictive than demographic variables of subsequent intubation or death among hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Santosh Dhakal
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Anna Yin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Zoe O Demko
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Nora Pisanic
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Trevor S Johnston
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - Kate Kruczynski
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - John S Lee
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Justin P Hardick
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Patrick Shea
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Janna R Shapiro
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Han-Sol Park
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Maclaine A Parish
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Christopher Caputo
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Abhinaya Ganesan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sarika K Mullapudi
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stephen J Gould
- Department of Biological Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Advanced Mammalian Biomanufacturing Innovation Center, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Christopher D Heaney
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Annukka A R Antar
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Yukari C Manabe
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Andrea L Cox
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Andrew H Karaba
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Felipe Andrade
- Division of Rheumatology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Scott L Zeger
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
21
|
Chagas JPS, de Oliveira JR, Brandão VA, Bellucio APB, Dutra JVDA, Dutra JR, Cerutti C. Evaluating imatinib in severe COVID-19: a systematic review and meta-analysis. Trans R Soc Trop Med Hyg 2024:trae095. [PMID: 39587918 DOI: 10.1093/trstmh/trae095] [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: 06/13/2024] [Revised: 08/09/2024] [Accepted: 10/17/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Considering the potential antiviral and immunomodulatory properties of imatinib, this drug has been investigated in several studies as a potential treatment option for severe cases of COVID-19, given that treatment modalities available remain limited. OBJECTIVES To evaluate the benefits or otherwise of imatinib for COVID-19 in severely ill patients, we performed a systematic review of studies that tested the efficacy and the safety of imatinib for COVID-19 and executed a meta-analysis. METHODS We searched Medline, Embase and Cochrane with the following search terms: 'coronavirus', 'SARS-Cov2', 'covid', 'covid-19' and 'imatinib'. The latest search date was November 2023. We used Cochrane Collaboration's tool to assess bias in randomized trials. RESULTS We included three randomized controlled trials with 561 participants. A total of 276 patients received imatinib and 285 received placebo. The mortality results showed no statistically significant differences between imatinib and controls (RR 0.61; 95% CI 0.37 to 1.01; p=0.06). There was no significant difference in length of hospital stay or severe adverse events occurring between groups. CONCLUSIONS Current evidence suggests that the potential benefits of imatinib should be further evaluated in randomized controlled trials in patients hospitalized for COVID-19.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Crispim Cerutti
- Departamento de Medicina Social, Universidade Federal do Espírito Santo, Vitória 29090-040, Brazil
| |
Collapse
|
22
|
Oh DS, Kim E, Normand R, Lu G, Shook LL, Lyall A, Jasset O, Demidkin S, Gilbert E, Kim J, Akinwunmi B, Tantivit J, Tirard A, Arnold BY, Slowikowski K, Goldberg MB, Filbin MR, Hacohen N, Nguyen LH, Chan AT, Yu XG, Li JZ, Yonker L, Fasano A, Perlis RH, Pasternak O, Gray KJ, Choi GB, Drew DA, Sen P, Villani AC, Edlow AG, Huh JR. SARS-CoV-2 infection elucidates features of pregnancy-specific immunity. Cell Rep 2024; 43:114933. [PMID: 39504241 PMCID: PMC11724703 DOI: 10.1016/j.celrep.2024.114933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 07/30/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024] Open
Abstract
Pregnancy is a risk factor for increased severity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other respiratory infections, but the mechanisms underlying this risk are poorly understood. To gain insight into the role of pregnancy in modulating immune responses at baseline and upon SARS-CoV-2 infection, we collected peripheral blood mononuclear cells and plasma from 226 women, including 152 pregnant individuals and 74 non-pregnant women. We find that SARS-CoV-2 infection is associated with altered T cell responses in pregnant women, including a clonal expansion of CD4-expressing CD8+ T cells, diminished interferon responses, and profound suppression of monocyte function. We also identify shifts in cytokine and chemokine levels in the sera of pregnant individuals, including a robust increase of interleukin-27, known to drive T cell exhaustion. Our findings reveal nuanced pregnancy-associated immune responses, which may contribute to the increased susceptibility of pregnant individuals to viral respiratory infection.
Collapse
Affiliation(s)
- Dong Sun Oh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Eunha Kim
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; BK21 Graduate Program, Department of Biomedical Sciences and Department of Neuroscience, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Rachelly Normand
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Guangqing Lu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Lydia L Shook
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amanda Lyall
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Olyvia Jasset
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stepan Demidkin
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Emily Gilbert
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Joon Kim
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Babatunde Akinwunmi
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jessica Tantivit
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alice Tirard
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Benjamin Y Arnold
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kamil Slowikowski
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Marcia B Goldberg
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA; Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Michael R Filbin
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nir Hacohen
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Long H Nguyen
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Xu G Yu
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Jonathan Z Li
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Lael Yonker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Roy H Perlis
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kathryn J Gray
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Gloria B Choi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David A Drew
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Pritha Sen
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA; Transplant, Oncology, and Immunocompromised Host Group, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Alexandra-Chloé Villani
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA.
| | - Andrea G Edlow
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
23
|
Cheng J, Wang H, Li C, Yu J, Zhu M. Characteristics of cytokines/chemokines associated with disease severity and adverse prognosis in COVID-19 patients. Front Immunol 2024; 15:1464545. [PMID: 39654886 PMCID: PMC11625740 DOI: 10.3389/fimmu.2024.1464545] [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: 07/14/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024] Open
Abstract
Background Cytokines and chemokines as crucial participants in innate immune response play significant roles during SARS-CoV-2 infection, yet excessive immune response exacerbates the severity of COVID-19. Purpose This study aims to investigate the involvement of which cytokines/chemokines in the cytokine storm of COVID-19, as well as the changes in cytokine/chemokine levels during the course of COVID-19, simultaneously exploring the diagnostic and prognostic value of the relevant cytokines/chemokines for COVID-19. Methods Flow cytometry was employed to detect the levels of cytokines and chemokines in the serum of 50 COVID-19 patients. Results Compared with severe COVID-19 patients, the levels of cytokines IL-6, IL-8, IL-10, sCD25, and chemokines IP-10 and MIG in the peripheral blood of non-severe patients were significantly reduced, while only IL-6, IL-10, and IP-10 levels were significantly decreased compared to non-survivors of COVID-19. Meanwhile, serum concentrations of IP-10, MCP-1, sTREM-1, IL-10, and the neutrophil-to-lymphocyte ratio (NLR) in peripheral blood could distinguish between COVID-19 survivors and non-survivors and were significantly associated with mortality. Among them, the concentration of IP-10 was shown to be the most powerful indicator for predicting adverse outcomes in COVID-19 patients (AUC: 0.715); however, its combined detection with the conventional inflammatory marker NLR did not improve the predictive value for adverse outcomes in COVID-19 patients. Additionally, serum IP-10 levels were negatively correlated with peripheral blood NK cell count and total lymphocyte count, while sTREM-1 levels were positively correlated with peripheral blood CD4+ T cell count and CD3+ T cell count. Meanwhile, IL-8 levels were positively correlated with total lymphocyte count in peripheral blood. Finally, the serum levels of cytokines/chemokines in non-survivors of COVID-19 increased significantly before death, while in survivors, they returned to normal levels before discharge. Conclusions Severely ill and non-surviving COVID-19 patients exhibit compromised immune function, with significantly higher levels of inflammation, cytokine/chemokine storms, and immune dysregulation compared to non-severe patients. Serum concentrations of IP-10, MCP-1, sTREM-1, and IL-10 levels can serve as biomarkers to predict adverse outcomes in COVID-19.
Collapse
Affiliation(s)
- Jianghao Cheng
- Department of Open Laboratory Medicine, Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Haozhen Wang
- Department of General Practice, Jinhua People’s Hospital, Jinhua, China
| | - Chaodan Li
- Department of Open Laboratory Medicine, Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianhua Yu
- Department of Infectious Diseases, Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Mingli Zhu
- Department of Open Laboratory Medicine, Hangzhou Xixi Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
24
|
Vukovic J, Jukic I, Tonkic A. The Challenges in Treating Inflammatory Bowel Diseases During the COVID-19 Pandemic: An Opinion. J Clin Med 2024; 13:7128. [PMID: 39685586 DOI: 10.3390/jcm13237128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
The COVID-19 pandemic posed significant challenges in the treatment of chronic diseases, particularly inflammatory bowel diseases (IBDs) such as Crohn's disease and ulcerative colitis. These challenges are multifaceted, encompassing difficulties in maintaining routine care, concerns about the safety of immunosuppressive therapies, disruptions in healthcare delivery, and the complexities of managing IBD in patients who contract COVID-19. This article explores the various obstacles faced in the treatment of IBD during the pandemic and discusses potential strategies to overcome these challenges.
Collapse
Affiliation(s)
- Jonatan Vukovic
- Department of Gastroenterology and Hepatology, Internal Clinic, Clinical Hospital Centre Split, Spinciceva 1, 21000 Split, Croatia
- Department of Internal Medicine, School of Medicine, University of Split, 21000 Split, Croatia
| | - Ivana Jukic
- Department of Gastroenterology and Hepatology, Internal Clinic, Clinical Hospital Centre Split, Spinciceva 1, 21000 Split, Croatia
- University Department of Health Studies, University of Split, 21000 Split, Croatia
| | - Ante Tonkic
- Department of Gastroenterology and Hepatology, Internal Clinic, Clinical Hospital Centre Split, Spinciceva 1, 21000 Split, Croatia
- Department of Internal Medicine, School of Medicine, University of Split, 21000 Split, Croatia
| |
Collapse
|
25
|
Singh T, Macintyre AN, Burke TW, Anderson J, Petzold E, Stover EL, French MJ, Oguin TH, Demarco T, McClain MT, Ko ER, Park LP, Denny T, Sempowski GD, Woods CW. Dynamics of cytokine and antibody responses in community versus hospital SARS-CoV-2 infections. Front Immunol 2024; 15:1468871. [PMID: 39650666 PMCID: PMC11621060 DOI: 10.3389/fimmu.2024.1468871] [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: 07/22/2024] [Accepted: 10/17/2024] [Indexed: 12/11/2024] Open
Abstract
Introduction Dysregulated host cytokine responses to SARS-CoV-2 infection are a primary cause of progression to severe disease, whereas early neutralizing antibody responses are considered protective. However, there are gaps in understanding the early temporal dynamics of these immune responses, and the profile of productive immune responses generated by non-hospitalized people with mild infections in the community. Methods Here we conducted a prospective cohort study of people with suspected infections/exposures in the US state of North Carolina, before vaccine availability. We recruited participants not only in hospitals/clinics, but also in their homes. With serial sampling, we compared virologic and immunologic factors in 258 community cases versus 114 hospital cases of COVID-19 to define factors associated with severity. Results We found that high early neutralizing antibodies were associated with lower nasal viral load, but not protection from hospitalization. Cytokine responses were evaluated in 125 cases, with subsets at first versus second week of illness to assess for time-dependent trajectories. The hospital group demonstrated a higher magnitude of serum IL-6, IL-1R antagonist, IP-10, and MIG; prolonged upregulation of IL-17; and lesser downregulation of GROα, IL-1R antagonist, and MCP1, in comparison to the community group suggesting that these factors may contribute to immunopathology. In the second week of illness, 2-fold increases in IL-6, IL-1R antagonist, and IP-10 were associated with 2.2, 1.8, and 10-fold higher odds of hospitalization respectively, whereas a 2-fold increase in IL-10 was associated with 63% reduction in odds of hospitalization (p<0.05). Moreover, antibody responses at 3-6 months post mild SARS-CoV-2 infections in the community revealed long-lasting antiviral IgM and IgA antibodies as well as a stable set point of neutralizing antibodies that were not waning. Discussion Our data provide valuable temporal cytokine benchmarks to track the progression of immunopathology in COVID-19 patients and guide improvements in immunotherapies.
Collapse
Affiliation(s)
- Tulika Singh
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, United States
- Duke Global Health Institute, Durham, NC, United States
| | - Andrew N. Macintyre
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Thomas W. Burke
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, NC, United States
| | - Jack Anderson
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, NC, United States
| | - Elizabeth Petzold
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, NC, United States
| | - Erica L. Stover
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Matthew J. French
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Thomas H. Oguin
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Todd Demarco
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Micah T. McClain
- Duke Global Health Institute, Durham, NC, United States
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, NC, United States
- Division of General Internal Medicine, Department of Medicine, Duke School of Medicine, Durham, NC, United States
| | - Emily R. Ko
- Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, NC, United States
- Division of General Internal Medicine, Department of Medicine, Duke School of Medicine, Durham, NC, United States
| | - Lawrence P. Park
- Duke Global Health Institute, Durham, NC, United States
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Thomas Denny
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Gregory D. Sempowski
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
- RTI International, Research Triangle Park, NC, United States
| | - Christopher W. Woods
- Duke Global Health Institute, Durham, NC, United States
- Division of Infectious Diseases, Department of Medicine, Duke University Medical Center, Durham, NC, United States
- Center for Infectious Disease Diagnostics and Innovation, Duke University, Durham, NC, United States
- Division of General Internal Medicine, Department of Medicine, Duke School of Medicine, Durham, NC, United States
| |
Collapse
|
26
|
Anshori I, Marcius D, Syaifie PH, Siregar KAAK, Syakuran LA, Jauhar MM, Arda AG, Shalannanda W, Mardliyati E. Therapeutic Potential of Propolis Extract in Managing Hyperinflammation and Long COVID-19: A Bioinformatics Study. Chem Biodivers 2024:e202401947. [PMID: 39576127 DOI: 10.1002/cbdv.202401947] [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: 08/13/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/06/2024]
Abstract
Hyperinflammation is a significant factor in long COVID, impacting over 65 million post-COVID-19 individuals globally. Herbal remedies, including propolis, show promise in reducing severity and pro-inflammatory cytokines. However, the natural pharmacological role of propolis in COVID-19 management remains underexplored. Employing network pharmacology and in silico techniques, we assessed propolis extract's potential in countering SARS-CoV-2-induced inflammation. We identified 80 flavonoids via LC-MS/MS QTOF and employed 11 anti-inflammatory drugs as references for inflammation target fishing. Utilizing in silico techniques encompassing target fishing, molecular docking, and dynamics, we examined propolis' effects. We identified 1105 gene targets connected to inflammation through multiple validated target predictors. By integrating SARS-CoV-2 DEGs from GSE147507 with these targets, we identify 25 inflammation-COVID-19-associated propolis targets, including STAT1, NOS2, CFB, EIF2K2, NPY5R, and BTK. Enrichment analyses highlighted primary pharmacological pathways related to Epstein-Barr virus infection and COVID-19. Molecular docking validated isokaempferide, iristectorigenin B, 3'-methoxypuerarin, cosmosiin, and baicalein-7-O-β-d-glucopyranoside, which exhibited strong binding affinity and stability with relevant genes. Moreover, our findings indicate that propolis ligands could potentially suppress reactivation of Epstein-Barr Virus infections in post-COVID-19 cases. However, this study has a limitation in that the concentration of each propolis compound has not been quantified. Therefore, further exploration of propolis compounds quantification and experimental validation are needed to support these findings.
Collapse
Affiliation(s)
- Isa Anshori
- School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, Indonesia
- Research Center for Nanosciences and Nanotechnology (RCNN), Bandung Institute of Technology, Bandung, Indonesia
| | - Donny Marcius
- School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, Indonesia
| | - Putri Hawa Syaifie
- Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, Banten, Indonesia
| | - Khalish Arsy Al Khairy Siregar
- Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, Banten, Indonesia
- Faculty of Pharmacy, Universitas Muhammadiyah Kalimantan Timur, Samarinda, East Borneo, Indonesia
| | | | | | | | - Wervyan Shalannanda
- School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung, Indonesia
| | - Etik Mardliyati
- Nano Center Indonesia, Jl. PUSPIPTEK, South Tangerang, Banten, Indonesia
- Research Center for Vaccine and Drugs, National Research and Innovation Agency (BRIN), Cibinong, Indonesia
| |
Collapse
|
27
|
Tan W, Qi L, Tan Z. Animal models of infection-induced acute lung injury. Exp Lung Res 2024; 50:221-241. [PMID: 39558475 DOI: 10.1080/01902148.2024.2428939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/19/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024]
Abstract
Aim: Acute lung injury (ALI) is characterized by severe hypoxemia, reduced lung elasticity, and notable pulmonary edema, often caused by infections and potentially progressing to ARDS. This article explores animal models of ALI and clarifies its main pathogenic mechanisms. Materials and Methods: we reviewed 20 years of ALI animal model advancements via PubMed, assessing clinical symptoms, histopathology, and reproducibility, and provided guidance on selecting models aligned with ALI pathogenesis. Results: key proinflammatory mediators and interleukins play a significant role in ALI development, though their interactions are not fully understood. Preclinical models are essential for investigating ALI causes and testing treatments. Animal models mimic ALI from sources such as infections, drugs, and I/R events, but differences between mouse and human lungs necessitate careful validation of these findings. Conclusions: A comprehensive strategy is essential to address clinical treatment and drug R&D challenges to prevent severe complications and reduce mortality rates.
Collapse
Affiliation(s)
- Wanying Tan
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Lingjun Qi
- Institute of Traditional Chinese Medicine Pharmacology and Toxicology, Sichuan academy of Chinese Medicine Sciences, Chengdu, China
| | - Zhenghuai Tan
- Affiliated Sichuan Gem Flower Hospital of North Sichuan Medical College, Chengdu, China
| |
Collapse
|
28
|
Niayesh-Mehr R, Kalantar M, Bontempi G, Montaldo C, Ebrahimi S, Allameh A, Babaei G, Seif F, Strippoli R. The role of epithelial-mesenchymal transition in pulmonary fibrosis: lessons from idiopathic pulmonary fibrosis and COVID-19. Cell Commun Signal 2024; 22:542. [PMID: 39538298 PMCID: PMC11558984 DOI: 10.1186/s12964-024-01925-y] [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: 08/16/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Despite the tremendous advancements in the knowledge of the pathophysiology and clinical aspects of SARS-CoV-2 infection, still many issues remain unanswered, especially in the long-term effects. Mounting evidence suggests that pulmonary fibrosis (PF) is one of the most severe complications associated with COVID-19. Therefore, understanding the molecular mechanisms behind its development is helpful to develop successful therapeutic strategies. Epithelial to mesenchymal transition (EMT) and its cell specific variants endothelial to mesenchymal transition (EndMT) and mesothelial to mesenchymal transition (MMT) are physio-pathologic cellular reprogramming processes induced by several infectious, inflammatory and biomechanical stimuli. Cells undergoing EMT acquire invasive, profibrogenic and proinflammatory activities by secreting several extracellular mediators. Their activity has been implicated in the pathogenesis of PF in a variety of lung disorders, including idiopathic pulmonary fibrosis (IPF) and COVID-19. Aim of this article is to provide an updated survey of the cellular and molecular mechanisms, with emphasis on EMT-related processes, implicated in the genesis of PF in IFP and COVID-19.
Collapse
Affiliation(s)
- Reyhaneh Niayesh-Mehr
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mojtaba Kalantar
- Department of Occupational Health, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | - Giulio Bontempi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Claudia Montaldo
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Saeedeh Ebrahimi
- Department of Medical Microbiology (Bacteriology and Virology), Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ghader Babaei
- Department of Clinical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Faezeh Seif
- Department of Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran.
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
- Gene Expression Laboratory, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy.
| |
Collapse
|
29
|
Chewaskulyong B, Satjaritanun P, Ketpueak T, Suksombooncharoen T, Charoentum C, Nuchpong N, Tantraworasin A. Neutralizing antibodies and safety of a COVID-19 vaccine against SARS-CoV-2 wild-type and Omicron variants in solid cancer patients. PLoS One 2024; 19:e0310781. [PMID: 39509358 PMCID: PMC11542819 DOI: 10.1371/journal.pone.0310781] [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: 07/13/2024] [Accepted: 09/05/2024] [Indexed: 11/15/2024] Open
Abstract
OBJECTIVE The aim of this study was to assess the seroconversion rate and percent inhibition of neutralizing antibodies against the wild-type and Omicron variants of SARS-CoV-2 in patients with solid cancer who received two COVID-19 vaccine doses by comparing chemotherapy and nonchemotherapy groups. METHODS This prospective cohort study enrolled 115 cancer patients from Maharaj Nakorn Chiang Mai Hospital, Sriphat Medical Center, Faculty of Medicine, Chiang Mai University, and Chiang Mai Klaimor Hospital, Chiang Mai, Thailand, between August 2021 and February 2022, with data from 91 patients who received two COVID-19 vaccine doses analyzed. Participants received vaccines as part of their personal vaccination programs, including various mRNA and non-mRNA vaccine combinations. Blood samples were collected at baseline, on day 28, and at 6 months post-second dose to assess neutralizing antibodies. The primary outcome was the seroconversion rate against the wild-type and Omicron variants on day 28. Secondary outcomes included seroconversion at 6 months, factors associated with seroconversion, and safety. RESULTS Among the participants, 45% were receiving chemotherapy. On day 28, seroconversion rates were 77% and 62% for the wild-type and Omicron variants, respectively. Chemotherapy did not significantly affect seroconversion rates (p = 0.789 for wild type, p = 0.597 for Omicron). The vaccine type administered was positively correlated with seroconversion, with an adjusted odds ratio (95% confidence interval) of 25.86 (1.39-478.06) for the wild type and 17.38 (3.65-82.66) for the Omicron variant with the primary heterologous vaccine regimen. Grades 1 and 2 adverse events were observed in 34.0% and 19.7% of participants, respectively. CONCLUSIONS Despite the lower seroconversion rate against the Omicron variant, no significant difference was observed between the chemotherapy and nonchemotherapy groups. COVID-19 vaccinations demonstrated good tolerability in this cohort. These findings highlight the importance of vaccine safety and immunogenicity in cancer patients and can inform tailored vaccination strategies for this vulnerable population.
Collapse
Affiliation(s)
- Busyamas Chewaskulyong
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pattarapong Satjaritanun
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thanika Ketpueak
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thatthan Suksombooncharoen
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chaiyut Charoentum
- Division of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nuttaphoom Nuchpong
- Medical Oncology Outpatient Clinic, Maharaj Nakorn Chiang Mai Hospital, Chiang Mai University, Chiang Mai, Thailand
| | - Apichat Tantraworasin
- Department of Surgery, General Thoracic Unit, Faculty of Medicine, and Clinical Surgical Research Center, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
30
|
Levitz D, Chao Foong Y, Sanfilippo P, Spelman T, Rath L, Roldan A, Lal A, Monif M, Jokubaitis V, Ozakbas S, Alroughani R, Boz C, Terzi M, Kalincik T, Blanco Y, Foschi M, Surcinelli A, Buzzard K, Skibina O, Laureys G, Van Hijfte L, Ramo-Tello C, Soysal A, Sanchez-Menoyo JL, Habek M, Cartechini E, Rojas JI, Karabudak R, Willekens B, Al-Harbi T, Fragoso Y, Castillo-Triviño T, Decoo D, Aragon de Vecino MC, Skromne E, Sirbu CA, Zhu C, Merlo D, Gresle M, Butzkueven H, Van Der Walt A. The impact of COVID-19 infection on multiple sclerosis disease course across 12 countries: a propensity-score-matched cohort study. Ther Adv Neurol Disord 2024; 17:17562864241278496. [PMID: 39525878 PMCID: PMC11544652 DOI: 10.1177/17562864241278496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 08/12/2024] [Indexed: 11/16/2024] Open
Abstract
Background The relationship between coronavirus disease 2019 (COVID-19) infection and multiple sclerosis (MS) relapse and disease progression remains unclear. Previous studies are limited by small sample sizes and most lack a propensity-matched control cohort. Objective To evaluate the effect of COVID-19 infection on MS disease course with a large propensity-matched cohort. Design This multi-centre cohort study analysed relapse and disability outcomes post-COVID-19 infection after balancing covariates using a propensity score matching method. The study period was from the 11th of September 2019 to the 16th of February 2023. The mean follow-up period was 1.7 years. Methods Data were retrieved from the MSBase Registry. Propensity scores were obtained based on age, sex, disease duration, baseline Expanded Disability Status Scale (EDSS), MS course, relapses pre-baseline, disease-modifying therapy (DMT) class and country. Primary outcomes were time to first relapse, annualised relapse rate (ARR) and time to confirm EDSS progression. Secondary outcomes were time to EDSS of 3, 4 or 6. Sensitivity analyses with baseline DMT classes were performed. Results The study included 2253 cases and 6441 controls. After matching, there were 2161 cases and an equal number of matched controls. Cases had a significantly higher ARR (ARR = 0.10 [95% CI 0.09-0.11]) compared to controls (ARR = 0.07 [95% CI 0.06-0.08]). Cases had a significantly greater hazard of time to first relapse compared to controls (hazard ratio (HR) = 1.54 [95% CI 1.29-1.84]). There was no association between COVID-19 infection and 24-week EDSS progression (HR = 1.18 [95% CI 0.92-1.52]), or time to EDSS of 3, 4 or 6. For patients on interferons and glatiramer acetate (BRACE), COVID-19 infection was associated with a greater hazard of time to first relapse (HR = 1.83 [95% CI 1.25-2.68]) and greater hazard of time to EDSS of 3 (HR = 2.04 [95% CI 1.06-3.90]) compared to patients on BRACE therapy without COVID-19 infection. Conclusion COVID-19 infection was associated with a significantly increased MS relapse rate and a shorter time to first relapse. There was no effect on confirmed EDSS progression over the short term. These results support ongoing COVID-19 risk minimisation strategies to protect patients with MS.
Collapse
Affiliation(s)
- David Levitz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Yi Chao Foong
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Paul Sanfilippo
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Tim Spelman
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Louise Rath
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Angie Roldan
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Anoushka Lal
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Mastura Monif
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Vilija Jokubaitis
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | | | - Raed Alroughani
- Division of Neurology, Department of Medicine, Amiri Hospital, Kuwait City, Kuwait
| | - Cavit Boz
- KTU Medical Faculty Farabi Hospital, Trabzon, Turkey
| | - Murat Terzi
- Medical Faculty, 19 Mayis University, Samsun, Turkey
| | - Tomas Kalincik
- Neuroimmunology Centre, Department of Neurology, The Royal Melbourne Hospital, Melbourne, VIC, Australia
- CORe, Department of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Yolanda Blanco
- Center of Neuroimmunology, Service of Neurology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Matteo Foschi
- Department of Neuroscience, Neurology Unit, S. Maria delle Croci Hospital of Ravenna, AUSL Romagna, Ravenna, Italy
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, L’Aquila, Italy
| | - Andrea Surcinelli
- Department of Neuroscience, Neurology Unit, S. Maria delle Croci Hospital of Ravenna, AUSL Romagna, Ravenna, Italy
| | - Katherine Buzzard
- Department of Neurology, Box Hill Hospital, Melbourne, VIC, Australia
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
| | - Olga Skibina
- Department of Neurology, The Alfred Hospital, Melbourne, VIC, Australia
- Department of Neurology, Box Hill Hospital, Melbourne, VIC, Australia
- Eastern Health Clinical School, Monash University, Box Hill, VIC, Australia
| | - Guy Laureys
- Department of Neurology, University Hospital Ghent, Ghent, Belgium
| | | | | | - Aysun Soysal
- Bakirkoy Education and Research Hospital for Psychiatric and Neurological Diseases, Istanbul, Turkey
| | - Jose Luis Sanchez-Menoyo
- Hospital de Galdakao-Usansolo, Galdakao, Spain
- Instituto de Investigacion sanitario Biocruces-Bizkaia, Barakaldo, Spain
| | - Mario Habek
- Department of Neurology, University Hospital Center Zagreb, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | | | | | | | - Barbara Willekens
- Department of Neurology, Antwerp University Hospital, Edegem, Belgium
- Translational Neurosciences Research Group, Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium
| | - Talal Al-Harbi
- King Fahad Specialist Hospital-Dammam, Dammam, Saudi Arabia
| | - Yara Fragoso
- Universidade Metropolitana de Santos, Santos, Brazil
| | | | | | | | - Eli Skromne
- Hospital Angeles de las Lomas, Instituto Mexicano de Neurociencias, Huixquilucan, Mexico
| | - Carmen-Adella Sirbu
- Central Military Emergency University Hospital, Bucharest, Romania
- Titu Maiorescu University, Bucharest, Romania
| | - Chao Zhu
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Daniel Merlo
- Department of Neurology, Box Hill Hospital, Melbourne, VIC, Australia
| | - Melissa Gresle
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, VIC, Australia
| | - Anneke Van Der Walt
- Department of Neuroscience, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia
- Department of Neurology, The Alfred Hospital, Melbourne, VIC, Australia
| |
Collapse
|
31
|
Zucchelli A, Parigi M, Giliani S, Vetrano DL, Lucente D, Marzetti E, Calvani R, Bellelli G, Marengoni A. Older patients affected by COVID-19: investigating the existence of biological phenotypes. BMC Geriatr 2024; 24:923. [PMID: 39511501 PMCID: PMC11542346 DOI: 10.1186/s12877-024-05473-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/15/2024] [Indexed: 11/15/2024] Open
Abstract
INTRODUCTION COVID-19 provides an opportunity to examine biological phenotypes (observable morphological, functional and biological characteristics) in individuals who experience the same acute condition, potentially revealing differences in response to acute external stressors. The aim our study was to investigate biological phenotypes in older patients hospitalized for COVID-19, exploiting a panel of aging biomarkers. METHODS Data were gathered from the FRACOVID Project, an observational multicenter study, aimed to evaluate the impact of frailty on health-related outcomes in patients 60 + with COVID-19 in Northern Italy. A hierarchical cluster analysis was run using log-transformed and scaled values of TNF-a, IL-1 beta, IL-6, PAI-1, GDF-15, NT-proBNP, and Cystatin C evaluated at admission. RESULTS Eighty-one participants (mean age 75.3 years; 60.5% male) were evaluated. Frailty was identified in 42% of the sample and 27.2% were unable to ambulate outdoors. The mean hospital stay was 24.7 days, with an in-hospital mortality rate of 18.5%. Three biological phenotypes were found: (1) 'inflammatory', with high inflammatory biomarkers; (2) 'organ dysfunction', characterized by elevated cystatin C and NT-proBNP, and lower inflammatory markers; and (3) 'unspecific', with lower NT-proBNP and GDF-15 levels, and intermediate concentrations of other biomarkers. The 'organ dysfunction' phenotype showed the highest mean age and prevalence of frailty, disability, and chronic diseases. The 'inflammatory' phenotype showed the highest burden of respiratory and systemic signs and symptoms of infection. CONCLUSION Biological phenotypes might be used to identify different clinical and functional phenotypes in individuals affected by COVID-19.
Collapse
Affiliation(s)
- Alberto Zucchelli
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, 171 77, Sweden.
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.
| | - Marta Parigi
- A. Nocivelli Institute for Molecular Medicine, ASST Spedali Civili, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Silvia Giliani
- A. Nocivelli Institute for Molecular Medicine, ASST Spedali Civili, Brescia, Italy
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Davide Liborio Vetrano
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, 171 77, Sweden
- Stockholm Gerontology Research Center, Stockholm, Sweden
| | - Daniela Lucente
- Fondazione "Ospedale e Casa di Riposo Nobile Paolo Richiedei", Brescia, Italy
| | - Emanuele Marzetti
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Riccardo Calvani
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Giuseppe Bellelli
- School of Medicine and Surgery, Milano-Bicocca University, Monza, Italy
- Acute Geriatric Unit, IRCCS San Gerardo Foundation, Monza, Italy
| | - Alessandra Marengoni
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, 171 77, Sweden
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
32
|
Li S, Wang J, Dai X, Li C, Li T, Chen L. The PDZ domain of the E protein in SARS-CoV induces carcinogenesis and poor prognosis in LUAD. Microbes Infect 2024; 26:105381. [PMID: 38914369 DOI: 10.1016/j.micinf.2024.105381] [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: 05/31/2024] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND In both lung adenocarcinoma (LUAD) and severe acute respiratory syndrome (SARS), uncontrolled inflammation can be detected in lung tissue. The PDZ-binding motif (PBM) in the SARS-CoV-1 E protein has been demonstrated to be a virulence factor that induces a cytokine storm. METHODS To identify gene expression fluctuations induced by PBM, microarray sequencing data of lung tissue infected with wild-type (SARS-CoV-1-E-wt) or recombinant virus (SARS-CoV-1-E-mutPBM) were analyzed, followed by functional enrichment analysis. To understand the role of the screened genes in LUAD, overall survival and immune correlation were calculated. RESULTS A total of 12 genes might participate in the initial and developmental stages of LUAD through expression variation and mutation. Moreover, dysregulation of a total of 12 genes could lead to a poorer prognosis. In addition, the downregulation of MAMDC2 and ITGA8 by PBM could also affect patient prognosis. Although the conserved PBM (-D-L-L-V-) can be found at the end of the carboxyl terminus in multiple E proteins of coronaviruses, the specific function of each protein depends on the entire amino acid sequence. CONCLUSIONS In summary, PBM containing the SARS-CoV-1 E protein promoted the carcinogenesis of LUAD by dysregulating important gene expression profiles and subsequently influencing the immune response and overall prognosis.
Collapse
Affiliation(s)
- Shun Li
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, China; Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Jinxuan Wang
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, China
| | - Xiaozhen Dai
- School of Biosciences and Technology, Chengdu Medical College, Chengdu 610500, China
| | - Churong Li
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, China
| | - Tao Li
- Radiotherapy Center, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Long Chen
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu 610500, China; Department of Immunology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan 610500, China.
| |
Collapse
|
33
|
Seethamraju H, Yang OO, Loftus R, Ogbuagu O, Sammartino D, Mansour A, Sacha JB, Ojha S, Hansen SG, Arman AC, Lalezari JP. A Randomized Placebo-Controlled Trial of Leronlimab in Mild-To-Moderate COVID-19. Clin Ther 2024; 46:891-899. [PMID: 39353749 DOI: 10.1016/j.clinthera.2024.08.019] [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: 04/24/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 10/04/2024]
Abstract
PURPOSE Early in the course of the SARS-CoV-2 pandemic it was hypothesised that host genetics played a role in the pathophysiology of COVID-19 including a suggestion that the CCR5-Δ32 mutation may be protective in SARS-CoV-2 infection. Leronlimab is an investigational CCR5-specific humanized IgG4 monoclonal antibody currently in development for HIV-1 infection. We aimed to explore the impact of leronlimab on the severity of disease symptoms among participants with mild-to-moderate COVID-19. METHODS The TEMPEST trial was a randomized, double-blind, placebo-controlled study in participants with mild-to-moderate COVID-19. Participants were randomly assigned in a 2:1 ratio to receive subcutaneous leronlimab (700 mg) or placebo on days 0 and 7. The primary efficacy endpoint was assessed by change in total symptom score based on fever, myalgia, dyspnea, and cough, at end of treatment (day 14). FINDINGS Overall, 84 participants were randomized and treated with leronlimab (n = 56) or placebo (n = 28). No difference was observed in change in total symptom score (P = 0.8184) or other pre-specified secondary endpoints between treatments. However, in a post hoc analysis, 50.0% of participants treated with leronlimab demonstrated improvements from baseline in National Early Warning Score 2 (NEWS2) at day 14, compared with 20·8% of participants in the placebo group (post hoc; p = 0.0223). Among participants in this trial with mild-to-moderate COVID-19 adverse events rates were numerically but not statistically significantly lower in leronlimab participants (33.9%) compared with placebo participants (50.0%). IMPLICATIONS At the time the TEMPEST trial was designed although CCR5 was known to be implicated in COVID-19 disease severity the exact pathophysiology of SARS-CoV-2 infection was poorly understood. Today it is well accepted that SARS-CoV-2 infection in asymptomatic-to-mild cases is primarily characterized by viral replication, with a heightened immune response, accompanied by diminished viral replication in moderate-to-severe disease and a peak in inflammatory responses with excessive production of pro-inflammatory cytokines in critical disease. It is therefore perhaps not surprising that no differences between treatments were observed in the primary endpoint or in pre-specified secondary endpoints among participants with mild-to-moderate COVID-19. However, the results of the exploratory post hoc analysis showing that participants in the leronlimab group had greater improvement in NEWS2 assessment compared to placebo provided a suggestion that leronlimab may be associated with a lower likelihood of people with mild-to-moderate COVID-19 progressing to more severe disease and needs to be confirmed in other appropriately designed clinical trials. CLINICALTRIALS gov number, NCT04343651 https://classic. CLINICALTRIALS gov/ct2/show/NCT04343651.
Collapse
Affiliation(s)
| | - Otto O Yang
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | | | | | | | | | - Jonah B Sacha
- Oregon Health & Science University, Portland, Oregon
| | - Sohita Ojha
- Oregon Health & Science University, Portland, Oregon
| | | | | | | |
Collapse
|
34
|
Vorobyeva DA, Potashnikova DM, Maryukhnich EV, Rusakovich GI, Tvorogova AV, Kalinskaya AI, Pinegina NV, Kovyrshina AV, Dolzhikova IV, Postnikov AB, Rozov FN, Sotnikova TN, Kanner DY, Logunov DY, Gintsburg AL, Vasilieva EJ, Margolis LB. Cytokine production in an ex vivo model of SARS-CoV-2 lung infection. Front Immunol 2024; 15:1448515. [PMID: 39497823 PMCID: PMC11532052 DOI: 10.3389/fimmu.2024.1448515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/25/2024] [Indexed: 11/07/2024] Open
Abstract
Introduction The mechanisms of the SARS-CoV-2-triggered complex alterations in immune cell activation and production of cytokines in lung tissue remain poorly understood, in part because of the limited use of adequate tissue models that simulate the structure and cell composition of the lung in vivo. We developed a novel ex vivo model of SARS-CoV-2 infection of lung explants, that maintains the intact tissue composition and the viral load for up to 7-10 days. Using this model, we studied cytokine production during SARS-CoV-2 infection. Materials and methods Lung tissue was monitored for viability and cell composition using flow cytometry and histological analysis. SARS-CoV-2 infection was verified immunohistochemically, viral loads in tissue and culture medium were monitored by qPCR. A panel of 41 cytokines was measured in culture medium using xMAP technology. Results The explant lung tissue was viable and maintained viral infection that influenced the cytokine production. Elevated concentrations of G-CSF, GM-CSF, GRO-a, IFN-g, IL-6, IL-8, IP-10, MCP-3, MIP-1a, PDGF-AA, and VEGF, and decreased IL-1RA concentration were observed in infected tissue compared to non-infected tissue. Discussion Our results generally reflect the data obtained in COVID-19 patients. GRO-a, IFN-g, IL-6, IL-8, MCP-1, MCP-3, and RANTES correlated with the viral load, forming a distinct pro-inflammatory cluster. Thus, our lung ex vivo model faithfully reproduces some aspects of cytokine alterations in COVID-19 patients at an early disease stage, making the investigation of SARS-CoV-2 infection mechanisms more accessible and providing a potential platform for antiviral drug testing.
Collapse
Affiliation(s)
- Daria A. Vorobyeva
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Daria M. Potashnikova
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Elena V. Maryukhnich
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - George I. Rusakovich
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Anna V. Tvorogova
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Anna I. Kalinskaya
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Natalia V. Pinegina
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Anna V. Kovyrshina
- Federal Government Budgetary Institution “The National Research Center for Epidemiology and Microbiology Named After Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Inna V. Dolzhikova
- Federal Government Budgetary Institution “The National Research Center for Epidemiology and Microbiology Named After Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | | | | | - Tatiana N. Sotnikova
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | | | - Denis Yu. Logunov
- Federal Government Budgetary Institution “The National Research Center for Epidemiology and Microbiology Named After Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Alexander L. Gintsburg
- Federal Government Budgetary Institution “The National Research Center for Epidemiology and Microbiology Named After Honorary Academician N.F. Gamaleya” of the Ministry of Health of the Russian Federation, Moscow, Russia
| | - Elena J. Vasilieva
- Laboratory of Atherothrombosis, Cardiology Department, Federal State Budgetary Educational Institution of Higher Education (FSBEI HE) “Russian University of Medicine” of the Ministry of Health of the Russian Federation, Moscow, Russia
- I.V. Davydovsky Moscow City Clinical Hospital, Moscow Department of Healthcare, Moscow, Russia
| | - Leonid B. Margolis
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
| |
Collapse
|
35
|
Avdonin PP, Blinova MS, Serkova AA, Komleva LA, Avdonin PV. Immunity and Coagulation in COVID-19. Int J Mol Sci 2024; 25:11267. [PMID: 39457048 PMCID: PMC11508857 DOI: 10.3390/ijms252011267] [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: 08/19/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Discovered in late 2019, the SARS-CoV-2 coronavirus has caused the largest pandemic of the 21st century, claiming more than seven million lives. In most cases, the COVID-19 disease caused by the SARS-CoV-2 virus is relatively mild and affects only the upper respiratory tract; it most often manifests itself with fever, chills, cough, and sore throat, but also has less-common mild symptoms. In most cases, patients do not require hospitalization, and fully recover. However, in some cases, infection with the SARS-CoV-2 virus leads to the development of a severe form of COVID-19, which is characterized by the development of life-threatening complications affecting not only the lungs, but also other organs and systems. In particular, various forms of thrombotic complications are common among patients with a severe form of COVID-19. The mechanisms for the development of thrombotic complications in COVID-19 remain unclear. Accumulated data indicate that the pathogenesis of severe COVID-19 is based on disruptions in the functioning of various innate immune systems. The key role in the primary response to a viral infection is assigned to two systems. These are the pattern recognition receptors, primarily members of the toll-like receptor (TLR) family, and the complement system. Both systems are the first to engage in the fight against the virus and launch a whole range of mechanisms aimed at its rapid elimination. Normally, their joint activity leads to the destruction of the pathogen and recovery. However, disruptions in the functioning of these innate immune systems in COVID-19 can cause the development of an excessive inflammatory response that is dangerous for the body. In turn, excessive inflammation entails activation of and damage to the vascular endothelium, as well as the development of the hypercoagulable state observed in patients seriously ill with COVID-19. Activation of the endothelium and hypercoagulation lead to the development of thrombosis and, as a result, damage to organs and tissues. Immune-mediated thrombotic complications are termed "immunothrombosis". In this review, we discuss in detail the features of immunothrombosis associated with SARS-CoV-2 infection and its potential underlying mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Pavel V. Avdonin
- Koltzov Institute of Developmental Biology RAS, ul. Vavilova, 26, 119334 Moscow, Russia; (P.P.A.)
| |
Collapse
|
36
|
Bédard-Matteau J, Soulé A, Liu KY, Fourcade L, Fraser DD, Emad A, Rousseau S. Circulating IL-17F, but not IL-17A, is elevated in severe COVID-19 and leads to an ERK1/2 and p38 MAPK-dependent increase in ICAM-1 cell surface expression and neutrophil adhesion on endothelial cells. Front Immunol 2024; 15:1452788. [PMID: 39493750 PMCID: PMC11527637 DOI: 10.3389/fimmu.2024.1452788] [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: 06/21/2024] [Accepted: 09/17/2024] [Indexed: 11/05/2024] Open
Abstract
Background Severe COVID-19 is associated with neutrophilic inflammation and immunothrombosis. Several members of the IL-17 cytokine family have been associated with neutrophilic inflammation and activation of the endothelium. Therefore, we investigated whether these cytokines were associated with COVID-19. Methods We investigated the association between COVID-19 and circulating plasma levels of IL-17 cytokine family members in participants to the Biobanque québécoise de la COVID-19 (BQC19), a prospective observational cohort and an independent cohort from Western University (London, Ontario). We measured the in vitro impact of IL-17F on intercellular adhesion molecule 1 (ICAM-1) cell surface expression and neutrophil adhesion on endothelial cells in culture. The contribution of two Mitogen Activated Protein Kinase (MAPK) pathways was determined using small molecule inhibitors PD184352 (a MKK1/MKK2 inhibitor) and BIRB0796 (a p38 MAPK inhibitor). Results We found increased IL-17D and IL-17F plasma levels when comparing SARS-CoV-2-positive vs negative hospitalized participants. Moreover, increased plasma levels of IL-17D, IL-17E and IL-17F were noted when comparing severe versus mild COVID-19. IL-17F, but not IL-17A, was significantly elevated in people with COVID-19 compared to healthy controls and with more severe disease. In vitro work on endothelial cells treated with IL-17F for 24h showed an increase cell surface expression of ICAM-1 accompanied by neutrophil adhesion. The introduction of two MAPK inhibitors significantly reduced the binding of neutrophils while also reducing ICAM-1 expression at the surface level of endothelial cells, but not its intracellular expression. Discussion Overall, these results have identified an association between two cytokines of the IL-17 family (IL-17D and IL-17F) with COVID-19 and disease severity. Considering that IL-17F stimulation promotes neutrophil adhesion to the endothelium in a MAPK-dependent manner, it is attractive to speculate that this pathway may contribute to pathogenic immunothrombosis in concert with other molecular effectors.
Collapse
Affiliation(s)
- Jérôme Bédard-Matteau
- The Meakins-Christie Laboratories, Research Institute of the McGill University Heath Centre, Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| | - Antoine Soulé
- Department of Electrical and Computer Engineering, McGill University, Montréal, QC, Canada
| | - Katelyn Yixiu Liu
- The Meakins-Christie Laboratories, Research Institute of the McGill University Heath Centre, Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Lyvia Fourcade
- The Meakins-Christie Laboratories, Research Institute of the McGill University Heath Centre, Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC, Canada
| | - Douglas D. Fraser
- Children’s Health Research Institute & Lawson Health Research Institute, London, ON, Canada
- Department of Pediatrics, Western University, London, ON, Canada
| | - Amin Emad
- Department of Electrical and Computer Engineering, McGill University, Montréal, QC, Canada
- Mila, Quebec AI Institute, Montréal, QC, Canada
| | - Simon Rousseau
- The Meakins-Christie Laboratories, Research Institute of the McGill University Heath Centre, Montréal, QC, Canada
- Department of Medicine, Faculty of Medicine, McGill University, Montréal, QC, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada
| |
Collapse
|
37
|
Sababathy M, Ramanathan G, Ganesan S, Sababathy S, Yasmin A, Ramasamy R, Foo J, Looi Q, Nur-Fazila S. Multipotent mesenchymal stromal/stem cell-based therapies for acute respiratory distress syndrome: current progress, challenges, and future frontiers. Braz J Med Biol Res 2024; 57:e13219. [PMID: 39417447 PMCID: PMC11484355 DOI: 10.1590/1414-431x2024e13219] [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: 02/15/2024] [Accepted: 07/30/2024] [Indexed: 10/19/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a critical, life-threatening condition marked by severe inflammation and impaired lung function. Mesenchymal stromal/stem cells (MSCs) present a promising therapeutic avenue due to their immunomodulatory, anti-inflammatory, and regenerative capabilities. This review comprehensively evaluates MSC-based strategies for ARDS treatment, including direct administration, tissue engineering, extracellular vesicles (EVs), nanoparticles, natural products, artificial intelligence (AI), gene modification, and MSC preconditioning. Direct MSC administration has demonstrated therapeutic potential but necessitates optimization to overcome challenges related to effective cell delivery, homing, and integration into damaged lung tissue. Tissue engineering methods, such as 3D-printed scaffolds and MSC sheets, enhance MSC survival and functionality within lung tissue. EVs and MSC-derived nanoparticles offer scalable and safer alternatives to cell-based therapies. Likewise, natural products and bioactive compounds derived from plants can augment MSC function and resilience, offering complementary strategies to enhance therapeutic outcomes. In addition, AI technologies could aid in optimizing MSC delivery and dosing, and gene editing tools like CRISPR/Cas9 allow precise modification of MSCs to enhance their therapeutic properties and target specific ARDS mechanisms. Preconditioning MSCs with hypoxia, growth factors, or pharmacological agents further enhances their therapeutic potential. While MSC therapies hold significant promise for ARDS, extensive research and clinical trials are essential to determine optimal protocols and ensure long-term safety and effectiveness.
Collapse
Affiliation(s)
- M. Sababathy
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - G. Ramanathan
- Faculty of Computer Science and Information Technology, University Malaya, Kuala Lumpur, Malaysia
| | - S. Ganesan
- School of Pharmacy, Management and Science University, Shah Alam, Selangor, Malaysia
| | - S. Sababathy
- Faculty of Medicine and Defence Health, National Defence University of Malaysia, Sungai Besi, Kuala Lumpur, Malaysia
| | - A.R. Yasmin
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, University Putra Malaysia, Serdang, Selangor, Malaysia
- Laboratory of Vaccines and Biomolecules, Institute of Bioscience, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - R. Ramasamy
- Department of Pathology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang, Selangor, Malaysia
| | - J.B. Foo
- Center for Drug Discovery and Molecular Pharmacology (CDDMP), Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor, Malaysia
| | - Q.H. Looi
- My Cytohealth Sdn. Bhd., Bandar Seri Petaling, Kuala Lumpur, Malaysia
| | - S.H. Nur-Fazila
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, University Putra Malaysia, Serdang, Selangor, Malaysia
- Laboratory of Vaccines and Biomolecules, Institute of Bioscience, University Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
38
|
Ochar K, Iwar K, Nair VD, Chung YJ, Ha BK, Kim SH. The Potential of Glucosinolates and Their Hydrolysis Products as Inhibitors of Cytokine Storms. Molecules 2024; 29:4826. [PMID: 39459194 PMCID: PMC11510469 DOI: 10.3390/molecules29204826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
A cytokine storm is an intense inflammatory response characterized by the overproduction of proinflammatory cytokines, resulting in tissue damage, and organ dysfunction. Cytokines play a crucial role in various conditions, such as coronavirus disease, in which the immune system becomes overactive and releases excessive levels of cytokines, including interleukins, tumor necrosis factor-alpha (TNF-α), and interferon-gamma (IFN-γ). This anomalous response often leads to acute respiratory distress syndrome (ARDS), disseminated intravascular coagulation (DIC), and multiple organ injury (MOI). Glucosinolates are plant secondary metabolites predominantly found in Brassica vegetables, but are also present in other species, such as Moringa Adens and Carica papaya L. When catalyzed by the enzyme myrosinase, glucosinolates produce valuable products, including sulforaphane, phenethyl isothiocyanate, 6-(methylsulfinyl) hexyl isothiocyanate, erucin, goitrin, and moringin. These hydrolyzed products regulate proinflammatory cytokine production by inhibiting the nuclear factor kappa-light-chain-enhancer of activated B-cell (NF-κB) signaling pathway and stimulating the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. This action can alleviate hyperinflammation in infected cells and modulate cytokine storms. In this review, we aimed to examine the potential role of glucosinolates in modulating cytokine storms and reducing inflammation in various conditions, such as coronavirus disease. Overall, we found that glucosinolates and their hydrolysis products can potentially attenuate cytokine production and the onset of cytokine storms in diseased cells. In summary, glucosinolates could be beneficial in regulating cytokine production and preventing complications related to cytokine storms.
Collapse
Affiliation(s)
- Kingsley Ochar
- Council for Scientific and Industrial Research, Plant Genetic Resources Research Institute, Bunso P.O. Box 7, Ghana;
- National Agrobiodiversity Center, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea;
| | - Kanivalan Iwar
- National Agrobiodiversity Center, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea;
| | - Vadakkemuriyil Divya Nair
- Department of Plant Sciences, Central University of Himachal Pradesh, Shahpur Campus, Kangra District, Shahpur 176206, HP, India;
| | - Yun-Jo Chung
- National Creative Research Laboratory for Ca Signaling Network, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea;
| | - Bo-Keun Ha
- Department of Applied Plant Science, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seong-Hoon Kim
- National Agrobiodiversity Center, National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea;
| |
Collapse
|
39
|
Matsui T, Ushigome E, Hamaguchi M, Sudo K, Kitagawa N, Kondo Y, Hasegawa Y, Imai D, Hattori T, Yamazaki M, Sawa T, Fukui M. Increased Insulin Requirements in Severe Cases of Covid-19 are Higher Than in Moderate Cases. Diabetes Metab Syndr Obes 2024; 17:3727-3733. [PMID: 39539455 PMCID: PMC11558443 DOI: 10.2147/dmso.s480598] [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: 05/30/2024] [Accepted: 10/01/2024] [Indexed: 11/16/2024] Open
Abstract
Purpose Despite the low overall death rate of coronavirus disease 2019 (COVID-19), no study has examined the association between COVID-19 severity and the total daily insulin dose required for glycemic control. The aim of this study was to determine the maximum total daily insulin dose required according to COVID-19 severity, and the number of days required to reach the maximum insulin dose in patients with COVID-19 who used insulin during hospitalization. Patients and Methods This retrospective cohort study included participants aged 20-90 years with a confirmed diagnosis of COVID-19 who used insulin during hospitalization at Kyoto Prefectural University of Medicine Hospital between March 4, 2020, and May 31, 2021. Factors associated with maximum insulin dose during hospitalization were evaluated using linear regression analyses. Results The maximum insulin doses were 31.8, 76.8, and 230.7 U/day, and the numbers of days between COVID-19 diagnosis and the need for maximum insulin were 15.6, 17.1, and 13.7 days in patients without ventilator management, with ventilator management, and with ventilator and extracorporeal membrane oxygenation management, respectively. Multivariate linear regression analyses revealed that hemoglobin A1c level (β = 15.87, P = 0.001), use of a ventilator (β = 50.53, P < 0.001), and use of extracorporeal membrane oxygenation (β = 150.36, P < 0.001) were independent determinants of maximum insulin dose. Conclusion Patients with severe COVID-19 required a significantly higher maximum insulin dose than did those with moderate COVID-19. The maximum insulin dose was reached approximately 2 weeks after onset. Furthermore, the hemoglobin A1c level on admission and the use of a ventilator or extracorporeal membrane oxygenation during hospitalization were associated with the need for maximum insulin dose.
Collapse
Affiliation(s)
- Takaaki Matsui
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Emi Ushigome
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Kazuki Sudo
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Nobuko Kitagawa
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Yuriko Kondo
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Yuka Hasegawa
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Dan Imai
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Tomohiro Hattori
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Masahiro Yamazaki
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Teiji Sawa
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| |
Collapse
|
40
|
Mihai N, Olariu MC, Ganea OA, Adamescu AI, Molagic V, Aramă ȘS, Tilișcan C, Aramă V. Risk of Hepatitis B Virus Reactivation in COVID-19 Patients Receiving Immunosuppressive Treatment: A Prospective Study. J Clin Med 2024; 13:6032. [PMID: 39457983 PMCID: PMC11508539 DOI: 10.3390/jcm13206032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Objectives: This study aimed to evaluate the risk of hepatitis B virus reactivation (HBVr) in COVID-19 patients receiving immunosuppressive treatment, which has been insufficiently studied to date. Secondarily, we aimed to evaluate the seroprevalence of HBV infection in COVID-19 patients. Methods: We performed HBV screening on all Romanian adults hospitalized in four COVID-19 wards between October 2021 and September 2022. We enrolled patients with positive hepatitis B core antibody (anti-HBc) without protective hepatitis B surface antibody (anti-HBs), HBV treatment, or baseline immunosuppressive conditions, and we conducted a virological follow-up on these patients at 3 months. Results: We identified 333/835 (39.9%) anti-HBc-positive patients. Follow-up was performed for 13 patients with positive hepatitis B surface antigen (HBsAg) and 19 HBsAg-negative/anti-HBc-positive patients. Among those who received immunosuppressants, 4/23 (17.4%) patients experienced HBVr: 1 out of 8 (12.5%) HBsAg-positive patients (with 1.99 log increase in HBV DNA level) and 3 out of 15 (20%) HBsAg-negative/anti-HBc-positive patients (with a de novo detectable HBV DNA level). Conclusions: Administration of COVID-19 immunosuppressants may result in a significant risk of HBVr in co-infected patients. We recommend performing an HBV triple screen panel (HBsAg, anti-HBs, anti-HBc) for all COVID-19 patients receiving immunosuppressive treatment. HBV prophylaxis may be indicated in certain patients. Larger studies are needed in order to establish appropriate and cost-effective management for these patients.
Collapse
Affiliation(s)
- Nicoleta Mihai
- Faculty of Medicine, University of Medicine and Pharmacy ‘Carol Davila’, No 37, Dionisie Lupu, 020021 Bucharest, Romania; (N.M.); (O.-A.G.); (V.M.); (V.A.)
- National Institute of Infectious Diseases ‘Matei Bals’, 1 Dr. Calistrat Grozovici, 021105 Bucharest, Romania; (A.-I.A.); (Ș.S.A.); (C.T.)
| | - Mihaela Cristina Olariu
- Faculty of Medicine, University of Medicine and Pharmacy ‘Carol Davila’, No 37, Dionisie Lupu, 020021 Bucharest, Romania; (N.M.); (O.-A.G.); (V.M.); (V.A.)
- National Institute of Infectious Diseases ‘Matei Bals’, 1 Dr. Calistrat Grozovici, 021105 Bucharest, Romania; (A.-I.A.); (Ș.S.A.); (C.T.)
| | - Oana-Alexandra Ganea
- Faculty of Medicine, University of Medicine and Pharmacy ‘Carol Davila’, No 37, Dionisie Lupu, 020021 Bucharest, Romania; (N.M.); (O.-A.G.); (V.M.); (V.A.)
- National Institute of Infectious Diseases ‘Matei Bals’, 1 Dr. Calistrat Grozovici, 021105 Bucharest, Romania; (A.-I.A.); (Ș.S.A.); (C.T.)
| | - Aida-Isabela Adamescu
- National Institute of Infectious Diseases ‘Matei Bals’, 1 Dr. Calistrat Grozovici, 021105 Bucharest, Romania; (A.-I.A.); (Ș.S.A.); (C.T.)
- Faculty of Dental Medicine, University of Medicine and Pharmacy ‘Carol Davila’, No 37, Dionisie Lupu, 020021 Bucharest, Romania
| | - Violeta Molagic
- Faculty of Medicine, University of Medicine and Pharmacy ‘Carol Davila’, No 37, Dionisie Lupu, 020021 Bucharest, Romania; (N.M.); (O.-A.G.); (V.M.); (V.A.)
- National Institute of Infectious Diseases ‘Matei Bals’, 1 Dr. Calistrat Grozovici, 021105 Bucharest, Romania; (A.-I.A.); (Ș.S.A.); (C.T.)
| | - Ștefan Sorin Aramă
- National Institute of Infectious Diseases ‘Matei Bals’, 1 Dr. Calistrat Grozovici, 021105 Bucharest, Romania; (A.-I.A.); (Ș.S.A.); (C.T.)
- Faculty of Dental Medicine, University of Medicine and Pharmacy ‘Carol Davila’, No 37, Dionisie Lupu, 020021 Bucharest, Romania
| | - Cătălin Tilișcan
- National Institute of Infectious Diseases ‘Matei Bals’, 1 Dr. Calistrat Grozovici, 021105 Bucharest, Romania; (A.-I.A.); (Ș.S.A.); (C.T.)
- Faculty of Dental Medicine, University of Medicine and Pharmacy ‘Carol Davila’, No 37, Dionisie Lupu, 020021 Bucharest, Romania
| | - Victoria Aramă
- Faculty of Medicine, University of Medicine and Pharmacy ‘Carol Davila’, No 37, Dionisie Lupu, 020021 Bucharest, Romania; (N.M.); (O.-A.G.); (V.M.); (V.A.)
- National Institute of Infectious Diseases ‘Matei Bals’, 1 Dr. Calistrat Grozovici, 021105 Bucharest, Romania; (A.-I.A.); (Ș.S.A.); (C.T.)
| |
Collapse
|
41
|
Yu Y, Lin K, Wu H, Hu M, Yang X, Wang J, Grillari J, Chen J. Targeting senescent cells in aging and COVID-19: from cellular mechanisms to therapeutic opportunities. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:20. [PMID: 39358480 PMCID: PMC11447201 DOI: 10.1186/s13619-024-00201-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/10/2024] [Indexed: 10/04/2024]
Abstract
The COVID-19 pandemic has caused a global health crisis and significant social economic burden. While most individuals experience mild or non-specific symptoms, elderly individuals are at a higher risk of developing severe symptoms and life-threatening complications. Exploring the key factors associated with clinical severity highlights that key characteristics of aging, such as cellular senescence, immune dysregulation, metabolic alterations, and impaired regenerative potential, contribute to disruption of tissue homeostasis of the lung and worse clinical outcome. Senolytic and senomorphic drugs, which are anti-aging treatments designed to eliminate senescent cells or decrease the associated phenotypes, have shown promise in alleviating age-related dysfunctions and offer a novel approach to treating diseases that share certain aspects of underlying mechanisms with aging, including COVID-19. This review summarizes the current understanding of aging in COVID-19 progression, and highlights recent findings on anti-aging drugs that could be repurposed for COVID-19 treatment to complement existing therapies.
Collapse
Affiliation(s)
- Yuan Yu
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kaixuan Lin
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Haoyu Wu
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Mingli Hu
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xuejie Yang
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jie Wang
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Johannes Grillari
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Molecular Biotechnology, BOKU University, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation With AUVA, 1200, Vienna, Austria
| | - Jiekai Chen
- Center for Cell Lineage and Atlas, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
42
|
Latarissa IR, Rendrayani F, Iftinan GN, Suhandi C, Meiliana A, Sormin IP, Barliana MI, Lestari K. The Efficacy of Oral/Intravenous Corticosteroid Use in COVID-19 Patients: A Systematic Review. J Exp Pharmacol 2024; 16:321-337. [PMID: 39371262 PMCID: PMC11453156 DOI: 10.2147/jep.s484596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024] Open
Abstract
The COVID-19 pandemic is prompting extensive investigation into potential treatments, including the use of corticosteroids to manage inflammation and mitigate severe disease outcomes. Therefore, this systematic review aimed to evaluate the efficacy of oral/intravenous corticosteroids in the management of COVID-19. A comprehensive search was conducted across major scientific databases such as MEDLINE, Scopus, and Cochrane for relevant studies published from 2019-2024. The inclusion criteria included studies investigating the use of oral/intravenous corticosteroids in COVID-19 patients >18 years with a randomized placebo-controlled trial method. Non-placebo-controlled studies, studies using combined treatments with other drugs, as well as protocol articles, conference proceedings, review articles, and non-English studies were excluded. A narrative synthesis approach was adopted given the significant methodological diversity. The results showed that a total of 12 studies met the inclusion criteria covering the use of three drugs, including dexamethasone (three), hydrocortisone (two), and methylprednisolone (seven). The outcome parameters used for each study were different. Among the total 12 studies, five showed insignificant results for hydrocortisone (two) and methylprednisolone (three), while others reported significant results. This systematic review suggested that oral/intravenous corticosteroids might confer clinical benefits in the management of COVID-19, particularly in reducing mortality and severe disease outcomes. However, further investigation was needed to establish standardized protocols regarding dosage, duration, and safety considerations to optimize efficacy and minimize potential adverse effects.
Collapse
Affiliation(s)
- Irma Rahayu Latarissa
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
| | - Farida Rendrayani
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Ghina Nadhifah Iftinan
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
| | - Cecep Suhandi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Anna Meiliana
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Prodia Clinical Laboratory, Central Jakarta, Indonesia
| | - Ida Paulina Sormin
- Faculty of Pharmacy, University of 17 August 1945 Jakarta, Jakarta, Indonesia
- Prodia Diacro Laboratories, Jakarta, Indonesia
| | - Melisa Intan Barliana
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, Indonesia
| | - Keri Lestari
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Medication Therapy Adherence Clinic (MTAC), Universitas Padjadjaran, Sumedang, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, Indonesia
| |
Collapse
|
43
|
Ibáñez-Prada ED, Guerrero JL, Bustos IG, León L, Fuentes YV, Santamaría-Torres M, Restrepo-Martínez JM, Serrano-Mayorga CC, Mendez L, Gomez-Duque S, Santacruz CA, Conway-Morris A, Martín-Loeches I, Gonzalez-Juarbe N, Cala MP, Reyes LF. The unique metabolic and lipid profiles of patients with severe COVID-19 compared to severe community-acquired pneumonia: a potential prognostic and therapeutic target. Expert Rev Respir Med 2024; 18:815-829. [PMID: 39327745 DOI: 10.1080/17476348.2024.2409264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Compare the changes and differences in metabolome and lipidome profiles among severe COVID-19 and CAP patients with ARF to identify biomarkers that could be used for personalized diagnosis, prognosis, and treatment. RESEARCH DESIGN AND METHODS Plasma samples were taken at hospital admission (baseline) and on the 5th day of hospitalization (follow-up) and examined by RP-LC-QTOF-MS and HILIC-LC-QTOF-MS. RESULTS 127 patients, 17 with CAP and 110 with COVID-19, were included. The analysis revealed 87 altered metabolites, suggesting changes in the metabolism of arachidonic acid, glycerolipids, glycerophospholipids, linoleic acid, pyruvate, glycolysis, among others. Most of these metabolites are involved in inflammatory, hypoxic, and thrombotic processes. At baseline, the greatest differences were found in phosphatidylcholine (PC) 31:4 (p < 0.001), phosphoserine (PS) 34:3 (p < 0.001), and phosphatidylcholine (PC) 36:5 (p < 0.001), all of which were notably decreased in COVID-19 patients. At follow-up, the most dysregulated metabolites were monomethyl-phosphatidylethanolamine (PE-Nme) 40:5 (p < 0.001) and phosphatidylcholine (PC) 38:4 (p < 0.001). CONCLUSIONS Metabolic and lipidic alterations suggest inhibition of innate anti-inflammatory and anti-thrombotic mechanisms in COVID-19 patients, which might lead to increased viral proliferation, uncontrolled inflammation, and thrombi formation. Results provide novel targets for predictive biomarkers against CAP and COVID-19. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Elsa D Ibáñez-Prada
- Unisabana Center for Translational Science, Universidad de La Sabana Chía, Colombia
- Clínica Universidad de La Sabana Chía, Colombia
| | - Jose L Guerrero
- MetCore-Metabolomics Core Facility, Vice-Presidency of Research and Knowledge Creation, Universidad de Los Andes, Bogotá, Colombia
| | - Ingrid G Bustos
- Unisabana Center for Translational Science, Universidad de La Sabana Chía, Colombia
| | - Lizeth León
- MetCore-Metabolomics Core Facility, Vice-Presidency of Research and Knowledge Creation, Universidad de Los Andes, Bogotá, Colombia
| | - Yuli V Fuentes
- Unisabana Center for Translational Science, Universidad de La Sabana Chía, Colombia
| | - Mary Santamaría-Torres
- MetCore-Metabolomics Core Facility, Vice-Presidency of Research and Knowledge Creation, Universidad de Los Andes, Bogotá, Colombia
| | | | | | - Lina Mendez
- Clínica Universidad de La Sabana Chía, Colombia
| | - Salome Gomez-Duque
- Unisabana Center for Translational Science, Universidad de La Sabana Chía, Colombia
| | - Carlos A Santacruz
- Fundación Santa Fe de Bogotá, Bogotá, Colombia
- Critical Care Department, Instituto de Ensino e Pesquisa do Pará, Brasil - IEPPA, Brazil
| | - Andrew Conway-Morris
- Division of Anesthesia, Department of Medicine, University of Cambridge, Cambridge, UK
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ignacio Martín-Loeches
- Department of Clinical Medicine, St James's Hospital, Multidisciplinary Intensive Care Research Organization (MICRO), Dublin, Ireland
| | | | - Mónica P Cala
- MetCore-Metabolomics Core Facility, Vice-Presidency of Research and Knowledge Creation, Universidad de Los Andes, Bogotá, Colombia
| | - Luis Felipe Reyes
- Unisabana Center for Translational Science, Universidad de La Sabana Chía, Colombia
- Clínica Universidad de La Sabana Chía, Colombia
- Pandemic Sciences Institute, University of Oxford, Oxford, UK
| |
Collapse
|
44
|
Fujieda K, Saito S, Tanaka A, Furuhashi K, Yasuda Y, Sano Y, Kato M, Maruyama S. A case of late-onset organizing pneumonia following COVID-19 infection in a post-kidney transplant patient. CEN Case Rep 2024; 13:346-350. [PMID: 38367183 PMCID: PMC11442844 DOI: 10.1007/s13730-023-00849-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/26/2023] [Indexed: 02/19/2024] Open
Abstract
A 50-year-old man who had undergone a living-donor kidney transplant 12 years prior for chronic renal failure due to autosomal dominant polycystic kidney disease contracted coronavirus disease 19 (COVID-19). He had a positive antigen test, mild symptoms, sore throat, and fever of 37.9 ℃. The patient was treated with molnupiravir for 5 days, and the symptoms disappeared 5 days after onset. However, 10 days after onset, he developed a fever of approximately 37 ℃ and a non-productive cough; 27 days after onset, the patient was hospitalized for anorexia and a worsening respiratory condition. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antigen test results on admission were negative, and no antiviral medications were administered against SARS-CoV-2. Computed tomography revealed extensive ground-glass opacities in both lung fields. The patient was treated with steroid pulse therapy, ceftriaxone, atovaquone, azithromycin, and respiratory management using a high-flow nasal cannula. The combined therapies were successful, and the patient was managed with a nasal oxygen cannula after 3 days. Oxygen administration was discontinued after 6 days of hospitalization, and the patient was discharged after 14 days. Based on the laboratory findings, bacterial, interstitial, and Pneumocystis pneumonia were unlikely. The success of the steroid pulse therapy suggested that respiratory failure was caused by pneumonia due to the immune response after COVID-19 infection.
Collapse
Affiliation(s)
- Kumiko Fujieda
- Department of Nephrology, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Shoji Saito
- Department of Nephrology, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Akihito Tanaka
- Department of Nephrology, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Kazuhiro Furuhashi
- Department of Nephrology, Nagoya University Hospital, Nagoya, Aichi, Japan.
| | - Yosinari Yasuda
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Yuta Sano
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masashi Kato
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| |
Collapse
|
45
|
Tandon A, Baral B, Saini V, Kandpal M, Dixit AK, Parmar HS, Meena AK, Chandra Jha H. The role of Helicobacter pylori in augmenting the severity of SARS-CoV-2 related gastrointestinal symptoms: An insight from molecular mechanism of co-infection. Heliyon 2024; 10:e37585. [PMID: 39364240 PMCID: PMC11447314 DOI: 10.1016/j.heliyon.2024.e37585] [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: 05/22/2024] [Revised: 08/24/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024] Open
Abstract
Coinfection of pathogenic bacteria and viruses is associated with multiple diseases. During the COVID-19 pandemic, the co-infection of other pathogens with SARS-CoV-2 was one of the important determinants of the severity. Although primarily a respiratory virus gastric manifestation of the SARS-CoV-2 infection was widely reported. This study highlights the possible consequences of SARS-CoV-2 -Helicobacter pylori coinfection in the gastrointestinal cells. We utilized the transfection and infection model for SARS-CoV-2 spike Delta (δ) and H. pylori respectively in colon carcinoma cell line HT-29 to develop the coinfection model to study inflammation, mitochondrial function, and cell death. The results demonstrate increased transcript levels of inflammatory markers like TLR2 (p < 0.01), IL10 (p < 0.05), TNFα (p < 0.05) and CXCL1 (p < 0.05) in pre-H. pylori infected cells as compared to the control. The protein levels of the β-Catenin (p < 0.01) and c-Myc (p < 0.01) were also significantly elevated in pre-H. pylori infected group in case of co-infection. Further investigation of apoptotic and necrotic markers (Caspase-3, Caspase-8, and RIP-1) reveals a necroptotic cell death in the coinfected cells. The infection and coinfection also damage the mitochondria in HT-29 cells, further implicating mitochondrial dysfunction in the necrotic cell death process. Our study also highlights the detrimental effect of pre-H. pylori exposure in the coinfection model compared to post-exposure and lone infection of H. pylori and SARS-CoV-2. This knowledge could aid in developing targeted interventions and therapeutic strategies to mitigate the severity of COVID-19 and improve patient outcomes.
Collapse
Affiliation(s)
- Akrati Tandon
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Budhadev Baral
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Vaishali Saini
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Meenakshi Kandpal
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| | - Amit Kumar Dixit
- Central Ayurveda Research Institute, Kolkata, 4-CN Block, Sector –V, Bidhannagar, Kolkata, 700 091, India
| | - Hamendra Singh Parmar
- School of Biotechnology, Devi Ahilya Vishwavidyalaya, Takshashila Campus, Indore, Madhya Pradesh, 452001, India
| | - Ajay Kumar Meena
- Regional Ayurveda Research Institute, Amkhoh, Gwalior, Madhya Pradesh, 474001, India
| | - Hem Chandra Jha
- Infection Bioengineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
- Centre for Rural Development and Technology, Indian Institute of Technology Indore, Simrol, Indore, Madhya Pradesh, 453552, India
| |
Collapse
|
46
|
Shen Z, Yin Z, Wei S, Cong Z, Zhao F, Zhang H, Zhu X. LIPS and PaO 2/FiO 2 Combined Plasma Biomarkers Predict Onset of Acute Respiratory Distress Syndrome in Patients of High Risks in SICU: A Prospective Exploratory Study. Mediators Inflamm 2024; 2024:4936265. [PMID: 39319361 PMCID: PMC11421942 DOI: 10.1155/2024/4936265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/10/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024] Open
Abstract
Objective To explore and validate the value of clinical parameters combined with plasma biomarkers for predicting acute respiratory distress syndrome (ARDS) in patients of high risks in the surgical intensive care unit (SICU). Materials and Methods We conducted a prospective, observational study from January 2020 to December 2023, which enrolled 263 patients of high risks in the SICU of Peking University Third Hospital consecutively; they were classified as ARDS and non-ARDS according to whether ARDS occurred after enrollment. Collected clinical characteristics and blood samples within 24 hr of admission to SICU. Blood samples from the first day to the seventh day of SICU were collected from patients without ARDS, and patients with ARDS were collected until 1 day after ARDS onset, forming data based on time series. ELISA and CBA were used to measure plasma biomarkers. Endpoint of the study was the onset of ARDS. Cox proportional hazard regression analysis was used to find independent risk factors of the onset of ARDS, then constructed a nomogram and tested its goodness-of-fit. Results About 84 of 263 patients ended with ARDS. Univariate analysis found 15 risk factors showed differences between ARDS and non-ARDS, namely, interleukin 6, interleukin 8 (IL-8), angiopoietin Ⅱ, LIPS, APACHEⅡ, SOFA, PaO2/FiO2, age, sex, shock, sepsis, acute abdomen, pulmonary contusion, pneumonia, hepatic dysfunction. We included factors with p < 0.2 in multivariate analysis and showed LIPS, PaO2/FiO2, IL-8, and receptor for advanced glycation end-products (RAGE) of the first day were independent risk factors for ARDS in SICU, a model combining them was good in predicting ARDS (C-index was 0.864 in total patients of high risks). The median of the C-index was 0.865, showed by fivefold cross-validation in the train cohort or validation cohort. The calibration curve shows an agreement between the probability of predicting ARDS and the actual probability of occurrence. Decision curve analysis indicated that the model had clinical use value. We constructed a nomogram that had the ability to predict ARDS in patients of high risks in SICU. Conclusions LIPS, PaO2/FiO2, plasma IL-8, and RAGE of the first day were independent risk factors of the onset of ARDS. The predictive ability for ARDS can be greatly improved when combining clinical parameters and plasma biomarkers.
Collapse
Affiliation(s)
- Ziyuan Shen
- Department of Critical Care MedicinePeking University Third Hospital, Beijing 100191, China
| | - Zhongnan Yin
- BiobankPeking University Third Hospital, Beijing 100191, China
| | - Senhao Wei
- Department of Critical Care MedicinePeking University Third Hospital, Beijing 100191, China
| | - Zhukai Cong
- Department of Critical Care MedicinePeking University Third Hospital, Beijing 100191, China
| | - Feng Zhao
- Department of Critical Care MedicinePeking University Third Hospital, Beijing 100191, China
| | - Hua Zhang
- Center of EpidemiologyPeking University Third Hospital, Beijing 100191, China
| | - Xi Zhu
- Department of Critical Care MedicinePeking University Third Hospital, Beijing 100191, China
| |
Collapse
|
47
|
Karim A, Pathan AM, Warayo A, Ahmed I, Betanai A, Dropati FNU, Ahmed S, Sarhandi PR, Kumari P, Bansari RB, Mehmoodi A, Malik J. Intranasal Insulin for Treatment of Persistent Post-COVID-19 Olfactory Dysfunction: A Scoping Review. J Community Hosp Intern Med Perspect 2024; 14:55-62. [PMID: 39399204 PMCID: PMC11466328 DOI: 10.55729/2000-9666.1390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/02/2024] [Accepted: 06/18/2024] [Indexed: 10/15/2024] Open
Abstract
Olfactory dysfunction has emerged as a prominent symptom of COVID-19, persisting in a subset of patients even after recovery. This scoping review aims to explore the potential of intranasal insulin as a treatment modality for persistent post-COVID-19 olfactory dysfunction. A comprehensive literature search was conducted to gather relevant studies examining the role of intranasal insulin in treating olfactory dysfunction, particularly in post-COVID-19 cases. Studies were included investigating intranasal insulin's mechanisms, efficacy, safety, and clinical outcomes. The review synthesizes findings from various studies suggesting the therapeutic potential of intranasal insulin in improving olfactory function. Research highlights the influence of intranasal insulin on neuroprotection, neurogenesis, and synaptic plasticity within the olfactory system, providing insights into its mechanisms of action. Furthermore, preliminary clinical evidence suggests improvements in olfactory sensitivity and intensity following intranasal insulin administration in post-COVID-19 patients with persistent olfactory dysfunction. While initial findings are encouraging, further rigorous investigations, including clinical trials with larger cohorts, are essential to validate these observations, ascertain optimal dosage regimens, and establish the safety and efficacy of intranasal insulin. This review provides a foundation for future research directions aimed at harnessing the therapeutic potential of intranasal insulin in addressing olfactory dysfunction following COVID-19.
Collapse
Affiliation(s)
- Ali Karim
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Arisha M.G. Pathan
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Allah Warayo
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Iftikhar Ahmed
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Aminullah Betanai
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - FNU Dropati
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Saqlain Ahmed
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Pir R.J. Sarhandi
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Poonam Kumari
- Department of Medicine, Liaquat University of Medical and Health Sciences, Jamshoro,
Pakistan
| | - Raveena B. Bansari
- epartment of Medicine, Peoples University Medical and Health Sciences for Women, Nawabshah,
Pakistan
| | - Amin Mehmoodi
- Department of Medicine, Ibn e Seena Hospital, Kabul,
Afghanistan
| | - Jahanzeb Malik
- Department of Cardiovascular Medicine, Cardiovascular Analytics Group, Islamabad,
Pakistan
| |
Collapse
|
48
|
Yu J, Zhang Y, Ye Z, Tang K, Ma Y, Fu L, Cui T, Kang H, Yuan Y, Pan W. A Multi-Machine Learning Consensus Model Based on Clinical Features Reveals That Interleukin-10 Derived from Monocytes Leads to a Poor Prognosis in Patients with Coronavirus Disease-2019. J Inflamm Res 2024; 17:5923-5942. [PMID: 39247837 PMCID: PMC11378990 DOI: 10.2147/jir.s472099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024] Open
Abstract
Background Despite ongoing interventions, SARS-CoV-2 continues to cause significant global morbidity and mortality. Early diagnosis and intervention are crucial for effective clinical management. However, prognostic features based on transcriptional data have shown limited effectiveness, highlighting the need for more precise biomarkers to improve COVID-19 treatment outcomes. Methods We retrospectively analyzed 149 clinical features from 189 COVID-19 patients, identifying prognostic features via univariate Cox regression. The cohort was split into training and validation sets, and 77 prognostic models were developed using seven machine learning algorithms. Among these, the least absolute shrinkage and selection operator (Lasso) method was employed to refine the selection of prognostic variables by ten-fold cross-validation strategy, which were then integrated with random survival forests (RSF) to build a robust COVID-19-related prognostic model (CRM). Model accuracy was evaluated across training, validation, and entire cohorts. The diagnostic relevance of interleukin-10 (IL-10) was confirmed in bulk transcriptional data and validated at the single-cell level, where we also examined changes in cellular communication between mononuclear cells with differing IL-10 expression and other immune cells. Results Univariate Cox regression identified 43 prognostic features. Among the 77 machine learning models, the combination of Lasso and RSF produced the most robust CRM. This model consistently performed well across training, validation, and entire cohorts. IL-10 emerged as a key prognostic feature within the CRM, validated by single-cell transcriptional data. Transcriptome analysis confirmed the stable diagnostic value of IL-10, with mononuclear cells identified as the primary IL-10 source. Moreover, differential IL-10 expression in these cells was linked to altered cellular communication in the COVID-19 immune microenvironment. Conclusion The CRM provides accurate prognostic predictions for COVID-19 patients. Additionally, the study underscores the importance of early IL-10 level testing upon hospital admission, which could inform therapeutic strategies.
Collapse
Affiliation(s)
- Jing Yu
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yike Zhang
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Zhixiong Ye
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Kun Tang
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yiming Ma
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Linlin Fu
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Tongtong Cui
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Hening Kang
- College of Basic Medicine, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Yadong Yuan
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Wensen Pan
- Second Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| |
Collapse
|
49
|
Mattos Pereira V, Thakar A, Nair S. Targeting iRhom2/ADAM17 attenuates COVID-19-induced cytokine release from cultured lung epithelial cells. Biochem Biophys Rep 2024; 39:101811. [PMID: 39253056 PMCID: PMC11382212 DOI: 10.1016/j.bbrep.2024.101811] [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: 11/16/2023] [Revised: 06/28/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, continues to pose a significant global health challenge, with acute respiratory distress syndrome (ARDS) being a major cause of mortality. Excessive cytokine release (cytokine storm) has been causally related to COVID-19-associated ARDS. While TNF-α inhibitors have shown potential in reducing inflammation, their broad effects on TNF-α signaling, including both pro- and anti-inflammatory pathways, present significant challenges and side effects in clinical use. Therefore, more precise therapeutic targets are urgently needed. ADAM17 is a key enzyme driving cytokine release, but its broad presence complicates direct inhibition. Targeting iRhom2, a regulator specific to immune cells that controls ADAM17's activity, offers a more focused and effective approach to reducing cytokine release. In this study, we hypothesized that targeted inhibition of ADAM-17/iRhom2 attenuates COVID-19-induced cytokine release in cultured lung epithelial cells. Human primary bronchial/tracheal epithelial cells challenged with COVID-19 pseudo-viral particles resulted in elevated cytokine release, which was attenuated following siRNA-mediated silencing of ADAM17 and iRhom2. Targeting ADAM-17/iRhom2 pathway may thus represent a strategy to overcome the COVID-19-associated ARDS.
Collapse
Affiliation(s)
- Vitoria Mattos Pereira
- University of Wyoming, School of Pharmacy and the Biomedical Sciences Interdisciplinary Graduate Program, Laramie, WY, 82071, USA
| | - Amit Thakar
- University of Wyoming, School of Pharmacy and the Biomedical Sciences Interdisciplinary Graduate Program, Laramie, WY, 82071, USA
| | - Sreejayan Nair
- University of Wyoming, School of Pharmacy and the Biomedical Sciences Interdisciplinary Graduate Program, Laramie, WY, 82071, USA
| |
Collapse
|
50
|
Chikopela T, Mwesigwa N, Masenga SK, Kirabo A, Shibao CA. The Interplay of HIV and Long COVID in Sub-Saharan Africa: Mechanisms of Endothelial Dysfunction. Curr Cardiol Rep 2024; 26:859-871. [PMID: 38958890 DOI: 10.1007/s11886-024-02087-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW Long COVID affects approximately 5 million people in Africa. This disease is characterized by persistent symptoms or new onset of symptoms after an acute SARS-CoV-2 infection. Specifically, the most common symptoms include a range of cardiovascular problems such as chest pain, orthostatic intolerance, tachycardia, syncope, and uncontrolled hypertension. Importantly, these conditions appear to have endothelial dysfunction as the common denominator, which is often due to impaired nitric oxide (NO) mechanisms. This review discusses the role of mechanisms contributing to endothelial dysfunction in Long COVID, particularly in people living with HIV. RECENT FINDINGS Recent studies have reported that increased inflammation and oxidative stress, frequently observed in Long COVID, may contribute to NO dysfunction, ultimately leading to decreased vascular reactivity. These mechanisms have also been reported in people living with HIV. In regions like Africa, where HIV infection is still a major public health challenge with a prevalence of approximately 26 million people in 2022. Specifically, endothelial dysfunction has been reported as a major mechanism that appears to contribute to cardiovascular diseases and the intersection with Long COVID mechanisms is of particular concern. Further, it is well established that this population is more likely to develop Long COVID following infection with SARS-CoV-2. Therefore, concomitant infection with SARS-CoV-2 may lead to accelerated cardiovascular disease. We outline the details of the worsening health problems caused by Long COVID, which exacerbate pre-existing conditions such as endothelial dysfunction. The overlapping mechanisms of HIV and SARS-CoV-2, particularly the prolonged inflammatory response and chronic hypoxia, may increase susceptibility to Long COVID. Addressing these overlapping health issues is critical as it provides clinical entry points for interventions that could improve and enhance outcomes and quality of life for those affected by both HIV and Long COVID in the region.
Collapse
Affiliation(s)
- Theresa Chikopela
- Department of Human Physiology, Faculty of Medicine, Lusaka Apex Medical University, Lusaka, Zambia
| | - Naome Mwesigwa
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA
| | - Sepiso K Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone, Zambia
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA
| | - Cyndya A Shibao
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA.
| |
Collapse
|