1
|
Costanza A, Amerio A, Aguglia A, Rossi M, Parise A, Magnani L, Serafini G, Amore M, Martins D, Nguyen KD. Reactive Astrocytosis-A Potential Contributor to Increased Suicide in Long COVID-19 Patients? Brain Sci 2024; 14:973. [PMID: 39451987 PMCID: PMC11505806 DOI: 10.3390/brainsci14100973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Long COVID-19 is an emerging chronic illness of significant public health concern due to a myriad of neuropsychiatric sequelae, including increased suicidal ideation (SI) and behavior (SB). METHODS This review provides a concise synthesis of clinical evidence that points toward the dysfunction of astrocytes, the most abundant glial cell type in the central nervous system, as a potential shared pathology between SI/SB and COVID-19. RESULTS Depression, a suicide risk factor, and SI/SB were both associated with reduced frequencies of various astrocyte subsets and complex proteomic/transcriptional changes of astrocyte-related markers in a brain-region-specific manner. Astrocyte-related circulating markers were increased in depressed subjects and, to a less consistent extent, in COVID-19 patients. Furthermore, reactive astrocytosis was observed in subjects with SI/SB and those with COVID-19. CONCLUSIONS Astrocyte dysfunctions occurred in depression, SI/SB, and COVID-19. Reactive-astrocyte-mediated loss of the blood-brain barrier (BBB) integrity and subsequent neuroinflammation-a factor previously linked to SI/SB development-might contribute to increased suicide in individuals with long COVID-19. As such, the formulation of new therapeutic strategies to restore astrocyte homeostasis, enhance BBB integrity, and mitigate neuroinflammation may reduce SI/SB-associated neuropsychiatric manifestations among long COVID-19 patients.
Collapse
Affiliation(s)
- Alessandra Costanza
- Department of Psychiatry, Faculty of Medicine, University of Geneva (UNIGE), 24 Rue du Général-Dufour, 1211 Geneva, Switzerland
- Department of Psychiatry, Faculty of Biomedical Sciences, University of Italian Switzerland (USI), Via Giuseppe Buffi 13, 6900 Lugano, Switzerland
- Department of Psychiatry, Adult Psychiatry Service, University Hospitals of Geneva (HUG), Rue Gabrielle-Perret-Gentil 4, 1205 Geneva, Switzerland
- “Nel Chiostro”, Medical and Study Center, Via Camillo Leone 29, 13100 Vercelli, Italy
| | - Andrea Amerio
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Via Balbi, 5, 16132 Genoa, Italy; (A.A.); (A.A.); (G.S.); (M.A.)
- IRCCS Polyclinic Hospital San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Andrea Aguglia
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Via Balbi, 5, 16132 Genoa, Italy; (A.A.); (A.A.); (G.S.); (M.A.)
- IRCCS Polyclinic Hospital San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Martina Rossi
- “Nel Chiostro”, Medical and Study Center, Via Camillo Leone 29, 13100 Vercelli, Italy
| | - Alberto Parise
- Geriatric-Rehabilitation Department, University Hospital of Parma, 43126 Parma, Italy;
| | - Luca Magnani
- Department of Psychiatry, San Maurizio Hospital of Bolzano, Via Lorenz Böhler, 5, 39100 Bolzano, Italy;
| | - Gianluca Serafini
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Via Balbi, 5, 16132 Genoa, Italy; (A.A.); (A.A.); (G.S.); (M.A.)
- IRCCS Polyclinic Hospital San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Mario Amore
- Section of Psychiatry, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Via Balbi, 5, 16132 Genoa, Italy; (A.A.); (A.A.); (G.S.); (M.A.)
- IRCCS Polyclinic Hospital San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN)—King’s College London (KCL), Strand Campus, London WC2R 2LS, UK;
- NIHR Maudesley BRC, 16 De Crespigny Park, SE5 8AF South London and Maudesley NHS Trust, Denmark Hill, London SE5 8AZ, UK
| | - Khoa D. Nguyen
- Program in Immunology, Stanford University, 450 Jane Stanford Way, Stanford, CA 94305, USA;
- Department of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
2
|
Velasco M, Posada-Ayala M, Pérez-Fernández E, Loria F, Amores M, Ramos JM, Jaime E, Guijarro C, Romero J, Pazos MR. Circulating endocannabinoid levels in SARS-CoV-2 infection and their potential role in the inflammatory response. Sci Rep 2024; 14:19558. [PMID: 39174572 PMCID: PMC11341693 DOI: 10.1038/s41598-024-70172-5] [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: 04/30/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
Plasma levels of endocannabinoids (eCBs) are very dynamic and variable in different circumstances and pathologies. The aim of the study was to determine the levels of the main eCBs and N-acylethanolamines (NAEs) in COVID-19 patients during the acute and post-acute phase of SARS-CoV-2 infection. Samples collected before December 31, 2020 were used for the determination of circulating eCB levels by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The association between plasma eCB measurements and biochemical and hematological parameters, as well as serum IL-6 levels, was evaluated. Samples of 64 individuals were analysed, n = 18 healthy donors, n = 30 acute, and n = 16 post-acute patients. Plasma levels of 2-arachidonoylglycerol (2-AG), were significantly elevated in COVID-19 patients when compared to healthy individuals. Plasma N-palmitoylethanolamide (PEA) and N-arachidonoylethanolamide (AEA) levels were found to be decreased in post-acute patient samples. These results suggest that 2-AG plays an important role in the inflammatory cascade in COVID-19 disease; in addition, eCBs might be involved in the post-acute pathogenesis of COVID-19. This study provides evidence of altered levels of circulating eCBs as a consequence of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Maria Velasco
- Unidad de Enfermedades Infecciosas, Hospital Universitario Fundación Alcorcón, 28922, Alcorcón, Madrid, Spain.
- Departamento de Especialidades Médicas y Salud Pública, Universidad Rey Juan Carlos, 28922, Alcorcón, Madrid, Spain.
- Unidad de Investigación, Hospital Universitario Fundación Alcorcón, 28922, Alcorcón, Madrid, Spain.
| | - Maria Posada-Ayala
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, 28223, Pozuelo de Alarcón, Madrid, Spain
| | - Elia Pérez-Fernández
- Unidad de Investigación, Hospital Universitario Fundación Alcorcón, 28922, Alcorcón, Madrid, Spain
| | - Frida Loria
- Unidad de Investigación, Hospital Universitario Fundación Alcorcón, 28922, Alcorcón, Madrid, Spain
| | - Mario Amores
- Unidad de Investigación, Hospital Universitario Fundación Alcorcón, 28922, Alcorcón, Madrid, Spain
| | - José Miguel Ramos
- Unidad de Medicina Interna, Hospital Universitario Fundación Alcorcón, 28922, Alcorcón, Madrid, Spain
| | - Elena Jaime
- Servicio de Análisis Clínicos, Hospital Universitario Fundación Alcorcón, 28922, Alcorcón, Madrid, Spain
| | - Carlos Guijarro
- Departamento de Especialidades Médicas y Salud Pública, Universidad Rey Juan Carlos, 28922, Alcorcón, Madrid, Spain
- Unidad de Medicina Interna, Hospital Universitario Fundación Alcorcón, 28922, Alcorcón, Madrid, Spain
| | - Julián Romero
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, 28223, Pozuelo de Alarcón, Madrid, Spain
| | - Maria Ruth Pazos
- Unidad de Investigación, Hospital Universitario Fundación Alcorcón, 28922, Alcorcón, Madrid, Spain.
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, 28223, Pozuelo de Alarcón, Madrid, Spain.
| |
Collapse
|
3
|
Salvio AL, Fernandes RA, Ferreira HFA, Duarte LA, Gutman EG, Raposo-Vedovi JV, Filho CHFR, da Costa Nunes Pimentel Coelho WL, Passos GF, Andraus MEC, da Costa Gonçalves JP, Cavalcanti MG, Amaro MP, Kader R, de Andrade Medronho R, Figueiredo CP, Amado-Leon LA, Alves-Leon SV. High Levels of NfL, GFAP, TAU, and UCH-L1 as Potential Predictor Biomarkers of Severity and Lethality in Acute COVID-19. Mol Neurobiol 2024; 61:3545-3558. [PMID: 37996731 PMCID: PMC11087339 DOI: 10.1007/s12035-023-03803-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023]
Abstract
Few studies showed that neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), total tubulin-associated unit (TAU), and ubiquitin carboxy-terminal hydrolase-L1 (UCH-L1) may be related to neurological manifestations and severity during and after SARS-CoV-2 infection. The objective of this work was to investigate the relationship among nervous system biomarkers (NfL, TAU, GFAP, and UCH-L1), biochemical parameters, and viral loads with heterogeneous outcomes in a cohort of severe COVID-19 patients admitted in Intensive Care Unit (ICU) of a university hospital. For that, 108 subjects were recruited within the first 5 days at ICU. In parallel, 16 mild COVID-19 patients were enrolled. Severe COVID-19 group was divided between "deceased" and "survivor." All subjects were positive for SARS-CoV-2 detection. NfL, total TAU, GFAP, and UCH-L1 quantification in plasma was performed using SIMOA SR-X platform. Of 108 severe patients, 36 (33.33%) presented neurological manifestation and 41 (37.96%) died. All four biomarkers - GFAP, NfL, TAU, and UCH-L1 - were significantly higher among deceased patients in comparison to survivors (p < 0.05). Analyzing biochemical biomarkers, higher Peak Serum Ferritin, D-Dimer Peak, Gamma-glutamyltransferase, and C-Reactive Protein levels were related to death (p < 0.0001). In multivariate analysis, GFAP, NfL, TAU, UCH-L1, and Peak Serum Ferritin levels were correlated to death. Regarding SARS-CoV-2 viral load, no statistical difference was observed for any group. Thus, Ferritin, NFL, GFAP, TAU, and UCH-L1 are early biomarkers of severity and lethality of SARS-COV-2 infection and may be important tools for therapeutic decision-making in the acute phase of disease.
Collapse
Affiliation(s)
- Andreza Lemos Salvio
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | - Renan Amphilophio Fernandes
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | - Helena França Alcaraz Ferreira
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | - Larissa Araujo Duarte
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | - Elisa Gouvea Gutman
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | - Jessica Vasques Raposo-Vedovi
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | | | | | | | - Maria Emília Cosenza Andraus
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
| | - João Paulo da Costa Gonçalves
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil
| | - Marta Guimarães Cavalcanti
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
- Epidemiology and Evaluation Service, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
| | - Marisa Pimentel Amaro
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
- School of Medicine, Post-Graduate Program in Infectious and Parasitic Diseases, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
| | - Rafael Kader
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
- School of Medicine, Post-Graduate Program in Infectious and Parasitic Diseases, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
| | - Roberto de Andrade Medronho
- Epidemiology and Evaluation Service, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil
| | | | - Luciane Almeida Amado-Leon
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil.
| | - Soniza Vieira Alves-Leon
- Laboratory of Translacional Neurosciences, Biomedical Institute, Federal University of the State of Rio de Janeiro-UNIRIO, Rio de Janeiro, 22290-240, Brazil.
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, 21941-901, Brazil.
| |
Collapse
|
4
|
Pilger BI, Castro A, Vasconcellos FF, Moura KF, Signini ÉDF, Marqueze LFB, Fiorenza-Neto EA, Rocha MT, Pedroso GS, Cavaglieri CR, Ferreira AG, Figueiredo C, Minuzzi LG, Gatti da Silva GH, Castro GS, Lira FS, Seelaender M, Pinho RA. Obesity-dependent molecular alterations in fatal COVID-19: A retrospective postmortem study of metabolomic profile of adipose tissue. J Cell Biochem 2024; 125:e30566. [PMID: 38591648 DOI: 10.1002/jcb.30566] [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: 01/26/2024] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/10/2024]
Abstract
We investigated the effects of obesity on metabolic, inflammatory, and oxidative stress parameters in the adipose tissue of patients with fatal COVID-19. Postmortem biopsies of subcutaneous adipose tissue were obtained from 25 unvaccinated inpatients who passed from COVID-19, stratified as nonobese (N-OB; body mass index [BMI], 26.5 ± 2.3 kg m-2) or obese (OB BMI 34.2 ± 5.1 kg m-2). Univariate and multivariate analyses revealed that body composition was responsible for most of the variations detected in the metabolome, with greater dispersion observed in the OB group. Fifteen metabolites were major segregation factors. Results from the OB group showed higher levels of creatinine, myo-inositol, O-acetylcholine, and succinate, and lower levels of sarcosine. The N-OB group showed lower levels of glutathione peroxidase activity, as well as higher content of IL-6 and adiponectin. We revealed significant changes in the metabolomic profile of the adipose tissue in fatal COVID-19 cases, with high adiposity playing a key role in these observed variations. These findings highlight the potential involvement of metabolic and inflammatory pathways, possibly dependent on hypoxia, shedding light on the impact of obesity on disease pathogenesis and suggesting avenues for further research and possible therapeutic targets.
Collapse
Affiliation(s)
- Bruna I Pilger
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Alex Castro
- Laboratory of Nuclear Magnetic Resonance, Department of Chemistry, Universidade Federal de São Carlos, São Carlos, Brazil
- Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Franciane F Vasconcellos
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Karen F Moura
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Étore De Favari Signini
- Cardiovascular Physical Therapy Laboratory, Department of Physical Therapy, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Luis Felipe B Marqueze
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Edson A Fiorenza-Neto
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Mateus T Rocha
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Giulia S Pedroso
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Claudia R Cavaglieri
- Exercise Physiology Laboratory, Faculty of Physical Education, University of Campinas, Campinas, Brazil
| | - Antonio G Ferreira
- Laboratory of Nuclear Magnetic Resonance, Department of Chemistry, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Caique Figueiredo
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Luciele G Minuzzi
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Guilherme H Gatti da Silva
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Gabriela S Castro
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Fábio S Lira
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista, Presidente Prudente, Brazil
| | - Marilia Seelaender
- Cancer Metabolism Research Group, Department of Surgery and LIM 26, Hospital das Clínicas, University of São Paulo, São Paulo, Brazil
| | - Ricardo A Pinho
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| |
Collapse
|
5
|
Li J, Wang Z, Li J, Zhao H, Ma Q. HMGB1: A New Target for Ischemic Stroke and Hemorrhagic Transformation. Transl Stroke Res 2024:10.1007/s12975-024-01258-5. [PMID: 38740617 DOI: 10.1007/s12975-024-01258-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 05/16/2024]
Abstract
Stroke in China is distinguished by its high rates of morbidity, recurrence, disability, and mortality. The ultra-early administration of rtPA is essential for restoring perfusion in acute ischemic stroke, though it concurrently elevates the risk of hemorrhagic transformation. High-mobility group box 1 (HMGB1) emerges as a pivotal player in neuroinflammation after brain ischemia and ischemia-reperfusion. Released passively by necrotic cells and actively secreted, including direct secretion of HMGB1 into the extracellular space and packaging of HMGB1 into intracellular vesicles by immune cells, glial cells, platelets, and endothelial cells, HMGB1 represents a prototypical damage-associated molecular pattern (DAMP). It is intricately involved in the pathogenesis of atherosclerosis, thromboembolism, and detrimental inflammation during the early phases of ischemic stroke. Moreover, HMGB1 significantly contributes to neurovascular remodeling and functional recovery in later stages. Significantly, HMGB1 mediates hemorrhagic transformation by facilitating neuroinflammation, directly compromising the integrity of the blood-brain barrier, and enhancing MMP9 secretion through its interaction with rtPA. As a systemic inflammatory factor, HMGB1 is also implicated in post-stroke depression and an elevated risk of stroke-associated pneumonia. The role of HMGB1 extends to influencing the pathogenesis of ischemia by polarizing various subtypes of immune and glial cells. This includes mediating excitotoxicity due to excitatory amino acids, autophagy, MMP9 release, NET formation, and autocrine trophic pathways. Given its multifaceted role, HMGB1 is recognized as a crucial therapeutic target and prognostic marker for ischemic stroke and hemorrhagic transformation. In this review, we summarize the structure and redox properties, secretion and pathways, regulation of immune cell activity, the role of pathophysiological mechanisms in stroke, and hemorrhage transformation for HMGB1, which will pave the way for developing new neuroprotective drugs, reduction of post-stroke neuroinflammation, and expansion of thrombolysis time window.
Collapse
Affiliation(s)
- Jiamin Li
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China
| | - Zixin Wang
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China
| | - Jiameng Li
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China
| | - Haiping Zhao
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China.
| | - Qingfeng Ma
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China.
| |
Collapse
|
6
|
Beck NS, Seo Y, Park T, Jun SS, Im JI, Hong SY. Oxidative stress in patients with coronavirus disease and end-stage renal disease: a pilot study. BMC Nephrol 2024; 25:155. [PMID: 38702607 PMCID: PMC11069245 DOI: 10.1186/s12882-024-03584-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Oxidative stress, an imbalance between reactive oxygen species production and antioxidant capacity, increases in patients with coronavirus disease (COVID-19) or renal impairment. We investigated whether combined COVID-19 and end-stage renal disease (ESRD) would increase oxidative stress levels compared to each disease alone. METHODS Oxidative stress was compared among three groups. Two groups comprised patients with COVID-19 referred to the hospital with or without renal impairment (COVID-ESRD group [n = 18]; COVID group [n = 17]). The third group (ESRD group [n = 18]) comprised patients without COVID-19 on maintenance hemodialysis at a hospital. RESULTS The total oxidative stress in the COVID-ESRD group was lower than in the COVID group (p = 0.047). The total antioxidant status was higher in the COVID-ESRD group than in the ESRD (p < 0.001) and COVID (p < 0.001) groups after controlling for covariates. The oxidative stress index was lower in the COVID-ESRD group than in the ESRD (p = 0.001) and COVID (p < 0.001) groups. However, the three oxidative parameters did not differ significantly between the COVID and COVID-ESRD groups. CONCLUSIONS The role of reactive oxygen species in the pathophysiology of COVID-19 among patients withESRD appears to be non-critical. Therefore, the provision of supplemental antioxidants may not confer a therapeutic advantage, particularly in cases of mild COVID-19 in ESRD patients receiving hemodialysis. Nonetheless, this area merits further research.
Collapse
Affiliation(s)
- Nam-Seon Beck
- Department of Pediatrics, Chung-Ang Jeil Hospital, Chungbuk, South Korea
| | - Yeonju Seo
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, South Korea
| | - Taesung Park
- Department of Statistics, Seoul National University, Seoul, South Korea
| | - Sang-Sin Jun
- Department of Neurology, Chung-Ang Jeil Hospital, Chungbuk, South Korea
| | - Joung-Il Im
- Department of Orthopedic Surgery, Chung-Ang Jeil Hospital, Chungbuk, South Korea
| | - Sae-Yong Hong
- Department of Nephrology, Chung-Ang Jeil Hospital, Chungbuk, South Korea.
| |
Collapse
|
7
|
Wulandari S, Nuryastuti T, Oktoviani FN, Daniwijaya MEW, Supriyati E, Arguni E, Hartono, Wibawa T. The association between high mobility group box 1 (HMGB1) and Interleukin-18 (IL-18) serum concentrations in COVID-19 inpatients. Heliyon 2024; 10:e26619. [PMID: 38434314 PMCID: PMC10907672 DOI: 10.1016/j.heliyon.2024.e26619] [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: 10/31/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 03/05/2024] Open
Abstract
Background High mobility group box 1 (HMGB1) and interleukin-18 (IL-18) are involved in various non-coronavirus disease pathogenesis and are reported as potential biomarkers for coronavirus disease (COVID-19). However, their association with COVID-19 pathogenesis has not yet been explored. Aim This study aimed to investigate the association between HMGB1 and IL-18 concentrations in the sera of COVID-19 patients versus non-COVID-19 patients. Material and methods We used stored serum samples obtained from 30 COVID-19 patients and 30 non-COVID-19 patients. We collected data on age, gender, treatment status, principal diagnosis, and comorbidity from patient medical records. HMGB1 and IL-18 concentrations were analyzed in the serum by enzyme-linked immunosorbent assay (ELISA). The swab samples' RT-PCR cycle threshold (CT) values were obtained from the laboratory database. Results HMGB1 concentrations were increased in the COVID-19 inpatients and non-COVID-19 inpatients compared to non-COVID-19 outpatients (COVID-19 inpatients vs. non-COVID-19 outpatients: 151.33 (90.27-192.38) vs. 80.75 (54.16-128.72) ng/ml; p = 0.0316; non-COVID-19 inpatients vs. non-COVID-19 outpatients: 152.66 (104.04-288.51) vs. 80.75 (54.16-128.72) ng/ml; p = 0.0199). IL-18 concentrations were also higher in the COVID-19 inpatients and non-COVID-19 inpatients compared to non-COVID-19 outpatients (COVID-19 inpatients vs. non-COVID-19 outpatients: 620.00 (461.50-849.6) vs. 403.10 (372.70-556.90) pg/ml; p = 0.0376; non-COVID-19 inpatients vs. non-COVID-19 outpatients: 835.70 (558.30-1602.00) vs. 403.10 (372.70-556.90) pg/ml; p = 0.0026). Moreover, HMGB1 was associated with IL-18 concentrations in the sera of COVID-19 inpatients (p = 0.0337; r = 0.5500). Conclusion The association of HMGB1 and IL-18 in COVID-19 might indicate the potential for a dangerous cycle leading to a cytokine storm to occur.
Collapse
Affiliation(s)
- Sri Wulandari
- Doctorate Program of Medicine and Health Science, Faculty of Medicine Public Health and Nursing Universitas Gadjah Mada, Yogyakarta, Indonesia
- Department of Physiology, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
| | - Titik Nuryastuti
- Department of Microbiology, Faculty of Medicine Public Health and Nursing Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Farida Nur Oktoviani
- Department of Microbiology, Faculty of Medicine Public Health and Nursing Universitas Gadjah Mada, Yogyakarta, Indonesia
| | | | - Endah Supriyati
- Centre for Tropical Medicine, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Eggi Arguni
- Department of Child Health, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Hartono
- Department of Physiology, Faculty of Medicine, Universitas Sebelas Maret, Surakarta, Indonesia
| | - Tri Wibawa
- Department of Microbiology, Faculty of Medicine Public Health and Nursing Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
8
|
Heil M. Self-DNA driven inflammation in COVID-19 and after mRNA-based vaccination: lessons for non-COVID-19 pathologies. Front Immunol 2024; 14:1259879. [PMID: 38439942 PMCID: PMC10910434 DOI: 10.3389/fimmu.2023.1259879] [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: 07/17/2023] [Accepted: 12/26/2023] [Indexed: 03/06/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic triggered an unprecedented concentration of economic and research efforts to generate knowledge at unequalled speed on deregulated interferon type I signalling and nuclear factor kappa light chain enhancer in B-cells (NF-κB)-driven interleukin (IL)-1β, IL-6, IL-18 secretion causing cytokine storms. The translation of the knowledge on how the resulting systemic inflammation can lead to life-threatening complications into novel treatments and vaccine technologies is underway. Nevertheless, previously existing knowledge on the role of cytoplasmatic or circulating self-DNA as a pro-inflammatory damage-associated molecular pattern (DAMP) was largely ignored. Pathologies reported 'de novo' for patients infected with Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV)-2 to be outcomes of self-DNA-driven inflammation in fact had been linked earlier to self-DNA in different contexts, e.g., the infection with Human Immunodeficiency Virus (HIV)-1, sterile inflammation, and autoimmune diseases. I highlight particularly how synergies with other DAMPs can render immunogenic properties to normally non-immunogenic extracellular self-DNA, and I discuss the shared features of the gp41 unit of the HIV-1 envelope protein and the SARS-CoV 2 Spike protein that enable HIV-1 and SARS-CoV-2 to interact with cell or nuclear membranes, trigger syncytia formation, inflict damage to their host's DNA, and trigger inflammation - likely for their own benefit. These similarities motivate speculations that similar mechanisms to those driven by gp41 can explain how inflammatory self-DNA contributes to some of most frequent adverse events after vaccination with the BNT162b2 mRNA (Pfizer/BioNTech) or the mRNA-1273 (Moderna) vaccine, i.e., myocarditis, herpes zoster, rheumatoid arthritis, autoimmune nephritis or hepatitis, new-onset systemic lupus erythematosus, and flare-ups of psoriasis or lupus. The hope is to motivate a wider application of the lessons learned from the experiences with COVID-19 and the new mRNA vaccines to combat future non-COVID-19 diseases.
Collapse
Affiliation(s)
- Martin Heil
- Departamento de Ingeniería Genética, Laboratorio de Ecología de Plantas, Centro de Investigación y de Estudios Avanzados (CINVESTAV)-Unidad Irapuato, Irapuato, Mexico
| |
Collapse
|
9
|
Katz AA, Wainwright S, Kelly MP, Albert P, Byrne R. Hyperbaric oxygen effectively addresses the pathophysiology of long COVID: clinical review. Front Med (Lausanne) 2024; 11:1354088. [PMID: 38449882 PMCID: PMC10916685 DOI: 10.3389/fmed.2024.1354088] [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: 12/11/2023] [Accepted: 01/31/2024] [Indexed: 03/08/2024] Open
Abstract
Background The World Health Organization defines long COVID as "the continuation or development of new symptoms 3 months after the initial SARS-CoV-2 infection, with these symptoms lasting for at least 2 months with no other explanation." Estimations of approximately 50 million individuals suffer from long COVID, reporting low health-related quality of life. Patients develop ongoing persistent symptoms that continue for more than 12 weeks that are not explained by another alternative diagnosis. To date, no current therapeutics are effective in treating the underlying pathophysiology of long COVID. Discussion A comprehensive literature search using PubMed and Google Scholar was conducted and all available articles from November 2021 to January 2024 containing keywords long covid and hyperbaric oxygen were reviewed. These published studies, including case series and randomized trials, demonstrate that utilizing Hyperbaric Oxygen Therapy (HBO) provided significant improvement in patients with long COVID. Conclusion A large cohort of patients suffer from long COVID or post-COVID-19 syndrome after recovery from their acute infection with no effective treatment options. HBO is a safe treatment and may provide benefit for this population and should continue to be researched for adjunctive treatment of long COVID.
Collapse
Affiliation(s)
- Alan A. Katz
- Hyperbaric Medical Solutions, New York, NY, United States
- Orlando College of Osteopathic Medicine, Winter Garden, FL, United States
| | - Sandra Wainwright
- Greenwich Hospital, Yale New Haven Health System, New Haven, CT, United States
| | - Matthew P. Kelly
- University of Alabama at Birmingham, Heersink School of Medicine, Birmingham, AL, United States
| | - Pradeep Albert
- Orlando College of Osteopathic Medicine, Winter Garden, FL, United States
| | - Rosemary Byrne
- Hyperbaric Medical Solutions, New York, NY, United States
| |
Collapse
|
10
|
Gavilán E, Medina-Guzman R, Bahatyrevich-Kharitonik B, Ruano D. Protein Quality Control Systems and ER Stress as Key Players in SARS-CoV-2-Induced Neurodegeneration. Cells 2024; 13:123. [PMID: 38247815 PMCID: PMC10814689 DOI: 10.3390/cells13020123] [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: 11/29/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
The COVID-19 pandemic has brought to the forefront the intricate relationship between SARS-CoV-2 and its impact on neurological complications, including potential links to neurodegenerative processes, characterized by a dysfunction of the protein quality control systems and ER stress. This review article explores the role of protein quality control systems, such as the Unfolded Protein Response (UPR), the Endoplasmic Reticulum-Associated Degradation (ERAD), the Ubiquitin-Proteasome System (UPS), autophagy and the molecular chaperones, in SARS-CoV-2 infection. Our hypothesis suggests that SARS-CoV-2 produces ER stress and exploits the protein quality control systems, leading to a disruption in proteostasis that cannot be solved by the host cell. This disruption culminates in cell death and may represent a link between SARS-CoV-2 and neurodegeneration.
Collapse
Affiliation(s)
- Elena Gavilán
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla (US), 41012 Sevilla, Spain; (R.M.-G.); (B.B.-K.); (D.R.)
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Junta de Andalucía, CSIC, University of Seville (US), 41013 Sevilla, Spain
| | - Rafael Medina-Guzman
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla (US), 41012 Sevilla, Spain; (R.M.-G.); (B.B.-K.); (D.R.)
| | - Bazhena Bahatyrevich-Kharitonik
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla (US), 41012 Sevilla, Spain; (R.M.-G.); (B.B.-K.); (D.R.)
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Junta de Andalucía, CSIC, University of Seville (US), 41013 Sevilla, Spain
| | - Diego Ruano
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla (US), 41012 Sevilla, Spain; (R.M.-G.); (B.B.-K.); (D.R.)
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Junta de Andalucía, CSIC, University of Seville (US), 41013 Sevilla, Spain
| |
Collapse
|
11
|
Yang X, Liu X, Nie Y, Zhan F, Zhu B. Oxidative stress and ROS-mediated cellular events in RSV infection: potential protective roles of antioxidants. Virol J 2023; 20:224. [PMID: 37798799 PMCID: PMC10557227 DOI: 10.1186/s12985-023-02194-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/27/2023] [Indexed: 10/07/2023] Open
Abstract
Respiratory syncytial virus (RSV), a member of the Pneumoviridae family, can cause severe acute lower respiratory tract infection in infants, young children, immunocompromised individuals and elderly people. RSV is associated with an augmented innate immune response, enhanced secretion of inflammatory cytokines, and necrosis of infected cells. Oxidative stress, which is mainly characterized as an imbalance in the production of reactive oxygen species (ROS) and antioxidant responses, interacts with all the pathophysiologic processes above and is receiving increasing attention in RSV infection. A gradual accumulation of evidence indicates that ROS overproduction plays an important role in the pathogenesis of severe RSV infection and serves as a major factor in pulmonary inflammation and tissue damage. Thus, antioxidants seem to be an effective treatment for severe RSV infection. This article mainly reviews the information on oxidative stress and ROS-mediated cellular events during RSV infection for the first time.
Collapse
Affiliation(s)
- Xue Yang
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, Hubei, China
| | - Xue Liu
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, Hubei, China
| | - Yujun Nie
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, Hubei, China
| | - Fei Zhan
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, Hubei, China
| | - Bin Zhu
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, Hubei, China.
| |
Collapse
|
12
|
Mrakic-Sposta S, Vezzoli A, Garetto G, Paganini M, Camporesi E, Giacon TA, Dellanoce C, Agrimi J, Bosco G. Hyperbaric Oxygen Therapy Counters Oxidative Stress/Inflammation-Driven Symptoms in Long COVID-19 Patients: Preliminary Outcomes. Metabolites 2023; 13:1032. [PMID: 37887357 PMCID: PMC10608857 DOI: 10.3390/metabo13101032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/15/2023] [Accepted: 09/22/2023] [Indexed: 10/28/2023] Open
Abstract
Long COVID-19 patients show systemic inflammation and persistent symptoms such as fatigue and malaise, profoundly affecting their quality of life. Since improving oxygenation can oppose inflammation at multiple tissue levels, we hypothesized that hyperbaric oxygen therapy (HBOT) could arrest inflammation progression and thus relieve symptoms of COVID-19. We evaluated oxy-inflammation biomarkers in long COVID-19 subjects treated with HBOT and monitored with non-invasive methods. Five subjects (two athletes and three patients with other comorbidities) were assigned to receive HBOT: 100% inspired O2 at 2.4 ATA in a multiplace hyperbaric chamber for 90 min (three athletes: 15 HBOT × 5 days/wk for 3 weeks; two patients affected by Idiopathic Sudden Sensorineural Hearing Loss: 30 HBOT × 5 days/wk for 6 weeks; and one patient with osteomyelitis: 30 HBOT × 5 days/wk for week for 6 weeks and, after a 30-day break, followed by a second cycle of 20 HBOT). Using saliva and/or urine samples, reactive oxygen species (ROS), antioxidant capacity, cytokines, lipids peroxidation, DNA damage, and renal status were assessed at T1_pre (basal level) and at T2_pre (basal level after treatment), and the results showed attenuated ROS production, lipid peroxidation, DNA damage, NO metabolites, and inflammation biomarker levels, especially in the athletes post-treatment. Thus, HBOT may represent an alternative non-invasive method for treating long COVID-19-induced long-lasting manifestations of oxy-inflammation.
Collapse
Affiliation(s)
- Simona Mrakic-Sposta
- Institute of Clinical Physiology, National Research Council (IFC-CNR), 20162 Milan, Italy;
| | - Alessandra Vezzoli
- Institute of Clinical Physiology, National Research Council (IFC-CNR), 20162 Milan, Italy;
| | | | - Matteo Paganini
- Department of Biomedical Sciences, University of Padova, 35122 Padova, Italy; (M.P.); (E.C.); (T.A.G.); (J.A.); (G.B.)
| | - Enrico Camporesi
- Department of Biomedical Sciences, University of Padova, 35122 Padova, Italy; (M.P.); (E.C.); (T.A.G.); (J.A.); (G.B.)
| | - Tommaso Antonio Giacon
- Department of Biomedical Sciences, University of Padova, 35122 Padova, Italy; (M.P.); (E.C.); (T.A.G.); (J.A.); (G.B.)
| | - Cinzia Dellanoce
- Institute of Clinical Physiology, National Research Council (IFC-CNR), 20162 Milan, Italy;
| | - Jacopo Agrimi
- Department of Biomedical Sciences, University of Padova, 35122 Padova, Italy; (M.P.); (E.C.); (T.A.G.); (J.A.); (G.B.)
| | - Gerardo Bosco
- Department of Biomedical Sciences, University of Padova, 35122 Padova, Italy; (M.P.); (E.C.); (T.A.G.); (J.A.); (G.B.)
| |
Collapse
|
13
|
Golomb BA, Han JH, Langsjoen PH, Dinkeloo E, Zemljic-Harpf AE. Statin Use in Relation to COVID-19 and Other Respiratory Infections: Muscle and Other Considerations. J Clin Med 2023; 12:4659. [PMID: 37510774 PMCID: PMC10380486 DOI: 10.3390/jcm12144659] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/04/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Statins have been widely advocated for use in COVID-19 based on large favorable observational associations buttressed by theoretical expected benefits. However, past favorable associations of statins to pre-COVID-19 infection outcomes (also buttressed by theoretical benefits) were unsupported in meta-analysis of RCTs, RR = 1.00. Initial RCTs in COVID-19 appear to follow this trajectory. Healthy-user/tolerator effects and indication bias may explain these disparities. Moreover, cholesterol drops in proportion to infection severity, so less severely affected individuals may be selected for statin use, contributing to apparent favorable statin associations to outcomes. Cholesterol transports fat-soluble antioxidants and immune-protective vitamins. Statins impair mitochondrial function in those most reliant on coenzyme Q10 (a mevalonate pathway product also transported on cholesterol)-i.e., those with existing mitochondrial compromise, whom data suggest bear increased risks from both COVID-19 and from statins. Thus, statin risks of adverse outcomes are amplified in those patients at risk of poor COVID-19 outcomes-i.e., those in whom adjunctive statin therapy may most likely be given. High reported rates of rhabdomyolysis in hospitalized COVID-19 patients underscore the notion that statin-related risks as well as benefits must be considered. Advocacy for statins in COVID-19 should be suspended pending clear evidence of RCT benefits, with careful attention to risk modifiers.
Collapse
Affiliation(s)
- Beatrice A. Golomb
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA;
| | - Jun Hee Han
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA;
| | | | - Eero Dinkeloo
- Navy and Marine Corps Public Health Center, Portsmouth, VA 23704, USA;
| | - Alice E. Zemljic-Harpf
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA 92093, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92093, USA
| |
Collapse
|
14
|
Bowen DR, Pathak S, Nadar RM, Parise RD, Ramesh S, Govindarajulu M, Moore A, Ren J, Moore T, Dhanasekaran M. Oxidative stress and COVID-19-associated neuronal dysfunction: mechanisms and therapeutic implications. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1153-1167. [PMID: 37357527 PMCID: PMC10465323 DOI: 10.3724/abbs.2023085] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 02/09/2023] [Indexed: 06/27/2023] Open
Abstract
Severe acute respiratory syndrome (SARS)-CoV-2 virus causes novel coronavirus disease 2019 (COVID-19), and there is a possible role for oxidative stress in the pathophysiology of neurological diseases associated with COVID-19. Excessive oxidative stress could be responsible for the thrombosis and other neuronal dysfunctions observed in COVID-19. This review discusses the role of oxidative stress associated with SARS-CoV-2 and the mechanisms involved. Furthermore, the various therapeutics implicated in treating COVID-19 and the oxidative stress that contributes to the etiology and pathogenesis of COVID-19-induced neuronal dysfunction are discussed. Further mechanistic and clinical research to combat COVID-19 is warranted to understand the exact mechanisms, and its true clinical effects need to be investigated to minimize neurological complications from COVID-19.
Collapse
Affiliation(s)
- Dylan R. Bowen
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Suhrud Pathak
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Rishi M. Nadar
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Rachel D. Parise
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Sindhu Ramesh
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Manoj Govindarajulu
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Austin Moore
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | - Jun Ren
- Department of CardiologyZhongshan Hospital Fudan UniversityShanghai200032China
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWA98195USA
| | - Timothy Moore
- Department of Drug Discovery and DevelopmentHarrison College of PharmacyAuburn UniversityAuburn-AL36849USA
| | | |
Collapse
|
15
|
Romani A, Sergi D, Zauli E, Voltan R, Lodi G, Vaccarezza M, Caruso L, Previati M, Zauli G. Nutrients, herbal bioactive derivatives and commensal microbiota as tools to lower the risk of SARS-CoV-2 infection. Front Nutr 2023; 10:1152254. [PMID: 37324739 PMCID: PMC10267353 DOI: 10.3389/fnut.2023.1152254] [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: 01/27/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
The SARS-CoV-2 outbreak has infected a vast population across the world, causing more than 664 million cases and 6.7 million deaths by January 2023. Vaccination has been effective in reducing the most critical aftermath of this infection, but some issues are still present regarding re-infection prevention, effectiveness against variants, vaccine hesitancy and worldwide accessibility. Moreover, although several old and new antiviral drugs have been tested, we still lack robust and specific treatment modalities. It appears of utmost importance, facing this continuously growing pandemic, to focus on alternative practices grounded on firm scientific bases. In this article, we aim to outline a rigorous scientific background and propose complementary nutritional tools useful toward containment, and ultimately control, of SARS-CoV-2 infection. In particular, we review the mechanisms of viral entry and discuss the role of polyunsaturated fatty acids derived from α-linolenic acid and other nutrients in preventing the interaction of SARS-CoV-2 with its entry gateways. In a similar way, we analyze in detail the role of herbal-derived pharmacological compounds and specific microbial strains or microbial-derived polypeptides in the prevention of SARS-CoV-2 entry. In addition, we highlight the role of probiotics, nutrients and herbal-derived compounds in stimulating the immunity response.
Collapse
Affiliation(s)
- Arianna Romani
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Domenico Sergi
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Enrico Zauli
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Rebecca Voltan
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Giada Lodi
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Mauro Vaccarezza
- Curtin Medical School & Curtin Health Innovation Research Institute (CHIRI), Faculty of Health Sciences, Curtin University, Perth, WA, Australia
| | - Lorenzo Caruso
- Department of Environmental and Prevention Sciences and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Maurizio Previati
- Department of Translational Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Giorgio Zauli
- Research Department, King Khaled Eye Specialistic Hospital, Riyadh, Saudi Arabia
| |
Collapse
|
16
|
Leng A, Shah M, Ahmad SA, Premraj L, Wildi K, Li Bassi G, Pardo CA, Choi A, Cho SM. Pathogenesis Underlying Neurological Manifestations of Long COVID Syndrome and Potential Therapeutics. Cells 2023; 12:816. [PMID: 36899952 PMCID: PMC10001044 DOI: 10.3390/cells12050816] [Citation(s) in RCA: 67] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
The development of long-term symptoms of coronavirus disease 2019 (COVID-19) more than four weeks after primary infection, termed "long COVID" or post-acute sequela of COVID-19 (PASC), can implicate persistent neurological complications in up to one third of patients and present as fatigue, "brain fog", headaches, cognitive impairment, dysautonomia, neuropsychiatric symptoms, anosmia, hypogeusia, and peripheral neuropathy. Pathogenic mechanisms of these symptoms of long COVID remain largely unclear; however, several hypotheses implicate both nervous system and systemic pathogenic mechanisms such as SARS-CoV2 viral persistence and neuroinvasion, abnormal immunological response, autoimmunity, coagulopathies, and endotheliopathy. Outside of the CNS, SARS-CoV-2 can invade the support and stem cells of the olfactory epithelium leading to persistent alterations to olfactory function. SARS-CoV-2 infection may induce abnormalities in innate and adaptive immunity including monocyte expansion, T-cell exhaustion, and prolonged cytokine release, which may cause neuroinflammatory responses and microglia activation, white matter abnormalities, and microvascular changes. Additionally, microvascular clot formation can occlude capillaries and endotheliopathy, due to SARS-CoV-2 protease activity and complement activation, can contribute to hypoxic neuronal injury and blood-brain barrier dysfunction, respectively. Current therapeutics target pathological mechanisms by employing antivirals, decreasing inflammation, and promoting olfactory epithelium regeneration. Thus, from laboratory evidence and clinical trials in the literature, we sought to synthesize the pathophysiological pathways underlying neurological symptoms of long COVID and potential therapeutics.
Collapse
Affiliation(s)
- Albert Leng
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Manuj Shah
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Syed Ameen Ahmad
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lavienraj Premraj
- Department of Neurology, Griffith University School of Medicine, Gold Coast, Brisbane, QLD 4215, Australia
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD 4032, Australia
| | - Karin Wildi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD 4032, Australia
| | - Gianluigi Li Bassi
- Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD 4032, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD 4072, Australia
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD 4000, Australia
- Intensive Care Unit, St Andrew’s War Memorial Hospital and the Wesley Hospital, Uniting Care Hospitals, Brisbane, QLD 4000, Australia
- Wesley Medical Research, Auchenflower, QLD 4066, Australia
| | - Carlos A. Pardo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alex Choi
- Division of Neurosciences Critical Care, Department of Neurosurgery, UT Houston, Houston, TX 77030, USA
| | - Sung-Min Cho
- Divisions of Neurosciences Critical Care and Cardiac Surgery, Departments of Neurology, Surgery, Anesthesiology and Critical Care Medicine and Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
17
|
Botto L, Lonati E, Russo S, Cazzaniga E, Bulbarelli A, Palestini P. Effects of PM2.5 Exposure on the ACE/ACE2 Pathway: Possible Implication in COVID-19 Pandemic. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:4393. [PMID: 36901403 PMCID: PMC10002082 DOI: 10.3390/ijerph20054393] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 06/18/2023]
Abstract
Particulate matter (PM) is a harmful component of urban air pollution and PM2.5, in particular, can settle in the deep airways. The RAS system plays a crucial role in the pathogenesis of pollution-induced inflammatory diseases: the ACE/AngII/AT1 axis activates a pro-inflammatory pathway counteracted by the ACE2/Ang(1-7)/MAS axis, which in turn triggers an anti-inflammatory and protective pathway. However, ACE2 acts also as a receptor through which SARS-CoV-2 penetrates host cells to replicate. COX-2, HO-1, and iNOS are other crucial proteins involved in ultrafine particles (UFP)-induced inflammation and oxidative stress, but closely related to the course of the COVID-19 disease. BALB/c male mice were subjected to PM2.5 sub-acute exposure to study its effects on ACE2 and ACE, COX-2, HO-1 and iNOS proteins levels, in the main organs concerned with the pathogenesis of COVID-19. The results obtained show that sub-acute exposure to PM2.5 induces organ-specific modifications which might predispose to greater susceptibility to severe symptomatology in the case of SARS-CoV-2 infection. The novelty of this work consists in using a molecular study, carried out in the lung but also in the main organs involved in the disease, to analyze the close relationship between exposure to pollution and the pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Laura Botto
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Elena Lonati
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Stefania Russo
- FIMP-Federazione Italiana Medici Pediatri, 00185 Rome, Italy
| | - Emanuela Cazzaniga
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- POLARIS Centre, University of Milano-Bicocca, 20126 Milan, Italy
| | - Alessandra Bulbarelli
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- POLARIS Centre, University of Milano-Bicocca, 20126 Milan, Italy
| | - Paola Palestini
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- POLARIS Centre, University of Milano-Bicocca, 20126 Milan, Italy
| |
Collapse
|
18
|
Waraich RS, Sohail FA, Khan G, Durr-E-Shahwar S, Khan B, Rafi S, Nasir S. Enhanced Expression of RAGE AXIS Is Associated with Severity of COVID-19 in Patients with Comorbidities. Metab Syndr Relat Disord 2023; 21:141-147. [PMID: 36787461 DOI: 10.1089/met.2022.0089] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
Background: There is a limited understanding of molecular and cellular events that derive disease progression in patients with corona virus disease 2019 (COVID-19). Receptor for advanced glycation end products (RAGE) is hyperactive in development and complications of several diseases by mediating oxidative stress and inflammation in the body. The present study aims to explore activation of RAGE signaling in patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with preexisting comorbidities, including hypertension and or diabetes. Methods: A total of 442 subjects with COVID-19, were recruited for the study. The molecular mechanism of Covid-19 was explored in blood cells, using ELISA, RT- PCR and Western blot. Results: Enhanced levels of ligands of RAGE, including AGEs, S100, and high-mobility group box-1 (HMGB-1) were observed in COVID-19 patients with severe diseases; however, their level was significantly higher in COVID-19 patients with comorbidities compared to COVID-19 patients without comorbidities. The expression of RAGE in parallel to ligands accumulation was significantly increased in patients with severe disease and comorbidities compared to COVID-19 patients with severe disease without comorbidities. The expression of downstream effectors of RAGE, including STAT-3 and nuclear factor kappa B (NF-kB), was also enhanced and their activity was increased in COVID-19 patients with comorbidities. Levels of inflammatory and oxidative stress biomarkers were markedly increased in COVID-19 patients with comorbidities. Conclusions: We conclude that upregulated RAGE axis plays critical role, to worsen the severity of the SARS-CoV-2 infection in patients with preexisting comorbidities and partly explain inflammatory and oxidative stress storm in severe COVID-19 patients.
Collapse
Affiliation(s)
- Rizwana Sanaullah Waraich
- Department of Biomedical and Biological Sciences, Biomedical Research Center, Sohail University, Karachi, Pakistan
| | - Fadieleh Adnan Sohail
- Department of Biomedical and Biological Sciences, Biomedical Research Center, Sohail University, Karachi, Pakistan.,Medical Education Department, Jinnah Medical and Dental College, Karachi, Pakistan
| | - Gulnaz Khan
- Department of Biomedical and Biological Sciences, Biomedical Research Center, Sohail University, Karachi, Pakistan
| | - Syeda Durr-E-Shahwar
- Department of Biomedical and Biological Sciences, Biomedical Research Center, Sohail University, Karachi, Pakistan
| | - Bushra Khan
- Department of Biomedical and Biological Sciences, Biomedical Research Center, Sohail University, Karachi, Pakistan
| | - Sidra Rafi
- Department of Biomedical and Biological Sciences, Biomedical Research Center, Sohail University, Karachi, Pakistan
| | - Shumaila Nasir
- Department of Biomedical and Biological Sciences, Biomedical Research Center, Sohail University, Karachi, Pakistan
| |
Collapse
|
19
|
Transient Changes in the Plasma of Astrocytic and Neuronal Injury Biomarkers in COVID-19 Patients without Neurological Syndromes. Int J Mol Sci 2023; 24:ijms24032715. [PMID: 36769057 PMCID: PMC9917569 DOI: 10.3390/ijms24032715] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
The levels of several glial and neuronal plasma biomarkers have been found to increase during the acute phase in COVID-19 patients with neurological symptoms. However, replications in patients with minor or non-neurological symptoms are needed to understand their potential as indicators of CNS injury or vulnerability. Plasma levels of glial fibrillary acidic protein (GFAP), neurofilament light chain protein (NfL), and total Tau (T-tau) were determined by Single molecule array (Simoa) immunoassays in 45 samples from COVID-19 patients in the acute phase of infection [moderate (n = 35), or severe (n = 10)] with minor or non-neurological symptoms; in 26 samples from fully recovered patients after ~2 months of clinical follow-up [moderate (n = 23), or severe (n = 3)]; and in 14 non-infected controls. Plasma levels of the SARS-CoV-2 receptor, angiotensin-converting enzyme 2 (ACE2), were also determined by Western blot. Patients with COVID-19 without substantial neurological symptoms had significantly higher plasma concentrations of GFAP, a marker of astrocytic activation/injury, and of NfL and T-tau, markers of axonal damage and neuronal degeneration, compared with controls. All these biomarkers were correlated in COVID-19 patients at the acute phase. Plasma GFAP, NfL and T-tau levels were all normalized after recovery. Recovery was also observed in the return to normal values of the quotient between the ACE2 fragment and circulating full-length species, following the change noticed in the acute phase of infection. None of these biomarkers displayed differences in plasma samples at the acute phase or recovery when the COVID-19 subjects were sub-grouped according to occurrence of minor symptoms at re-evaluation 3 months after the acute episode (so called post-COVID or "long COVID"), such as asthenia, myalgia/arthralgia, anosmia/ageusia, vision impairment, headache or memory loss. Our study demonstrated altered plasma GFAP, NfL and T-tau levels in COVID-19 patients without substantial neurological manifestation at the acute phase of the disease, providing a suitable indication of CNS vulnerability; but these biomarkers fail to predict the occurrence of delayed minor neurological symptoms.
Collapse
|
20
|
Gauthier AG, Lin M, Zefi S, Kulkarni A, Thakur GA, Ashby CR, Mantell LL. GAT107-mediated α7 nicotinic acetylcholine receptor signaling attenuates inflammatory lung injury and mortality in a mouse model of ventilator-associated pneumonia by alleviating macrophage mitochondrial oxidative stress via reducing MnSOD-S-glutathionylation. Redox Biol 2023; 60:102614. [PMID: 36717349 PMCID: PMC9950665 DOI: 10.1016/j.redox.2023.102614] [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: 11/05/2022] [Revised: 01/09/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Supraphysiological concentrations of oxygen (hyperoxia) can compromise host defense and increase susceptibility to bacterial and viral infections, causing ventilator-associated pneumonia (VAP). Compromised host defense and inflammatory lung injury are mediated, in part, by high extracellular concentrations of HMGB1, which can be decreased by GTS-21, a partial agonist of α7 nicotinic acetylcholine receptor (α7nAChR). Here, we report that a novel α7nAChR agonistic positive allosteric modulator (ago-PAM), GAT107, at 3.3 mg/kg, i.p., significantly decreased animal mortality and markers of inflammatory injury in mice exposed to hyperoxia and subsequently infected with Pseudomonas aeruginosa. The incubation of macrophages with 3.3 μM of GAT107 significantly decreased hyperoxia-induced extracellular HMGB1 accumulation and HMGB1-induced macrophage phagocytic dysfunction. Hyperoxia-compromised macrophage function was correlated with impaired mitochondrial membrane integrity, increased superoxide levels, and decreased manganese superoxide dismutase (MnSOD) activity. This compromised MnSOD activity is due to a significant increase in its level of glutathionylation. The incubation of hyperoxic macrophages with 3.3 μM of GAT107 significantly decreases the levels of glutathionylated MnSOD, and restores MnSOD activity and mitochondrial membrane integrity. Thus, GAT107 restored hyperoxia-compromised phagocytic functions by decreasing HMGB1 release, most likely via a mitochondrial-directed pathway. Overall, our results suggest that GAT107 may be a potential treatment to decrease acute inflammatory lung injury by increasing host defense in patients with VAP.
Collapse
Affiliation(s)
- Alex G. Gauthier
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Mosi Lin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Sidorela Zefi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | | | | | - Charles R. Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Lin L. Mantell
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA,Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA,Corresponding author. Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, 128 St. Albert Hall, 8000 Utopia Parkway, Queens, NY, 11439, USA.
| |
Collapse
|
21
|
García-García T, Fernández-Rodríguez R, Redondo N, de Lucas-Rius A, Zaldívar-López S, López-Ayllón BD, Suárez-Cárdenas JM, Jiménez-Marín Á, Montoya M, Garrido JJ. Impairment of antiviral immune response and disruption of cellular functions by SARS-CoV-2 ORF7a and ORF7b. iScience 2022; 25:105444. [PMID: 36310646 PMCID: PMC9597514 DOI: 10.1016/j.isci.2022.105444] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/20/2022] [Accepted: 10/21/2022] [Indexed: 11/09/2022] Open
Abstract
SARS-CoV-2, the causative agent of the present COVID-19 pandemic, possesses eleven accessory proteins encoded in its genome, and some have been implicated in facilitating infection and pathogenesis through their interaction with cellular components. Among these proteins, accessory protein ORF7a and ORF7b functions are poorly understood. In this study, A549 cells were transduced to express ORF7a and ORF7b, respectively, to explore more in depth the role of each accessory protein in the pathological manifestation leading to COVID-19. Bioinformatic analysis and integration of transcriptome results identified defined canonical pathways and functional groupings revealing that after expression of ORF7a or ORF7b, the lung cells are potentially altered to create conditions more favorable for SARS-CoV-2, by inhibiting the IFN-I response, increasing proinflammatory cytokines release, and altering cell metabolic activity and adhesion. Based on these results, it is plausible to suggest that ORF7a or ORF7b could be used as biomarkers of progression in this pandemic.
Collapse
Affiliation(s)
- Tránsito García-García
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonoses and Emergent Diseases ENZOEM, Department of Genetics, University of Córdoba, Córdoba, Spain
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), GA-14 Research Group, Córdoba, Spain
| | - Raúl Fernández-Rodríguez
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonoses and Emergent Diseases ENZOEM, Department of Genetics, University of Córdoba, Córdoba, Spain
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), GA-14 Research Group, Córdoba, Spain
| | - Natalia Redondo
- Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid 28040, Spain
| | - Ana de Lucas-Rius
- Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid 28040, Spain
| | - Sara Zaldívar-López
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonoses and Emergent Diseases ENZOEM, Department of Genetics, University of Córdoba, Córdoba, Spain
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), GA-14 Research Group, Córdoba, Spain
| | - Blanca Dies López-Ayllón
- Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid 28040, Spain
| | - José M. Suárez-Cárdenas
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonoses and Emergent Diseases ENZOEM, Department of Genetics, University of Córdoba, Córdoba, Spain
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), GA-14 Research Group, Córdoba, Spain
| | - Ángeles Jiménez-Marín
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonoses and Emergent Diseases ENZOEM, Department of Genetics, University of Córdoba, Córdoba, Spain
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), GA-14 Research Group, Córdoba, Spain
| | - María Montoya
- Molecular Biomedicine Department, Centro de Investigaciones Biológicas Margarita Salas (CIB), CSIC, Madrid 28040, Spain
- Corresponding author
| | - Juan J. Garrido
- Immunogenomics and Molecular Pathogenesis Group, UIC Zoonoses and Emergent Diseases ENZOEM, Department of Genetics, University of Córdoba, Córdoba, Spain
- Maimónides Biomedical Research Institute of Córdoba (IMIBIC), GA-14 Research Group, Córdoba, Spain
- Corresponding author
| |
Collapse
|
22
|
Cavalcante LTDF, da Fonseca GC, Amado Leon LA, Salvio AL, Brustolini OJ, Gerber AL, Guimarães APDC, Marques CAB, Fernandes RA, Ramos Filho CHF, Kader RL, Pimentel Amaro M, da Costa Gonçalves JP, Vieira Alves-Leon S, Vasconcelos ATR. Buffy Coat Transcriptomic Analysis Reveals Alterations in Host Cell Protein Synthesis and Cell Cycle in Severe COVID-19 Patients. Int J Mol Sci 2022; 23:13588. [PMID: 36362378 PMCID: PMC9659271 DOI: 10.3390/ijms232113588] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/25/2023] Open
Abstract
Transcriptome studies have reported the dysregulation of cell cycle-related genes and the global inhibition of host mRNA translation in COVID-19 cases. However, the key genes and cellular mechanisms that are most affected by the severe outcome of this disease remain unclear. For this work, the RNA-seq approach was used to study the differential expression in buffy coat cells of two groups of people infected with SARS-CoV-2: (a) Mild, with mild symptoms; and (b) SARS (Severe Acute Respiratory Syndrome), who were admitted to the intensive care unit with the severe COVID-19 outcome. Transcriptomic analysis revealed 1009 up-regulated and 501 down-regulated genes in the SARS group, with 10% of both being composed of long non-coding RNA. Ribosome and cell cycle pathways were enriched among down-regulated genes. The most connected proteins among the differentially expressed genes involved transport dysregulation, proteasome degradation, interferon response, cytokinesis failure, and host translation inhibition. Furthermore, interactome analysis showed Fibrillarin to be one of the key genes affected by SARS-CoV-2. This protein interacts directly with the N protein and long non-coding RNAs affecting transcription, translation, and ribosomal processes. This work reveals a group of dysregulated processes, including translation and cell cycle, as key pathways altered in severe COVID-19 outcomes.
Collapse
Affiliation(s)
| | | | - Luciane Almeida Amado Leon
- Laboratório de Desenvolvimento Tecnológico em Virologia, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro 21040-360, Brazil
| | - Andreza Lemos Salvio
- Laboratório de Neurociências Translacional, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro 20211-040, Brazil
| | - Otávio José Brustolini
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro 25651-076, Brazil
| | - Alexandra Lehmkuhl Gerber
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro 25651-076, Brazil
| | - Ana Paula de Campos Guimarães
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro 25651-076, Brazil
| | - Carla Augusta Barreto Marques
- Laboratório de Neurociências Translacional, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro 20211-040, Brazil
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-617, Brazil
| | - Renan Amphilophio Fernandes
- Laboratório de Neurociências Translacional, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro 20211-040, Brazil
| | | | - Rafael Lopes Kader
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-617, Brazil
| | - Marisa Pimentel Amaro
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-617, Brazil
| | - João Paulo da Costa Gonçalves
- Laboratório de Neurociências Translacional, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro 20211-040, Brazil
- Yale New Haven Hospital, New Haven, CT 06510, USA
| | - Soniza Vieira Alves-Leon
- Laboratório de Neurociências Translacional, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro 20211-040, Brazil
- Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-617, Brazil
| | - Ana Tereza Ribeiro Vasconcelos
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro 25651-076, Brazil
| |
Collapse
|
23
|
Glycation-Driven Inflammation: COVID-19 Severity in Pregnant Women and Perinatal Outcomes. Nutrients 2022; 14:nu14194037. [PMID: 36235696 PMCID: PMC9570639 DOI: 10.3390/nu14194037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 11/17/2022] Open
Abstract
The link between being pregnant and overweight or obese and the infectivity and virulence of the SARS CoV-2 virus is likely to be caused by SARS-CoV-2 spike protein glycosylation, which may work as a glycan shield. Methylglyoxal (MGO), an important advanced glycation end-product (AGE), and glycated albumin (GA) are the results of poor subclinical glucose metabolism and are indices of oxidative stress. Forty-one consecutive cases of SARS-CoV-2-positive pregnant patients comprising 25% pre-pregnancy overweight women and 25% obese women were recruited. The aim of our study was to compare the blood levels of MGO and GA in pregnant women with asymptomatic and symptomatic SARS-CoV-2 infection with pregnant women without SARS-CoV-2 infection with low risk and uneventful pregnancies and to evaluate the relative perinatal outcomes. The MGO and GA values of the SARS-CoV-2 cases were statistically significantly higher than those of the negative control subjects. In addition, the SARS-CoV-2-positive pregnant patients who suffered of moderate to severe COVID-19 syndrome had higher values of GA than those infected and presenting with mild symptoms or those with asymptomatic infection. Premature delivery and infants of a small size for their gestational age were overrepresented in this cohort, even in mild-asymptomatic patients for whom delivery was not indicated by the COVID-19 syndrome. Moreover, ethnic minorities were overrepresented among the severe cases. The AGE–RAGE oxidative stress axis on the placenta and multiple organs caused by MGO and GA levels, associated with the biological mechanisms of the glycation of the SARS-CoV-2 spike protein, could help to explain the infectivity and virulence of this virus in pregnant patients affected by being overweight or obese or having gestational diabetes, and the increased risk of premature delivery and/or low newborn weight.
Collapse
|
24
|
Notarbartolo V, Montante C, Ferrante G, Giuffrè M. Antioxidant Effects of Dietary Supplements on Adult COVID-19 Patients: Why Do We Not Also Use Them in Children? Antioxidants (Basel) 2022; 11:antiox11091638. [PMID: 36139712 PMCID: PMC9495518 DOI: 10.3390/antiox11091638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/09/2022] [Accepted: 08/19/2022] [Indexed: 11/16/2022] Open
Abstract
Respiratory tract infections (RTIs) are very common in children, especially in the first five years of life, and several viruses, such as the influenza virus, Respiratory Syncytial Virus, and Rhinovirus, are triggers for symptoms that usually affect the upper airways. It has been known that during respiratory viral infections, a condition of oxidative stress (OS) occurs, and many studies have suggested the potential use of antioxidants as complementary components in prophylaxis and/or therapy of respiratory viral infections. Preliminary data have demonstrated that antioxidants may also interfere with the new coronavirus 2’s entry and replication in human cells, and that they have a role in the downregulation of several pathogenetic mechanisms involved in disease severity. Starting from preclinical data, the aim of this narrative review is to evaluate the current evidence about the main antioxidants that are potentially useful for preventing and treating Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection in adults and to speculate on their possible use in children by exploring the most relevant issues affecting their use in clinical practice, as well as the associated evidence gaps and research limitations.
Collapse
Affiliation(s)
- Veronica Notarbartolo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialities, University of Palermo, 90128 Palermo, Italy
- Correspondence:
| | - Claudio Montante
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialities, University of Palermo, 90128 Palermo, Italy
| | - Giuliana Ferrante
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Pediatric Division, University of Verona, 37134 Verona, Italy
| | - Mario Giuffrè
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialities, University of Palermo, 90128 Palermo, Italy
| |
Collapse
|
25
|
Tsermpini EE, Glamočlija U, Ulucan-Karnak F, Redenšek Trampuž S, Dolžan V. Molecular Mechanisms Related to Responses to Oxidative Stress and Antioxidative Therapies in COVID-19: A Systematic Review. Antioxidants (Basel) 2022; 11:1609. [PMID: 36009328 PMCID: PMC9405444 DOI: 10.3390/antiox11081609] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/25/2022] Open
Abstract
The coronavirus disease (COVID-19) pandemic is a leading global health and economic challenge. What defines the disease's progression is not entirely understood, but there are strong indications that oxidative stress and the defense against reactive oxygen species are crucial players. A big influx of immune cells to the site of infection is marked by the increase in reactive oxygen and nitrogen species. Our article aims to highlight the critical role of oxidative stress in the emergence and severity of COVID-19 and, more importantly, to shed light on the underlying molecular and genetic mechanisms. We have reviewed the available literature and clinical trials to extract the relevant genetic variants within the oxidative stress pathway associated with COVID-19 and the anti-oxidative therapies currently evaluated in the clinical trials for COVID-19 treatment, in particular clinical trials on glutathione and N-acetylcysteine.
Collapse
Affiliation(s)
- Evangelia Eirini Tsermpini
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Una Glamočlija
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Faculty of Pharmacy, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina
- School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Fulden Ulucan-Karnak
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Department of Medical Biochemistry, Faculty of Medicine, Ege University, Bornova, 35100 İzmir, Turkey
| | - Sara Redenšek Trampuž
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
26
|
Endothelial Dysfunction, HMGB1, and Dengue: An Enigma to Solve. Viruses 2022; 14:v14081765. [PMID: 36016387 PMCID: PMC9414358 DOI: 10.3390/v14081765] [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: 07/11/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Dengue is a viral infection caused by dengue virus (DENV), which has a significant impact on public health worldwide. Although most infections are asymptomatic, a series of severe clinical manifestations such as hemorrhage and plasma leakage can occur during the severe presentation of the disease. This suggests that the virus or host immune response may affect the protective function of endothelial barriers, ultimately being considered the most relevant event in severe and fatal dengue pathogenesis. The mechanisms that induce these alterations are diverse. It has been suggested that the high mobility group box 1 protein (HMGB1) may be involved in endothelial dysfunction. This non-histone nuclear protein has different immunomodulatory activities and belongs to the alarmin group. High concentrations of HMGB1 have been detected in patients with several infectious diseases, including dengue, and it could be considered as a biomarker for the early diagnosis of dengue and a predictor of complications of the disease. This review summarizes the main features of dengue infection and describes the known causes associated with endothelial dysfunction, highlighting the involvement and possible relationship between HMGB1 and DENV.
Collapse
|
27
|
Wang M, Yu B, Wang J, Wang Y, Liang L. Exploring the role of Xingren on
COVID
‐19 based on network pharmacology and molecular docking. J Food Biochem 2022; 46:e14363. [PMID: 35933696 PMCID: PMC9539046 DOI: 10.1111/jfbc.14363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/20/2022] [Accepted: 07/18/2022] [Indexed: 11/29/2022]
Abstract
Since the outbreak of novel Coronavirus Pneumonia 2019 (COVID‐19), the role of Almonds (Xingren) in the protection and treatment of COVID‐19 is not clear. Network pharmacology and molecular docking were used to explore the potential mechanism and potential key targets of Xingren on COVID‐19. A total of nine common targets between them were obtained, and these targets were involved in multiple related processes of GO and KEGG pathway enrichment analysis. Molecular docking showed that licochalcone B has the best binding energy (−9.33 kJ·mol−1) to PTGS2. They are maybe the important ingredient and key potential target. Its possible mechanism is to intervene anxiety disorder in the process of disease development, such as regulation of blood pressure, reactive oxygen species metabolic process, leishmaniasis peroxisome, and IL‐17 signaling pathway.
Collapse
Affiliation(s)
- Maoru Wang
- Drug Dispensing Department The Third Hospital of Mianyang, Sichuan Mental Health Center Mianyang China
| | - Bin Yu
- Department of Pharmacy, Mianyang Central Hospital School of Medicine, University of Electronic Science and Technology of China Mianyang China
| | - Jisheng Wang
- Drug Dispensing Department The Third Hospital of Mianyang, Sichuan Mental Health Center Mianyang China
| | - Yu Wang
- Department of Pharmacy, Mianyang Central Hospital School of Medicine, University of Electronic Science and Technology of China Mianyang China
| | - Libo Liang
- Drug Dispensing Department The Third Hospital of Mianyang, Sichuan Mental Health Center Mianyang China
| |
Collapse
|
28
|
Effect of Parecoxib Sodium Preemptive Analgesia on the Recovery Period of General Anesthesia in Patients Undergoing Glioma Resection. JOURNAL OF ONCOLOGY 2022; 2022:4934343. [PMID: 35734224 PMCID: PMC9208977 DOI: 10.1155/2022/4934343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/26/2022] [Accepted: 02/15/2022] [Indexed: 11/30/2022]
Abstract
Objective To investigate the effect of parecoxib sodium preemptive analgesia on postoperative complications and postoperative recovery of patients undergoing glioma resection. Methods A total of 200 eligible patients with low-grade gliomas in the functional brain area scheduled for an awake craniotomy between January 2017 and December 2020 were reviewed. The subjects were divided into two groups: the study group (n = 100) given dexmedetomidine plus parecoxib sodium for pre-emptive analgesia 30 minutes preoperatively, and the control group (n = 100) receiving dexmedetomidine alone. Venous blood was collected before surgery, at the time of postoperative recovery, and 24 hours after operation, mean artery pressure (MAP) and heart rate (HR) were recorded during surgery. Sedation satisfaction, agitation rate, numerical pain score (NRS), postoperative complications, minimental state examination (MMSE) scores, quality of life (QoL) scores, and incidence of adverse events were also investigated after the surgery. Results There were no significant differences in operation time, awakening time, intraoperative awakening time, and extubation time between the two groups (P > 0.05). Compared with the control group, the ΔMAP (7.26 ± 2.21 versus 5.78 ± 2.36 mmHg) and the ΔHR (11.35 ± 3.66 versus 8.84 ± 2.47 beats/min) were significantly lower in the study group (P < 0.05). Compared with the control group, the satisfaction was higher and agitation rate was lower in the study group (P < 0.05). The incidence of intracranial infection and pulmonary infection decreased after operation (P < 0.05). The NRS of the study group was remarkably lower than the control group at 12 hours postoperatively Preoperative MMSE score and QoL score showed no statistical difference (P > 0.05), while postoperative MMSE and QoL scores showed statistical difference (P < 0.05). Conclusion This study suggests that parecoxib sodium can significantly improve the level of sedation and analgesia in patients undergoing glioma resection, reduce the incidence of intracranial infection and pulmonary infection.
Collapse
|
29
|
Allen CNS, Santerre M, Arjona SP, Ghaleb LJ, Herzi M, Llewellyn MD, Shcherbik N, Sawaya BE. SARS-CoV-2 Causes Lung Inflammation through Metabolic Reprogramming and RAGE. Viruses 2022; 14:983. [PMID: 35632725 PMCID: PMC9143006 DOI: 10.3390/v14050983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 12/26/2022] Open
Abstract
Clinical studies indicate that patients infected with SARS-CoV-2 develop hyperinflammation, which correlates with increased mortality. The SARS-CoV-2/COVID-19-dependent inflammation is thought to occur via increased cytokine production and hyperactivity of RAGE in several cell types, a phenomenon observed for other disorders and diseases. Metabolic reprogramming has been shown to contribute to inflammation and is considered a hallmark of cancer, neurodegenerative diseases, and viral infections. Malfunctioning glycolysis, which normally aims to convert glucose into pyruvate, leads to the accumulation of advanced glycation end products (AGEs). Being aberrantly generated, AGEs then bind to their receptor, RAGE, and activate several pro-inflammatory genes, such as IL-1b and IL-6, thus, increasing hypoxia and inducing senescence. Using the lung epithelial cell (BEAS-2B) line, we demonstrated that SARS-CoV-2 proteins reprogram the cellular metabolism and increase pyruvate kinase muscle isoform 2 (PKM2). This deregulation promotes the accumulation of AGEs and senescence induction. We showed the ability of the PKM2 stabilizer, Tepp-46, to reverse the observed glycolysis changes/alterations and restore this essential metabolic process.
Collapse
Affiliation(s)
- Charles N. S. Allen
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
| | - Maryline Santerre
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
| | - Sterling P. Arjona
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
| | - Lea J. Ghaleb
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
| | - Muna Herzi
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
| | - Megan D. Llewellyn
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
| | - Natalia Shcherbik
- Department for Cell Biology and Neuroscience, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA;
| | - Bassel E. Sawaya
- Molecular Studies of Neurodegenerative Diseases Lab., FELS Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (M.S.); (S.P.A.); (L.J.G.); (M.H.); (M.D.L.)
- Departments of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Cancer and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
30
|
Vollbracht C, Kraft K. Oxidative Stress and Hyper-Inflammation as Major Drivers of Severe COVID-19 and Long COVID: Implications for the Benefit of High-Dose Intravenous Vitamin C. Front Pharmacol 2022; 13:899198. [PMID: 35571085 PMCID: PMC9100929 DOI: 10.3389/fphar.2022.899198] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/14/2022] [Indexed: 12/25/2022] Open
Abstract
Oxidative stress is a pivotal point in the pathophysiology of COVID-19 and presumably also in Long-COVID. Inflammation and oxidative stress are mutually reinforcing each other, thus contributing to the systemic hyperinflammatory state and coagulopathy which are cardinal pathological mechanisms of severe stages. COVID-19 patients, like other critically ill patients e.g. with pneumonia, very often show severe deficiency of the antioxidant vitamin C. So far, it has not been investigated how long this deficiency lasts or whether patients with long COVID symptoms also suffer from deficiencies. A vitamin C deficit has serious pathological consequences because vitamin C is one of the most effective antioxidants, but also co-factor of many enzymatic processes that affect the immune and nervous system, blood circulation and energy metabolism. Because of its anti-oxidative, anti-inflammatory, endothelial-restoring, and immunomodulatory effects the supportive intravenous (iv) use of supraphysiological doses has been investigated so far in 12 controlled or observational studies with altogether 1578 inpatients with COVID-19. In these studies an improved oxygenation, a decrease in inflammatory markers and a faster recovery were observed. In addition, early treatment with iv high dose vitamin C seems to reduce the risks of severe courses of the disease such as pneumonia and also mortality. Persistent inflammation, thrombosis and a dysregulated immune response (auto-immune phenomena and/or persistent viral load) seem to be major contributors to Long-COVID. Oxidative stress and inflammation are involved in the development and progression of fatigue and neuro-psychiatric symptoms in various diseases by disrupting tissue (e.g. autoantibodies), blood flow (e.g. immune thrombosis) and neurotransmitter metabolism (e.g. excitotoxicity). In oncological diseases, other viral infections and autoimmune diseases, which are often associated with fatigue, cognitive disorders, pain and depression similar to Long-COVID, iv high dose vitamin C was shown to significantly relieve these symptoms. Supportive iv vitamin C in acute COVID-19 might therefore reduce the risk of severe courses and also the development of Long-COVID.
Collapse
Affiliation(s)
- Claudia Vollbracht
- Medical Science Department, Pascoe Pharmazeutische Präparate GmbH, Giessen, Germany
| | - Karin Kraft
- Chair of Naturopathy, University Medicine Rostock, Rostock, Germany
| |
Collapse
|
31
|
Nelson AR. Peripheral Pathways to Neurovascular Unit Dysfunction, Cognitive Impairment, and Alzheimer’s Disease. Front Aging Neurosci 2022; 14:858429. [PMID: 35517047 PMCID: PMC9062225 DOI: 10.3389/fnagi.2022.858429] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/03/2022] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. It was first described more than a century ago, and scientists are acquiring new data and learning novel information about the disease every day. Although there are nuances and details continuously being unraveled, many key players were identified in the early 1900’s by Dr. Oskar Fischer and Dr. Alois Alzheimer, including amyloid-beta (Aβ), tau, vascular abnormalities, gliosis, and a possible role of infections. More recently, there has been growing interest in and appreciation for neurovascular unit dysfunction that occurs early in mild cognitive impairment (MCI) before and independent of Aβ and tau brain accumulation. In the last decade, evidence that Aβ and tau oligomers are antimicrobial peptides generated in response to infection has expanded our knowledge and challenged preconceived notions. The concept that pathogenic germs cause infections generating an innate immune response (e.g., Aβ and tau produced by peripheral organs) that is associated with incident dementia is worthwhile considering in the context of sporadic AD with an unknown root cause. Therefore, the peripheral amyloid hypothesis to cognitive impairment and AD is proposed and remains to be vetted by future research. Meanwhile, humans remain complex variable organisms with individual risk factors that define their immune status, neurovascular function, and neuronal plasticity. In this focused review, the idea that infections and organ dysfunction contribute to Alzheimer’s disease, through the generation of peripheral amyloids and/or neurovascular unit dysfunction will be explored and discussed. Ultimately, many questions remain to be answered and critical areas of future exploration are highlighted.
Collapse
|
32
|
Allen CNS, Arjona SP, Santerre M, Sawaya BE. Hallmarks of Metabolic Reprogramming and Their Role in Viral Pathogenesis. Viruses 2022; 14:602. [PMID: 35337009 PMCID: PMC8955778 DOI: 10.3390/v14030602] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/07/2023] Open
Abstract
Metabolic reprogramming is a hallmark of cancer and has proven to be critical in viral infections. Metabolic reprogramming provides the cell with energy and biomass for large-scale biosynthesis. Based on studies of the cellular changes that contribute to metabolic reprogramming, seven main hallmarks can be identified: (1) increased glycolysis and lactic acid, (2) increased glutaminolysis, (3) increased pentose phosphate pathway, (4) mitochondrial changes, (5) increased lipid metabolism, (6) changes in amino acid metabolism, and (7) changes in other biosynthetic and bioenergetic pathways. Viruses depend on metabolic reprogramming to increase biomass to fuel viral genome replication and production of new virions. Viruses take advantage of the non-metabolic effects of metabolic reprogramming, creating an anti-apoptotic environment and evading the immune system. Other non-metabolic effects can negatively affect cellular function. Understanding the role metabolic reprogramming plays in viral pathogenesis may provide better therapeutic targets for antivirals.
Collapse
Affiliation(s)
- Charles N. S. Allen
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (S.P.A.); (M.S.)
| | - Sterling P. Arjona
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (S.P.A.); (M.S.)
| | - Maryline Santerre
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (S.P.A.); (M.S.)
| | - Bassel E. Sawaya
- Molecular Studies of Neurodegenerative Diseases Lab, FELS Cancer Institute for Personalized Medicine Institute, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; (C.N.S.A.); (S.P.A.); (M.S.)
- Departments of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Department of Cancer and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|