1
|
Ashraf MS, Tuli K, Moiz S, Sharma SK, Sharma D, Adnan M. AMP kinase: A promising therapeutic drug target for post-COVID-19 complications. Life Sci 2024:123202. [PMID: 39489398 DOI: 10.1016/j.lfs.2024.123202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, has resulted in severe respiratory issues and persistent complications, particularly affecting glucose metabolism. Patients with or without pre-existing diabetes often experience worsened symptoms, highlighting the need for innovative therapeutic approaches. AMPK, a crucial regulator of cellular energy balance, plays a pivotal role in glucose metabolism, insulin sensitivity, and inflammatory responses. AMPK activation, through allosteric or kinase-dependent mechanisms, impacts cellular processes like glucose uptake, fatty acid oxidation, and autophagy. The tissue-specific distribution of AMPK emphasizes its role in maintaining metabolic homeostasis throughout the body. Intriguingly, SARS-CoV-2 infection inhibits AMPK, contributing to metabolic dysregulation and post-COVID-19 complications. AMPK activators like capsaicinoids, curcumin, phytoestrogens, cilostazol, and momordicosides have demonstrated the potential to regulate AMPK activity. Compounds from various sources improve fatty acid oxidation and insulin sensitivity, with metformin showing opposing effects on AMPK activation compared to the virus, suggesting potential therapeutic options. The diverse effects of AMPK activation extend to its role in countering viral infections, further highlighting its significance in COVID-19. This review explores AMPK activation mechanisms, its role in metabolic disorders, and the potential use of natural compounds to target AMPK for post-COVID-19 complications. Also, it aims to review the possible methods of activating AMPK to prevent post-COVID-19 diabetes and cardiovascular complications. It also explores the use of natural compounds for their therapeutic effects in targeting the AMPK pathways. Targeting AMPK activation emerges as a promising avenue to mitigate the long-term effects of COVID-19, offering hope for improved patient outcomes and a better quality of life.
Collapse
Affiliation(s)
- Mohammad Saquib Ashraf
- Department of Medical Laboratory Science College of Pharmacy, Nursing and Medical Science Riyadh ELM University, Riyadh, P.O. Box 12734, Saudi Arabia.
| | - Kanika Tuli
- Guru Nanak Institute of Pharmacy, Dalewal, Hoshiarpur 144208, Punjab, India
| | - Shadman Moiz
- Department of Biotechnology, Lalit Narayan Mithila University, Darbhanga 846004, Bihar, India
| | - Satish Kumar Sharma
- Department of Pharmacology, Glocal School of Pharmacy, The Glocal University, Saharanpur, India
| | - Deepa Sharma
- UMM Matrix Innovations Private Limited, Delhi 110044, India
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha'il, Ha'il, P.O. Box 2440, Saudi Arabia
| |
Collapse
|
2
|
Suri C, Pande B, Sahithi LS, Sahu T, Verma HK. Interplay between Lung Diseases and Viral Infections: A Comprehensive Review. Microorganisms 2024; 12:2030. [PMID: 39458339 PMCID: PMC11510474 DOI: 10.3390/microorganisms12102030] [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: 08/25/2024] [Revised: 09/16/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
The intricate relationship between chronic lung diseases and viral infections is a significant concern in respiratory medicine. We explore how pre-existing lung conditions, including chronic obstructive pulmonary disease, asthma, and interstitial lung diseases, influence susceptibility, severity, and outcomes of viral infections. We also examine how viral infections exacerbate and accelerate the progression of lung disease by disrupting immune responses and triggering inflammatory pathways. By summarizing current evidence, this review highlights the bidirectional nature of these interactions, where underlying lung diseasesincrease vulnerability to viral infections, while these infections, in turn, worsen the clinical course. This review underscores the importance of preventive measures, such as vaccination, early detection, and targeted therapies, to mitigate adverse outcomes in patients with chronic lung conditions. The insights provided aim to inform clinical strategies that can improve patient management and reduce the burden of chronic lung diseases exacerbated by viral infections.
Collapse
Affiliation(s)
- Chahat Suri
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada;
| | - Babita Pande
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | | | - Tarun Sahu
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India; (B.P.); (T.S.)
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lungs Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Neuherberg, 85764 Munich, Germany
| |
Collapse
|
3
|
Katusic ZS, d’Uscio LV, He T. Cerebrovascular Endothelial Dysfunction: Role of BACE1. Arterioscler Thromb Vasc Biol 2024; 44:1737-1747. [PMID: 38868939 PMCID: PMC11269044 DOI: 10.1161/atvbaha.124.320798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
Dysfunctional endothelium is increasingly recognized as a mechanistic link between cardiovascular risk factors and dementia, including Alzheimer disease. BACE1 (β-site amyloid-β precursor protein-cleaving enzyme 1) is responsible for β-processing of APP (amyloid-β precursor protein), the first step in the production of Aβ (amyloid-β) peptides, major culprits in the pathogenesis of Alzheimer disease. Under pathological conditions, excessive activation of BACE1 exerts detrimental effects on endothelial function by Aβ-dependent and Aβ-independent mechanisms. High local concentration of Aβ in the brain blood vessels is responsible for the loss of key vascular protective functions of endothelial cells. More recent studies recognized significant contribution of Aβ-independent proteolytic activity of endothelial BACE1 to the pathogenesis of endothelial dysfunction. This review critically evaluates existing evidence supporting the concept that excessive activation of BACE1 expressed in the cerebrovascular endothelium impairs key homeostatic functions of the brain blood vessels. This concept has important therapeutic implications. Indeed, improved understanding of the mechanisms of endothelial dysfunction may help in efforts to develop new approaches to the protection and preservation of healthy cerebrovascular function.
Collapse
Affiliation(s)
- Zvonimir S. Katusic
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota 55902, USA
- Department of Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Livius V. d’Uscio
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota 55902, USA
- Department of Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Tongrong He
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota 55902, USA
- Department of Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota 55902, USA
| |
Collapse
|
4
|
Poyatos P, Luque N, Sabater G, Eizaguirre S, Bonnin M, Orriols R, Tura-Ceide O. Endothelial dysfunction and cardiovascular risk in post-COVID-19 patients after 6- and 12-months SARS-CoV-2 infection. Infection 2024; 52:1269-1285. [PMID: 38324145 PMCID: PMC11289012 DOI: 10.1007/s15010-024-02173-5] [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: 09/18/2023] [Accepted: 01/01/2024] [Indexed: 02/08/2024]
Abstract
INTRODUCTION SARS-CoV-2 infection causes severe endothelial damage, an essential step for cardiovascular complications. Endothelial-colony forming cells (ECFCs) act as a biomarker of vascular damage but their role in SARS-CoV-2 remain unclear. The aim of this study was to assess whether the number of ECFCs and angiogenic biomarkers remained altered after 6 and 12-months post-infection and whether this imbalance correlated with the presence of long-COVID syndrome and other biological parameters measured. METHODS Seventy-two patients were recruited at different time-points after overcoming COVID-19 and thirty-one healthy controls. All subjects were matched for age, gender, BMI, and comorbidities. ECFCs were obtained from peripheral blood and cultured with specific conditions. RESULTS The results confirm the presence of a long-term sequela in post-COVID-19 patients, with an abnormal increase in ECFC production compared to controls (82.8% vs. 48.4%, P < 0.01) that is maintained up to 6-months (87.0% vs. 48.4%, P < 0.01) and 12-months post-infection (85.0% vs. 48.4%, P < 0.01). Interestingly, post-COVID-19 patients showed a significant downregulation of angiogenesis-related proteins compared to controls indicating a clear endothelial injury. Troponin, NT-proBNP and ferritin levels, markers of cardiovascular risk and inflammation, remained elevated up to 12-months post-infection. Patients with lower numbers of ECFC exhibited higher levels of inflammatory markers, such as ferritin, suggesting that ECFCs may play a protective role. Additionally, long-COVID syndrome was associated with higher ferritin levels and with female gender. CONCLUSIONS These findings highlight the presence of vascular sequela that last up to 6- and 12-months post-infection and point out the need for preventive measures and patient follow-up.
Collapse
Affiliation(s)
- Paula Poyatos
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190, Girona, Spain
- Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Neus Luque
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190, Girona, Spain
| | - Gladis Sabater
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190, Girona, Spain
- Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Saioa Eizaguirre
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190, Girona, Spain
- Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Marc Bonnin
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190, Girona, Spain
- Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain
| | - Ramon Orriols
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190, Girona, Spain.
- Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain.
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain.
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Dr. Josep Trueta University Hospital de Girona, Santa Caterina Hospital de Salt and the Girona Biomedical Research Institute (IDIBGI), 17190, Girona, Spain.
- Department of Medical Sciences, Faculty of Medicine, University of Girona, Girona, Spain.
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Madrid, Spain.
- Department of Pulmonary Medicine, Servei de Pneumologia, Hospital Clínic-Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), University of Barcelona, Villarroel, 170, 08036, Barcelona, Spain.
| |
Collapse
|
5
|
Krivdić Dupan Z, Periša V, Suver Stević M, Mihalj M, Tolušić Levak M, Guljaš S, Salha T, Loinjak D, Kos M, Šapina M, Canjko I, Šambić Penc M, Štefančić M, Nešković N. The Impact of Pentraxin 3 Serum Levels and Angiotensin-Converting Enzyme Polymorphism on Pulmonary Infiltrates and Mortality in COVID-19 Patients. Biomedicines 2024; 12:1618. [PMID: 39062191 PMCID: PMC11275229 DOI: 10.3390/biomedicines12071618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
OBJECTIVES The aim of this study was to examine the impact of the pentraxin 3 (PTX3) serum level and angiotensin-converting enzyme (ACE) gene insertion/deletion (I/D) polymorphism on the severity of radiographic pulmonary infiltrates and the clinical outcomes of COVID-19. METHODS The severity of COVID-19 pulmonary infiltrates was evaluated within a week of admission by analyzing chest X-rays (CXR) using the modified Brixia (MBrixa) scoring system. The insertion (I)/deletion (D) polymorphism of the ACE gene and the serum levels of PTX3 were determined for all patients included in the study. RESULTS This study included 80 patients. Using a cut-off serum level of PTX3 ≥ 2.765 ng/mL, the ROC analysis (AUC 0.871, 95% CI 0.787-0.954, p < 0.001) showed a sensitivity of 85.7% and specificity of 78.8% in predicting severe MBrixa scores. Compared to ACE I/I polymorphism, D/D polymorphism significantly increased the risk of severe CXR infiltrates, OR 7.7 (95% CI: 1.9-30.1), and p = 0.002. Significant independent predictors of severe CXR infiltrates include hypertension (OR 7.71), PTX3 (OR 1.20), and ACE D/D polymorphism (OR 18.72). Hypertension (OR 6.91), PTX3 (OR 1.47), and ACE I/I polymorphism (OR 0.09) are significant predictors of poor outcomes. CONCLUSION PTX3 and ACE D/D polymorphism are significant predictors of the severity of COVID-19 pneumonia. PTX3 is a significant predictor of death.
Collapse
Affiliation(s)
- Zdravka Krivdić Dupan
- Department of Radiology, Osijek University Hospital, 31000 Osijek, Croatia
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Vlatka Periša
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Hematology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Mirjana Suver Stević
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Transfusion Medicine, Osijek University Hospital, 31000 Osijek, Croatia
| | - Martina Mihalj
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Dermatology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Maja Tolušić Levak
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Dermatology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Silva Guljaš
- Department of Radiology, Osijek University Hospital, 31000 Osijek, Croatia
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Tamer Salha
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Faculty of Dental Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Domagoj Loinjak
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Internal Medicine, Osijek University Hospital, 31000 Osijek, Croatia
| | - Martina Kos
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Pediatrics, Osijek University Hospital, 31000 Osijek, Croatia
| | - Matej Šapina
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Pediatrics, Osijek University Hospital, 31000 Osijek, Croatia
| | - Ivana Canjko
- Department of Radiotherapy and Oncology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Mirela Šambić Penc
- Department of Radiotherapy and Oncology, Osijek University Hospital, 31000 Osijek, Croatia
| | - Marin Štefančić
- Department of Radiology, National Memorial Hospital Vukovar, 32000 Vukovar, Croatia
| | - Nenad Nešković
- Medical Faculty Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- International Medical Center Priora, 31431 Cepin, Croatia
| |
Collapse
|
6
|
Subramaniam S, Kenney D, Jayaraman A, O’Connell AK, Walachowski S, Montanaro P, Reinhardt C, Colucci G, Crossland NA, Douam F, Bosmann M. Aging is associated with an insufficient early inflammatory response of lung endothelial cells in SARS-CoV-2 infection. Front Immunol 2024; 15:1397990. [PMID: 38911865 PMCID: PMC11190167 DOI: 10.3389/fimmu.2024.1397990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
Advanced age is associated with an increased susceptibility to Coronavirus Disease (COVID)-19 and more severe outcomes, although the underlying mechanisms are understudied. The lung endothelium is located next to infected epithelial cells and bystander inflammation may contribute to thromboinflammation and COVID-19-associated coagulopathy. Here, we investigated age-associated SARS-CoV-2 pathogenesis and endothelial inflammatory responses using humanized K18-hACE2 mice. Survival was reduced to 20% in aged mice (85-112 weeks) versus 50% in young mice (12-15 weeks) at 10 days post infection (dpi). Bulk RNA-sequencing of endothelial cells from mock and infected mice at 2dpi of both age groups (aged: 72-85 weeks; young: 15 weeks) showed substantially lower significant differentially regulated genes in infected aged mice than in young mice (712 versus 2294 genes). Viral recognition and anti-viral pathways such as RIG-I-like receptor signaling, NOD-like receptor signaling and interferon signaling were regulated in response to SARS-CoV-2. Young mice showed several fold higher interferon responses (Ifitm3, Ifit1, Isg15, Stat1) and interferon-induced chemokines (Cxcl10 and Cxcl11) than aged mice. Endothelial cells from infected young mice displayed elevated expression of chemokines (Cxcl9, Ccl2) and leukocyte adhesion markers (Icam1) underscoring that inflammation of lung endothelium during infection could facilitate leukocyte adhesion and thromboinflammation. TREM1 and acute phase response signaling were particularly prominent in endothelial cells from infected young mice. Immunohistochemistry was unable to detect viral protein in pulmonary endothelium. In conclusion, our data demonstrate that the early host response of the endothelium to SARS-CoV-2 infection declines with aging, which could be a potential contributor to disease severity.
Collapse
Affiliation(s)
- Saravanan Subramaniam
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Devin Kenney
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Department of Virology, Immunology and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Archana Jayaraman
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Aoife Kateri O’Connell
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Sarah Walachowski
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Paige Montanaro
- Department of Virology, Immunology and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Giuseppe Colucci
- Outer Corelab, Viollier AG, Allschwil, Switzerland
- Department of Hematology, University of Basel, Basel, Switzerland
| | - Nicholas A. Crossland
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Department of Virology, Immunology and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Florian Douam
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Department of Virology, Immunology and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Markus Bosmann
- Pulmonary Center, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, United States
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
7
|
Kikinis Z, Castañeyra-Perdomo A, González-Mora JL, Rushmore RJ, Toppa PH, Haggerty K, Papadimitriou G, Rathi Y, Kubicki M, Kikinis R, Heller C, Yeterian E, Besteher B, Pallanti S, Makris N. Investigating the structural network underlying brain-immune interactions using combined histopathology and neuroimaging: a critical review for its relevance in acute and long COVID-19. Front Psychiatry 2024; 15:1337888. [PMID: 38590789 PMCID: PMC11000670 DOI: 10.3389/fpsyt.2024.1337888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/23/2024] [Indexed: 04/10/2024] Open
Abstract
Current views on immunity support the idea that immunity extends beyond defense functions and is tightly intertwined with several other fields of biology such as virology, microbiology, physiology and ecology. It is also critical for our understanding of autoimmunity and cancer, two topics of great biological relevance and for critical public health considerations such as disease prevention and treatment. Central to this review, the immune system is known to interact intimately with the nervous system and has been recently hypothesized to be involved not only in autonomic and limbic bio-behaviors but also in cognitive function. Herein we review the structural architecture of the brain network involved in immune response. Furthermore, we elaborate upon the implications of inflammatory processes affecting brain-immune interactions as reported recently in pathological conditions due to SARS-Cov-2 virus infection, namely in acute and post-acute COVID-19. Moreover, we discuss how current neuroimaging techniques combined with ad hoc clinical autopsies and histopathological analyses could critically affect the validity of clinical translation in studies of human brain-immune interactions using neuroimaging. Advances in our understanding of brain-immune interactions are expected to translate into novel therapeutic avenues in a vast array of domains including cancer, autoimmune diseases or viral infections such as in acute and post-acute or Long COVID-19.
Collapse
Affiliation(s)
- Zora Kikinis
- Department of Psychiatry, Psychiatry Neuroimaging Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Agustin Castañeyra-Perdomo
- Universidad de La Laguna, Área de Anatomía y Fisiología. Departamento de Ciencias Médicas Básicas, Facultad de Ciencias de la Salud, San Cristobal de la Laguna, Spain
| | - José Luis González-Mora
- Universidad de La Laguna, Área de Anatomía y Fisiología. Departamento de Ciencias Médicas Básicas, Facultad de Ciencias de la Salud, San Cristobal de la Laguna, Spain
- Universidad de La Laguna, Instituto Universitario de Neurosciencias, Facultad de Ciencias de la Salud, San Cristobal de la Laguna, Spain
| | - Richard Jarrett Rushmore
- Department of Psychiatry, Psychiatry Neuroimaging Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Department of Anatomy and Neurobiology, Boston University School of Medicine, San Cristobal de la Laguna, Spain
- Departments of Psychiatry and Neurology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Poliana Hartung Toppa
- Departments of Psychiatry and Neurology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Kayley Haggerty
- Departments of Psychiatry and Neurology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - George Papadimitriou
- Departments of Psychiatry and Neurology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Yogesh Rathi
- Department of Psychiatry, Psychiatry Neuroimaging Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Departments of Psychiatry and Neurology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Marek Kubicki
- Department of Psychiatry, Psychiatry Neuroimaging Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Departments of Psychiatry and Neurology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Ron Kikinis
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Carina Heller
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Edward Yeterian
- Departments of Psychiatry and Neurology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Psychology, Colby College, Waterville, ME, United States
| | - Bianca Besteher
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - Stefano Pallanti
- Department of Psychiatry and Behavioural Science, Albert Einstein College of Medicine, Bronx, NY, United States
- Istituto di Neuroscienze, Florence, Italy
| | - Nikos Makris
- Department of Psychiatry, Psychiatry Neuroimaging Laboratory, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
- Universidad de La Laguna, Área de Anatomía y Fisiología. Departamento de Ciencias Médicas Básicas, Facultad de Ciencias de la Salud, San Cristobal de la Laguna, Spain
- Universidad de La Laguna, Instituto Universitario de Neurosciencias, Facultad de Ciencias de la Salud, San Cristobal de la Laguna, Spain
- Department of Anatomy and Neurobiology, Boston University School of Medicine, San Cristobal de la Laguna, Spain
- Departments of Psychiatry and Neurology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
8
|
de Azambuja Pias Weber A, Viero FT, Pillat MM, de Lima Gonçalves T. Changes in markers of inflammation and their correlation with death in patients with COVID-19 in the intensive care unit. Cytokine 2024; 175:156509. [PMID: 38241964 DOI: 10.1016/j.cyto.2024.156509] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/07/2023] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
This study aimed to characterize the changes in serum inflammatory mediators in hospitalized patients with COVID-19 correlating with death. The study includes 58 participants: i) inpatients (n = 37): patients suffering from severe acute respiratory syndrome due to COVID-19, who were admitted at Intensive Care Unit (ICU) recovered and who died and ii) control group (n = 21): community volunteers. Inflammatory mediators evaluated interleukin-2 (IL-2), interleukin-4 (IL-4), interleukin-6 (IL-6), interleukin-10 (IL-10), interleukin-17 (IL-17), interferon-gamma (IFN-γ) and interferon-gamma protein levels (IFN-γ), as well as, Urea, LDH, D-dimer, TAP/INR, AST, ALT and lymphocytes. Our results suggest that high levels of inflammatory markers, such as pro-inflammatory cytokines, and laboratory parameters, such as low levels of lymphocytes and high levels of IL-6, are associated with disease severity, especially in individuals who died. Constant measurement and monitoring of these inflammatory parameters is an effective tool in clinical practice, and it can help choosing appropriate therapies during the course of the disease.
Collapse
Affiliation(s)
- Andressa de Azambuja Pias Weber
- Postgraduate Program in Pharmaceutical Sciences, Department of Clinical and Toxicology Analysis, Center of Healthy Sciences, Federal University of Santa Maria (UFSM), Santa Maria, Brazil.
| | - Fernanda Tibolla Viero
- Postgraduate Program in Pharmacology, Department of Microbiology and Parasitology, Center of Healthy Sciences, Federal University of Santa Maria (UFSM), Santa Maria, Brazil.
| | - Micheli Mainardi Pillat
- Postgraduate Program in Pharmacology, Department of Microbiology and Parasitology, Center of Healthy Sciences, Federal University of Santa Maria (UFSM), Santa Maria, Brazil.
| | - Thissiane de Lima Gonçalves
- Postgraduate Program in Pharmaceutical Sciences, Department of Clinical and Toxicology Analysis, Center of Healthy Sciences, Federal University of Santa Maria (UFSM), Santa Maria, Brazil.
| |
Collapse
|
9
|
Maybin J, Watters M, Rowley B, Walker C, Sharp G, Alvergne A. COVID-19 and abnormal uterine bleeding: potential associations and mechanisms. Clin Sci (Lond) 2024; 138:153-171. [PMID: 38372528 PMCID: PMC10876417 DOI: 10.1042/cs20220280] [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: 10/11/2023] [Revised: 01/16/2024] [Accepted: 01/29/2024] [Indexed: 02/20/2024]
Abstract
The impact of COVID-19 on menstruation has received a high level of public and media interest. Despite this, uncertainty exists about the advice that women and people who menstruate should receive in relation to the expected impact of SARS-CoV-2 infection, long COVID or COVID-19 vaccination on menstruation. Furthermore, the mechanisms leading to these reported menstrual changes are poorly understood. This review evaluates the published literature on COVID-19 and its impact on menstrual bleeding, discussing the strengths and limitations of these studies. We present evidence consistent with SARS-CoV-2 infection and long COVID having an association with changes in menstrual bleeding parameters and that the impact of COVID vaccination on menstruation appears less significant. An overview of menstrual physiology and known causes of abnormal uterine bleeding (AUB) is provided before discussing potential mechanisms which may underpin the menstrual disturbance reported with COVID-19, highlighting areas for future scientific study. Finally, consideration is given to the effect that menstruation may have on COVID-19, including the impact of the ovarian sex hormones on acute COVID-19 severity and susceptibility and reported variation in long COVID symptoms across the menstrual cycle. Understanding the current evidence and addressing gaps in our knowledge in this area are essential to inform public health policy, direct the treatment of menstrual disturbance and facilitate development of new therapies, which may reduce the severity of COVID-19 and improve quality of life for those experiencing long COVID.
Collapse
Affiliation(s)
- Jacqueline A. Maybin
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, U.K
| | - Marianne Watters
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, U.K
| | - Bethan Rowley
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, U.K
| | - Catherine A. Walker
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, U.K
| | | | - Alexandra Alvergne
- ISEM, Univ Montpellier, CNRS, IRD, Montpellier, France
- School of Anthropology and Museum Ethnography, Oxford, U.K
| |
Collapse
|
10
|
Agoston DV. Traumatic Brain Injury in the Long-COVID Era. Neurotrauma Rep 2024; 5:81-94. [PMID: 38463416 PMCID: PMC10923549 DOI: 10.1089/neur.2023.0067] [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] [Indexed: 03/12/2024] Open
Abstract
Major determinants of the biological background or reserve, such as age, biological sex, comorbidities (diabetes, hypertension, obesity, etc.), and medications (e.g., anticoagulants), are known to affect outcome after traumatic brain injury (TBI). With the unparalleled data richness of coronavirus disease 2019 (COVID-19; ∼375,000 and counting!) as well as the chronic form, long-COVID, also called post-acute sequelae SARS-CoV-2 infection (PASC), publications (∼30,000 and counting) covering virtually every aspect of the diseases, pathomechanisms, biomarkers, disease phases, symptomatology, etc., have provided a unique opportunity to better understand and appreciate the holistic nature of diseases, interconnectivity between organ systems, and importance of biological background in modifying disease trajectories and affecting outcomes. Such a holistic approach is badly needed to better understand TBI-induced conditions in their totality. Here, I briefly review what is known about long-COVID/PASC, its underlying-suspected-pathologies, the pathobiological changes induced by TBI, in other words, the TBI endophenotypes, discuss the intersection of long-COVID/PASC and TBI-induced pathobiologies, and how by considering some of the known factors affecting the person's biological background and the inclusion of mechanistic molecular biomarkers can help to improve the clinical management of TBI patients.
Collapse
Affiliation(s)
- Denes V. Agoston
- Department of Anatomy, Physiology, and Genetics, School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| |
Collapse
|
11
|
Ansone L, Rovite V, Brīvība M, Jagare L, Pelcmane L, Borisova D, Thews A, Leiminger R, Kloviņš J. Longitudinal NMR-Based Metabolomics Study Reveals How Hospitalized COVID-19 Patients Recover: Evidence of Dyslipidemia and Energy Metabolism Dysregulation. Int J Mol Sci 2024; 25:1523. [PMID: 38338803 PMCID: PMC10855192 DOI: 10.3390/ijms25031523] [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: 11/26/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Long COVID, or post-acute sequelae of SARS-CoV-2 infection (PASC), can manifest as long-term symptoms in multiple organ systems, including respiratory, cardiovascular, neurological, and metabolic systems. In patients with severe COVID-19, immune dysregulation is significant, and the relationship between metabolic regulation and immune response is of great interest in determining the pathophysiological mechanisms. We aimed to characterize the metabolomic footprint of recovering severe COVID-19 patients at three consecutive timepoints and compare metabolite levels to controls. Our findings add proof of dysregulated amino acid metabolism in the acute phase and dyslipidemia, glycoprotein level alterations, and energy metabolism disturbances in severe COVID-19 patients 3-4 months post-hospitalization.
Collapse
Affiliation(s)
- Laura Ansone
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (V.R.); (M.B.); (L.J.); (L.P.); (D.B.)
| | - Vita Rovite
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (V.R.); (M.B.); (L.J.); (L.P.); (D.B.)
| | - Monta Brīvība
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (V.R.); (M.B.); (L.J.); (L.P.); (D.B.)
| | - Lauma Jagare
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (V.R.); (M.B.); (L.J.); (L.P.); (D.B.)
| | - Līva Pelcmane
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (V.R.); (M.B.); (L.J.); (L.P.); (D.B.)
| | - Daniella Borisova
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (V.R.); (M.B.); (L.J.); (L.P.); (D.B.)
| | - Anne Thews
- Bruker BioSpin GmbH & Co., Rudolf-Plank-Straße 23, 76275 Ettlingen, Germany; (A.T.); (R.L.)
| | - Roland Leiminger
- Bruker BioSpin GmbH & Co., Rudolf-Plank-Straße 23, 76275 Ettlingen, Germany; (A.T.); (R.L.)
| | - Jānis Kloviņš
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (V.R.); (M.B.); (L.J.); (L.P.); (D.B.)
| |
Collapse
|
12
|
S B MJ, Chacko B, Selvarajan S, Peter JV, Geevar T, Dave RG, Georgy JT, Zachariah A, George T, Sathyendra S, Hansdak SG, Krishnaswami RK, Thangakunam B, Gupta R, Karuppusami R, Nair SC, Srivastava A. Biomarkers of coagulation, endothelial, platelet function, and fibrinolysis in patients with COVID-19: a prospective study. Sci Rep 2024; 14:2011. [PMID: 38263377 PMCID: PMC10805716 DOI: 10.1038/s41598-024-51908-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/11/2024] [Indexed: 01/25/2024] Open
Abstract
Prospective and sequential evaluation of homeostatic changes leading to thrombosis across COVID 19 disease severity spectrum are limited. In this prospective observational study, haemostasis was evaluated in patients with mild, moderate-severe, and critical COVID-19 infection. Markers of endothelial activation [Soluble thrombomodulin (sTM), von Willebrand Factor (VWF)], platelet activation [Soluble P-selectin, beta-thromboglobulin (BTG)] and global haemostasis [Rotational thromboelastometry (ROTEM)] were evaluated on days 1 and 5 after admission. The study cohort comprised of 100 adult patients (mild = 20, moderate-severe = 22, critical = 58). Sixty-five patients received anticoagulation for 10 (7-14) days. Thrombotic events were seen in 9 patients. In-hospital mortality was 21%. Endothelial activation markers were elevated at baseline in all subgroups, with levels in moderate-severe (sTM = 4.92 ng/ml, VWF = 295 U/dl) [reference-ranges: sTM = 2.26-4.55 ng/ml; Soluble P-selectin = 13.5-31.5 ng/ml; BTG = 0.034-1.99 ng/ml] and critical patients (sTM = 6.07 ng/ml, VWF = 294 U/dl) being significantly higher than in the mild group (sTM = 4.18 ng/ml, VWF = 206 U/dl). In contrast, platelet activation markers were elevated only in critically ill patients at baseline (Soluble P-selectin = 37.3 ng/ml, BTG = 2.51 ng/ml). The critical group had significantly lower fibrinolysis on days 1 and 5 when compared with the moderate-severe arm. COVID-19 infection was associated with graded endothelial activation and lower fibrinolysis that correlated with illness severity.
Collapse
Affiliation(s)
- Manoj Job S B
- Department of Critical Care, Christian Medical College, Vellore, 632004, India.
| | - Binila Chacko
- Department of Critical Care, Christian Medical College, Vellore, 632004, India
| | - Sushil Selvarajan
- Department of Clinical Hematology, Christian Medical College, Vellore, India
| | - John Victor Peter
- Department of Critical Care, Christian Medical College, Vellore, 632004, India
| | - Tulasi Geevar
- Department of Transfusion Medicine, Christian Medical College, Vellore, India
| | - Rutvi Gautam Dave
- Department of Transfusion Medicine, Christian Medical College, Vellore, India
| | - Josh Thomas Georgy
- Department of General Medicine, Christian Medical College, Vellore, India
| | - Anand Zachariah
- Department of General Medicine, Christian Medical College, Vellore, India
| | - Tina George
- Department of General Medicine, Christian Medical College, Vellore, India
| | - Sowmya Sathyendra
- Department of General Medicine, Christian Medical College, Vellore, India
| | | | | | | | - Richa Gupta
- Department of Respiratory Medicine, Christian Medical College, Vellore, India
| | - Reka Karuppusami
- Department of Biostatistics, Christian Medical College, Vellore, India
| | | | - Alok Srivastava
- Department of Clinical Hematology, Christian Medical College, Vellore, India
| |
Collapse
|
13
|
Liu Y, Gu X, Li H, Zhang H, Xu J. Mechanisms of long COVID: An updated review. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:231-240. [PMID: 39171285 PMCID: PMC11332859 DOI: 10.1016/j.pccm.2023.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Indexed: 08/23/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has been ongoing for more than 3 years, with an enormous impact on global health and economies. In some patients, symptoms and signs may remain after recovery from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, which cannot be explained by an alternate diagnosis; this condition has been defined as long COVID. Long COVID may exist in patients with both mild and severe disease and is prevalent after infection with different SARS-CoV-2 variants. The most common symptoms include fatigue, dyspnea, and other symptoms involving multiple organs. Vaccination results in lower rates of long COVID. To date, the mechanisms of long COVID remain unclear. In this narrative review, we summarized the clinical presentations and current evidence regarding the pathogenesis of long COVID.
Collapse
Affiliation(s)
- Yan Liu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing 100029, China
- Department of Infectious Disease, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Xiaoying Gu
- Department of Clinical Research and Data Management, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China
| | - Haibo Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing 100029, China
| | - Hui Zhang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing 100029, China
- Department of Pulmonary and Critical Care Medicine, China–Japan Friendship Hospital, Capital Medical University, Beijing 100029, China
| | - Jiuyang Xu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China–Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
14
|
Negi V, Gavlock D, Miedel MT, Lee JK, Shun T, Gough A, Vernetti L, Stern AM, Taylor DL, Yechoor VK. Modeling mechanisms underlying differential inflammatory responses to COVID-19 in type 2 diabetes using a patient-derived microphysiological organ-on-a-chip system. LAB ON A CHIP 2023; 23:4514-4527. [PMID: 37766577 DOI: 10.1039/d3lc00285c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
Background: COVID-19 pandemic has caused more than 6 million deaths worldwide. Co-morbid conditions such as Type 2 Diabetes (T2D) have increased mortality in COVID-19. With limited translatability of in vitro and small animal models to human disease, human organ-on-a-chip models are an attractive platform to model in vivo disease conditions and test potential therapeutics. Methods: T2D or non-diabetic patient-derived macrophages and human liver sinusoidal endothelial cells were seeded, along with normal hepatocytes and stellate cells in the liver-on-a-chip (LAMPS - liver acinus micro physiological system), perfused with media mimicking non-diabetic fasting or T2D (high levels of glucose, fatty acids, insulin, glucagon) states. The macrophages and endothelial cells were transduced to overexpress the SARS-CoV2-S (spike) protein with appropriate controls before their incorporation into LAMPS. Cytokine concentrations in the efflux served as a read-out of the effects of S-protein expression in the different experimental conditions (non-diabetic-LAMPS, T2D-LAMPS), including incubation with tocilizumab, an FDA-approved drug for severe COVID-19. Findings: S-protein expression in the non-diabetic LAMPS led to increased cytokines, but in the T2D-LAMPS, this was significantly amplified both in the number and magnitude of key pro-inflammatory cytokines (IL6, CCL3, IL1β, IL2, TNFα, etc.) involved in cytokine storm syndrome (CSS), mimicking severe COVID-19 infection in T2D patients. Compared to vehicle control, tocilizumab (IL6-receptor antagonist) decreased the pro-inflammatory cytokine secretion in T2D-COVID-19-LAMPS but not in non-diabetic-COVID-19-LAMPS. Interpretation: macrophages and endothelial cells play a synergistic role in the pathophysiology of the hyper-inflammatory response seen with COVID-19 and T2D. The effect of Tocilizumab was consistent with large clinical trials that demonstrated Tocilizumab's efficacy only in critically ill patients with severe disease, providing confirmatory evidence that the T2D-COVID-19-LAMPS is a robust platform to model human in vivo pathophysiology of COVID-19 in T2D and for screening potential therapeutics.
Collapse
Affiliation(s)
- Vinny Negi
- Diabetes and Beta Cell Biology Center, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Dillon Gavlock
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mark T Miedel
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeong Kyung Lee
- Diabetes and Beta Cell Biology Center, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Tongying Shun
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Albert Gough
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lawrence Vernetti
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andrew M Stern
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - D Lansing Taylor
- Drug Discovery Institute and Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Vijay K Yechoor
- Diabetes and Beta Cell Biology Center, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
Greistorfer T, Jud P. Pathophysiological Aspects of COVID-19-Associated Vasculopathic Diseases. Thromb Haemost 2023; 123:931-944. [PMID: 37172941 DOI: 10.1055/s-0043-1768969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Since the beginning of coronavirus disease 2019 (COVID-19) pandemic, numerous data reported potential effects on the cardiovascular system due to infection by severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), which may lead to COVID-19-associated vasculopathies during the acute phase and measurable vascular changes in the convalescent phase. Infection by SARS-CoV-2 seems to have specific direct and indirect effects on the endothelium, immune and coagulation systems thus promoting endothelial dysfunction, immunothrombosis, and formation of neutrophil extracellular traps although the exact mechanisms still need to be elucidated. This review represents a recent update of pathophysiological pathways of the respective three major mechanisms contributing to COVID-19 vasculopathies and vascular changes and includes clinical implications and significance of outcome data.
Collapse
Affiliation(s)
- Thiemo Greistorfer
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Philipp Jud
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
16
|
Muyayalo KP, Gong GS, Kiyonga Aimeé K, Liao AH. Impaired immune response against SARS-CoV-2 infection is the major factor indirectly altering reproductive function in COVID-19 patients: a narrative review. HUM FERTIL 2023; 26:778-796. [PMID: 37811836 DOI: 10.1080/14647273.2023.2262757] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/25/2023] [Indexed: 10/10/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease affecting multiple systems and organs, including the reproductive system. SARS-CoV-2, the virus that causes COVID-19, can damage reproductive organs through direct (angiotensin converting enzyme-2, ACE-2) and indirect mechanisms. The immune system plays an essential role in the homeostasis and function of the male and female reproductive systems. Therefore, an altered immune response related to infectious and inflammatory diseases can affect reproductive function and fertility in both males and females. This narrative review discussed the dysregulation of innate and adaptive systems induced by SARS-CoV-2 infection. We reviewed the evidence showing that this altered immune response in COVID-19 patients is the major indirect mechanism leading to adverse reproduction outcomes in these patients. We summarized studies reporting the long-term effect of SARS-CoV-2 infection on women's reproductive function and proposed the chronic inflammation and chronic autoimmunity characterizing long COVID as potential underlying mechanisms. Further studies are needed to clarify the role of autoimmunity and chronic inflammation (long COVID) in altered female reproduction function in COVID-19.
Collapse
Affiliation(s)
- Kahindo P Muyayalo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
- Department of Obstetrics and Gynecology, University of Kinshasa, Kinshasa, D. R. Congo
| | - Guang-Shun Gong
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Kahindo Kiyonga Aimeé
- Department of Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, People's Republic of China
- Department of Tropical Medicine Infectious and Parasitic Diseases, University of Kinshasa, Kinshasa, D. R. Congo
| | - Ai-Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| |
Collapse
|
17
|
Andreeva VD, Ehlers H, R C AK, Presselt M, J van den Broek L, Bonnet S. Combining nitric oxide and calcium sensing for the detection of endothelial dysfunction. Commun Chem 2023; 6:179. [PMID: 37644120 PMCID: PMC10465535 DOI: 10.1038/s42004-023-00973-8] [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: 02/01/2023] [Accepted: 08/01/2023] [Indexed: 08/31/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide and are not typically diagnosed until the disease has manifested. Endothelial dysfunction is an early, reversible precursor in the irreversible development of cardiovascular diseases and is characterized by a decrease in nitric oxide production. We believe that more reliable and reproducible methods are necessary for the detection of endothelial dysfunction. Both nitric oxide and calcium play important roles in the endothelial function. Here we review different types of molecular sensors used in biological settings. Next, we review the current nitric oxide and calcium sensors available. Finally, we review methods for using both sensors for the detection of endothelial dysfunction.
Collapse
Affiliation(s)
| | - Haley Ehlers
- Mimetas B.V., De limes 7, 2342 DH, Oegstgeest, The Netherlands
- Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Aswin Krishna R C
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Martin Presselt
- Leibniz Institute of Photonic Technology (Leibniz-IPHT), Albert-Einstein-Str. 9, 07745, Jena, Germany
- Sciclus GmbH & Co. KG, Moritz-von-Rohr-Str. 1a, 07745, Jena, Germany
| | | | - Sylvestre Bonnet
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
18
|
Bele A, Wagh V, Munjewar PK. A Comprehensive Review on Cardiovascular Complications of COVID-19: Unraveling the Link to Bacterial Endocarditis. Cureus 2023; 15:e44019. [PMID: 37746510 PMCID: PMC10517725 DOI: 10.7759/cureus.44019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
The global pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has ushered in a new era of understanding the multifaceted nature of infectious diseases. Beyond its well-documented respiratory impact, COVID-19 has unveiled intricate interactions with the cardiovascular system, with potential implications that extend to bacterial endocarditis. This review explores the complex interplay between COVID-19 and bacterial endocarditis, elucidating shared risk factors, theoretical mechanisms, and clinical implications. We examine the diverse cardiovascular manifestations of COVID-19, ranging from myocarditis and thromboembolic events to arrhythmias, and delve into the pathogenesis, clinical features, and diagnostic challenges of bacterial endocarditis. By analyzing potential connections, such as viral-induced endothelial disruption and immune modulation, we shed light on the plausible relationship between COVID-19 and bacterial endocarditis. Our synthesis highlights the significance of accurate diagnosis, optimal management, and interdisciplinary collaboration in addressing the challenges posed by these intricate interactions. In addition, we underscore the importance of future research, emphasizing prospective studies on bacterial endocarditis incidence and investigations into the long-term cardiovascular effects of COVID-19. As the boundaries of infectious diseases and cardiovascular complications converge, this review calls for continued research, vigilance, and coordinated efforts to enhance patient care and public health strategies in a rapidly evolving landscape.
Collapse
Affiliation(s)
- Anurag Bele
- Internal Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Vasant Wagh
- Community Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Pratiksha K Munjewar
- Medical Surgical Nursing, Smt. Radhikabai Meghe Memorial College of Nursing, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
19
|
Salvucci F, Codella R, Coppola A, Zacchei I, Grassi G, Anti ML, Nitisoara N, Luzi L, Gazzaruso C. Antihistamines improve cardiovascular manifestations and other symptoms of long-COVID attributed to mast cell activation. Front Cardiovasc Med 2023; 10:1202696. [PMID: 37529714 PMCID: PMC10388239 DOI: 10.3389/fcvm.2023.1202696] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/06/2023] [Indexed: 08/03/2023] Open
Abstract
Introduction Long-COVID is a broadly defined condition and there are no effective therapies. Cardiovascular manifestations of long-COVID include high heart rate, postural tachycardia, and palpitations. Previous studies have suggested that mast cell activation (MCA) may play a role in the pathophysiology of long-COVID, including in the mechanisms of its cardiovascular manifestations. The present study aimed to evaluate the effectiveness of a treatment with blockers of histamine receptors in patients with long-COVID who did not respond to other therapies. Methods In all, 14 patients (F/M = 9/5; 49.5 ± 11.5 years) and 13 controls (F/M = 8/5; 47.3 ± 8.0 years) with long-COVID symptoms attributed to MCA were evaluated. Patients were treated with fexofenadine (180 mg/day) and famotidine (40 mg/day). Fatigue, brain fog, abdominal disorders, and increased heart rate were evaluated in treated and untreated patients at baseline and 20 days later. Results Long-COVID symptoms disappeared completely in 29% of treated patients. There was a significant improvement in each of the considered symptoms (improved or disappeared) in all treated patients, and the improvement grade was significantly greater in treated patients compared to controls. No significant differences in the outcomes were observed in the controls. Conclusions Our data confirm that histamine receptors blockade may be an effective target to successfully treat long-COVID. Our finding supports the underlying role of MCA in the pathophysiology of long-COVID.
Collapse
Affiliation(s)
- Fabrizio Salvucci
- Internal Medicine, Clinica Santa Rita del Gruppo Policlinico di Monza, Vercelli, Italy
| | - Roberto Codella
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Adriana Coppola
- Centre for Applied Clinical Research (Ce.R.C.A.), Istituto Clinico Beato Matteo, Vigevano, Italy
| | - Irene Zacchei
- Cardiovascular and Metabolic Department, Ticinello Cardiovascular and Metabolic Centre, Pavia, Italy
| | - Gabriella Grassi
- Cardiovascular and Metabolic Department, Ticinello Cardiovascular and Metabolic Centre, Pavia, Italy
| | - Maria Luisa Anti
- Cardiovascular and Metabolic Department, Ticinello Cardiovascular and Metabolic Centre, Pavia, Italy
| | - Nicolita Nitisoara
- Internal Medicine, Clinica Santa Rita del Gruppo Policlinico di Monza, Vercelli, Italy
| | - Livio Luzi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- Department of Endocrinology, IRCCS Multimedica, Milan, Italy
| | - Carmine Gazzaruso
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| |
Collapse
|
20
|
Rostamzadeh F, Najafipour H, Nakhaei S, Yazdani R, Langari AA. Low Ang-(1-7) and high des-Arg9 bradykinin serum levels are correlated with cardiovascular risk factors in patients with COVID-19. Open Med (Wars) 2023; 18:20230741. [PMID: 37415613 PMCID: PMC10320568 DOI: 10.1515/med-2023-0741] [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: 01/30/2023] [Revised: 05/31/2023] [Accepted: 05/31/2023] [Indexed: 07/08/2023] Open
Abstract
It is predictable that the renin-angiotensin-aldosterone and kinin-kallikrein systems are dysregulated in COVID-19 (COV) patients because SARS-CoV-2 requires ACE2 to cause an infection. This study aimed to assess the serum levels of des-arg(9)-bradykinin (DABK) and angiotensin 1-7 (ang-(1-7)) in patients with COV who had the above-mentioned cardiovascular disease risk factors. In a cross-sectional study, 69 COV patients were selected among patients referred to the main referral center for these patients, in Kerman, Iran, and 73 matched control (non-COV) individuals among individuals who participated in the KERCARD cohort study. Serum levels of DABK and ang-(1-7) were measured by ELISA in the groups of CTL (healthy), HTN, DM, OB, COV, COV + HTN, COV + DM, and COV + OB. Ang-(1-7) levels were lower in the COV + HTN group compared to the HTN group. DABK levels were higher in the COV, HTN, and OB groups and in DM + COV subjects compared to their corresponding control group. The levels of ang-(1-7) and DABK were related to HTN and OB, respectively. According to the findings, we can infer that an increase in DABK production in those with the cardiovascular disease risk factors of diabetes, obesity, and hypertension or a decrease in ang-(1-7) in those with hypertension may contribute to the adverse outcomes of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Farzaneh Rostamzadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Najafipour
- Cardiovascular and Respiratory Physiology, Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Samira Nakhaei
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Rostam Yazdani
- Department of Internal Medicine, School of Medicine, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Ahmad Alinaghi Langari
- Gastroenterology, and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
21
|
Xu SW, Ilyas I, Weng JP. Endothelial dysfunction in COVID-19: an overview of evidence, biomarkers, mechanisms and potential therapies. Acta Pharmacol Sin 2023; 44:695-709. [PMID: 36253560 PMCID: PMC9574180 DOI: 10.1038/s41401-022-00998-0] [Citation(s) in RCA: 132] [Impact Index Per Article: 132.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 09/11/2022] [Indexed: 12/15/2022] Open
Abstract
The fight against coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 infection is still raging. However, the pathophysiology of acute and post-acute manifestations of COVID-19 (long COVID-19) is understudied. Endothelial cells are sentinels lining the innermost layer of blood vessel that gatekeep micro- and macro-vascular health by sensing pathogen/danger signals and secreting vasoactive molecules. SARS-CoV-2 infection primarily affects the pulmonary system, but accumulating evidence suggests that it also affects the pan-vasculature in the extrapulmonary systems by directly (via virus infection) or indirectly (via cytokine storm), causing endothelial dysfunction (endotheliitis, endothelialitis and endotheliopathy) and multi-organ injury. Mounting evidence suggests that SARS-CoV-2 infection leads to multiple instances of endothelial dysfunction, including reduced nitric oxide (NO) bioavailability, oxidative stress, endothelial injury, glycocalyx/barrier disruption, hyperpermeability, inflammation/leukocyte adhesion, senescence, endothelial-to-mesenchymal transition (EndoMT), hypercoagulability, thrombosis and many others. Thus, COVID-19 is deemed as a (micro)vascular and endothelial disease. Of translational relevance, several candidate drugs which are endothelial protective have been shown to improve clinical manifestations of COVID-19 patients. The purpose of this review is to provide a latest summary of biomarkers associated with endothelial cell activation in COVID-19 and offer mechanistic insights into the molecular basis of endothelial activation/dysfunction in macro- and micro-vasculature of COVID-19 patients. We envisage further development of cellular models and suitable animal models mimicking endothelial dysfunction aspect of COVID-19 being able to accelerate the discovery of new drugs targeting endothelial dysfunction in pan-vasculature from COVID-19 patients.
Collapse
Affiliation(s)
- Suo-Wen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230001, China.
| | - Iqra Ilyas
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230001, China
| | - Jian-Ping Weng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
22
|
Naz A, Asif S, Alwutayd KM, Sarfaraz S, Abbasi SW, Abbasi A, Alenazi AM, Hasan ME. Repurposing FIASMAs against Acid Sphingomyelinase for COVID-19: A Computational Molecular Docking and Dynamic Simulation Approach. Molecules 2023; 28:molecules28072989. [PMID: 37049752 PMCID: PMC10096053 DOI: 10.3390/molecules28072989] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Over the past few years, COVID-19 has caused widespread suffering worldwide. There is great research potential in this domain and it is also necessary. The main objective of this study was to identify potential inhibitors against acid sphingomyelinase (ASM) in order to prevent coronavirus infection. Experimental studies revealed that SARS-CoV-2 causes activation of the acid sphingomyelinase/ceramide pathway, which in turn facilitates the viral entry into the cells. The objective was to inhibit acid sphingomyelinase activity in order to prevent the cells from SARS-CoV-2 infection. Previous studies have reported functional inhibitors against ASM (FIASMAs). These inhibitors can be exploited to block the entry of SARS-CoV-2 into the cells. To achieve our objective, a drug library containing 257 functional inhibitors of ASM was constructed. Computational molecular docking was applied to dock the library against the target protein (PDB: 5I81). The potential binding site of the target protein was identified through structural alignment with the known binding pocket of a protein with a similar function. AutoDock Vina was used to carry out the docking steps. The docking results were analyzed and the inhibitors were screened based on their binding affinity scores and ADME properties. Among the 257 functional inhibitors, Dutasteride, Cepharanthine, and Zafirlukast presented the lowest binding affinity scores of −9.7, −9.6, and −9.5 kcal/mol, respectively. Furthermore, computational ADME analysis of these results revealed Cepharanthine and Zafirlukast to have non-toxic properties. To further validate these findings, the top two inhibitors in complex with the target protein were subjected to molecular dynamic simulations at 100 ns. The molecular interactions and stability of these compounds revealed that these inhibitors could be a promising tool for inhibiting SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Aliza Naz
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad 45320, Pakistan
- Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad 44000, Pakistan
| | - Sumbul Asif
- Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad 44000, Pakistan
- School of Interdisciplinary Engineering and Sciences, National University of Sciences and Technology, Islamabad 44000, Pakistan
| | - Khairiah Mubarak Alwutayd
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Sara Sarfaraz
- Department of Bioinformatics, Kohsar University Murree, Murree 47150, Pakistan
- Correspondence:
| | - Sumra Wajid Abbasi
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Asim Abbasi
- Department of Environmental Sciences, Kohsar University Murree, Murree 47150, Pakistan
| | - Abdulkareem M. Alenazi
- Pediatric Senior Registrar, King Salman Armed Forces Hospital in Northwestern Region (KSAFH), Tabuk 47512, Saudi Arabia
| | - Mohamed E. Hasan
- Bioinformatic Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City 32897, Egypt
| |
Collapse
|
23
|
COVID-19 and Pulmonary Angiogenesis: The Possible Role of Hypoxia and Hyperinflammation in the Overexpression of Proteins Involved in Alveolar Vascular Dysfunction. Viruses 2023; 15:v15030706. [PMID: 36992415 PMCID: PMC10057465 DOI: 10.3390/v15030706] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023] Open
Abstract
COVID-19 has been considered a vascular disease, and inflammation, intravascular coagulation, and consequent thrombosis may be associated with endothelial dysfunction. These changes, in addition to hypoxia, may be responsible for pathological angiogenesis. This research investigated the impact of COVID-19 on vascular function by analyzing post-mortem lung samples from 24 COVID-19 patients, 10 H1N1pdm09 patients, and 11 controls. We evaluated, through the immunohistochemistry technique, the tissue immunoexpressions of biomarkers involved in endothelial dysfunction, microthrombosis, and angiogenesis (ICAM-1, ANGPT-2, and IL-6, IL-1β, vWF, PAI-1, CTNNB-1, GJA-1, VEGF, VEGFR-1, NF-kB, TNF-α and HIF-1α), along with the histopathological presence of microthrombosis, endothelial activation, and vascular layer hypertrophy. Clinical data from patients were also observed. The results showed that COVID-19 was associated with increased immunoexpression of biomarkers involved in endothelial dysfunction, microthrombosis, and angiogenesis compared to the H1N1 and CONTROL groups. Microthrombosis and vascular layer hypertrophy were found to be more prevalent in COVID-19 patients. This study concluded that immunothrombosis and angiogenesis might play a key role in COVID-19 progression and outcome, particularly in patients who die from the disease.
Collapse
|
24
|
Treating COVID-19: Targeting the Host Response, Not the Virus. Life (Basel) 2023; 13:life13030712. [PMID: 36983871 PMCID: PMC10054780 DOI: 10.3390/life13030712] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/19/2023] [Accepted: 01/31/2023] [Indexed: 03/09/2023] Open
Abstract
In low- and middle-income countries (LMICs), inexpensive generic drugs like statins, ACE inhibitors, and ARBs, especially if used in combination, might be the only practical way to save the lives of patients with severe COVID-19. These drugs will already be available in all countries on the first pandemic day. Because they target the host response to infection instead of the virus, they could be used to save lives during any pandemic. Observational studies show that inpatient statin treatment reduces 28–30-day mortality but randomized controlled trials have failed to show this benefit. Combination treatment has been tested for antivirals and dexamethasone but, with the exception of one observational study in Belgium, not for inexpensive generic drugs. Future pandemic research must include testing combination generic drug treatments that could be used in LMICs.
Collapse
|
25
|
Beneficial Effects of L-Arginine in Patients Hospitalized for COVID-19: New Insights from a Randomized Clinical Trial. Pharmacol Res 2023; 191:106702. [PMID: 36804278 PMCID: PMC9928676 DOI: 10.1016/j.phrs.2023.106702] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
We have recently demonstrated in a double-blind randomized trial the beneficial effects of L-Arginine in patients hospitalized for COVID-19. We hypothesize that one of the mechanisms underlying the favorable effects of L-Arginine is its action on inflammatory cytokines. To verify our hypothesis, we measured longitudinal plasma levels of pro-inflammatory and anti-inflammatory cytokines implied in the pathophysiology of COVID-19 in patients randomized to receive oral L-Arginine or placebo. The study was successfully completed by 169 patients. Patients in the L-Arginine arm had a reduced respiratory support evaluated at 10 and 20 days; moreover, the time to hospital discharge was significantly shorter in the L-Arginine group. The assessment of circulating cytokines revealed that L-Arginine significantly reduced the circulating levels of pro-inflammatory IL-2, IL-6, and IFN-γ and increased the levels of the anti-inflammatory IL-10. Taken together, these findings indicate that adding L-Arginine to standard therapy in COVID-19 patients markedly reduces the need of respiratory support and the duration of in-hospital stay; moreover, L-Arginine significantly regulates circulating levels of pro-inflammatory and anti-inflammatory cytokines.
Collapse
|
26
|
Cardiovascular and Renal Comorbidities Included into Neural Networks Predict the Outcome in COVID-19 Patients Admitted to an Intensive Care Unit: Three-Center, Cross-Validation, Age- and Sex-Matched Study. J Cardiovasc Dev Dis 2023; 10:jcdd10020039. [PMID: 36826535 PMCID: PMC9967447 DOI: 10.3390/jcdd10020039] [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: 11/28/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/25/2023] Open
Abstract
Here, we performed a multicenter, age- and sex-matched study to compare the efficiency of various machine learning algorithms in the prediction of COVID-19 fatal outcomes and to develop sensitive, specific, and robust artificial intelligence tools for the prompt triage of patients with severe COVID-19 in the intensive care unit setting. In a challenge against other established machine learning algorithms (decision trees, random forests, extra trees, neural networks, k-nearest neighbors, and gradient boosting: XGBoost, LightGBM, and CatBoost) and multivariate logistic regression as a reference, neural networks demonstrated the highest sensitivity, sufficient specificity, and excellent robustness. Further, neural networks based on coronary artery disease/chronic heart failure, stage 3-5 chronic kidney disease, blood urea nitrogen, and C-reactive protein as the predictors exceeded 90% sensitivity and 80% specificity, reaching AUROC of 0.866 at primary cross-validation and 0.849 at secondary cross-validation on virtual samples generated by the bootstrapping procedure. These results underscore the impact of cardiovascular and renal comorbidities in the context of thrombotic complications characteristic of severe COVID-19. As aforementioned predictors can be obtained from the case histories or are inexpensive to be measured at admission to the intensive care unit, we suggest this predictor composition is useful for the triage of critically ill COVID-19 patients.
Collapse
|
27
|
Ambrosino P, Moretta P, Lanzillo A, Formisano R, Maniscalco M. Implementing Translational Research to Understand the Future of COVID-19 and Its Long-Term Consequences: A Degrowth Perspective or the Transformation of a Global Emergency? Biomedicines 2023; 11:117. [PMID: 36672625 PMCID: PMC9855765 DOI: 10.3390/biomedicines11010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/05/2023] Open
Abstract
It has now been three years since the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) first gave rise to a global health crisis [...].
Collapse
Affiliation(s)
- Pasquale Ambrosino
- Istituti Clinici Scientifici Maugeri IRCCS, Directorate of Telese Terme Institute, 82037 Telese Terme, Italy
| | - Pasquale Moretta
- Istituti Clinici Scientifici Maugeri IRCCS, Neuromotor Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy
| | - Anna Lanzillo
- Istituti Clinici Scientifici Maugeri IRCCS, Neuromotor Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy
| | - Roberto Formisano
- Istituti Clinici Scientifici Maugeri IRCCS, Cardiac Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy
| | - Mauro Maniscalco
- Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| |
Collapse
|
28
|
Ambrosino P, Sanduzzi Zamparelli S, Mosella M, Formisano R, Molino A, Spedicato GA, Papa A, Motta A, Di Minno MND, Maniscalco M. Clinical assessment of endothelial function in convalescent COVID-19 patients: a meta-analysis with meta-regressions. Ann Med 2022; 54:3234-3249. [PMID: 36382632 PMCID: PMC9673781 DOI: 10.1080/07853890.2022.2136403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Endothelial dysfunction has been proposed to play a key role in the pathogenesis of coronavirus disease 2019 (COVID-19) and its post-acute sequelae. Flow-mediated dilation (FMD) is recognized as an accurate clinical method to assess endothelial function. Thus, we performed a meta-analysis of the studies evaluating FMD in convalescent COVID-19 patients and controls with no history of COVID-19. METHODS A systematic literature search was conducted in the main scientific databases according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Using the random effects method, differences between cases and controls were expressed as mean difference (MD) with 95% confidence intervals (95% CI). The protocol was registered on PROSPERO with reference number CRD42021289684. RESULTS Twelve studies were included in the final analysis. A total of 644 convalescent COVID-19 patients showed significantly lower FMD values as compared to 662 controls (MD: -2.31%; 95% CI: -3.19, -1.44; p < 0.0001). Similar results were obtained in the sensitivity analysis of the studies that involved participants in either group with no cardiovascular risk factors or history of coronary artery disease (MD: -1.73%; 95% CI: -3.04, -0.41; p = 0.010). Interestingly, when considering studies separately based on enrolment within or after 3 months of symptom onset, results were further confirmed in both short- (MD: -2.20%; 95% CI: -3.35, -1.05; p < 0.0001) and long-term follow-up (MD: -2.53%; 95% CI: -4.19, -0.86; p = 0.003). Meta-regression models showed that an increasing prevalence of post-acute sequelae of COVID-19 was linked to a higher difference in FMD between cases and controls (Z-score: -2.09; p = 0.037). CONCLUSIONS Impaired endothelial function can be documented in convalescent COVID-19 patients, especially when residual clinical manifestations persist. Targeting endothelial dysfunction through pharmacological and rehabilitation strategies may represent an attractive therapeutic option.Key messagesThe mechanisms underlying the post-acute sequelae of coronavirus disease 2019 (COVID-19) have not been fully elucidated.Impaired endothelial function can be documented in convalescent COVID-19 patients for up to 1 year after infection, especially when residual clinical manifestations persist.Targeting endothelial dysfunction may represent an attractive therapeutic option in the post-acute phase of COVID-19.
Collapse
Affiliation(s)
- Pasquale Ambrosino
- Cardiac Rehabilitation Unit of Telese Terme Institute, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | | | - Marco Mosella
- Neurological Rehabilitation Unit of Telese Terme Institute, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Roberto Formisano
- Cardiac Rehabilitation Unit of Telese Terme Institute, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Antonio Molino
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | | | - Antimo Papa
- Cardiac Rehabilitation Unit of Telese Terme Institute, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council, Pozzuoli, Italy
| | | | - Mauro Maniscalco
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.,Pulmonary Rehabilitation Unit of Telese Terme Institute, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| |
Collapse
|
29
|
Izzo R, Trimarco V, Mone P, Aloè T, Capra Marzani M, Diana A, Fazio G, Mallardo M, Maniscalco M, Marazzi G, Messina N, Mininni S, Mussi C, Pelaia G, Pennisi A, Santus P, Scarpelli F, Tursi F, Zanforlin A, Santulli G, Trimarco B. Combining L-Arginine with vitamin C improves long-COVID symptoms: The LINCOLN Survey. Pharmacol Res 2022; 183:106360. [PMID: 35868478 PMCID: PMC9295384 DOI: 10.1016/j.phrs.2022.106360] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Recent evidence suggests that oxidative stress and endothelial dysfunction play critical roles in the pathophysiology of COVID-19 and Long-COVID. We hypothesized that a supplementation combining L-Arginine (to improve endothelial function) and Vitamin C (to reduce oxidation) could have favorable effects on Long-COVID symptoms. METHODS We designed a survey (LINCOLN: L-Arginine and Vitamin C improves Long-COVID), assessing several symptoms that have been associated with Long-COVID to be administered nationwide to COVID-19 survivors; the survey also included effort perception, measured using the Borg scale. Patients receiving the survey were divided in two groups, with a 2:1 ratio: the first group included patients that received L-Arginine + Vitamin C, whereas the second group received a multivitamin combination (alternative treatment). RESULTS 1390 patients successfully completed the survey. Following a 30-day treatment in both groups, the survey revealed that patients in the L-Arginine + Vitamin C treatment arm had significantly lower scores compared to patients who had received the multivitamin combination. There were no other significant differences between the two groups. When examining effort perception, we observed a significantly lower value (p < 0.0001) in patients receiving L-Arginine + Vitamin C compared to the alternative-treatment arm. CONCLUSIONS Our survey indicates that the supplementation with L-Arginine + Vitamin C has beneficial effects in Long-COVID, in terms of attenuating its typical symptoms and improving effort perception.
Collapse
Affiliation(s)
- Raffaele Izzo
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Valentina Trimarco
- Department of Neuroscience, Reproductive Sciences and Dentistry, Federico II University, Naples, Italy
| | - Pasquale Mone
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | | | | | | | | | | | | | | | | | - Simone Mininni
- Associazione Scientifica Interdisciplinare Aggiornamento Medico (ASIAM), Florence, Italy
| | - Chiara Mussi
- Department of Biomedical and Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, Modena, Italy
| | - Girolamo Pelaia
- Department of Health Science, Magna Graecia University, Catanzaro, Italy
| | | | | | | | | | | | - Gaetano Santulli
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy; Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA.
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
30
|
Gogu AE, Motoc AG, Docu Axelerad A, Stroe AZ, Gogu AA, Jianu DC. Tolosa-Hunt Syndrome and Hemorrhagic Encephalitis Presenting in a Patient after COVID-19 Vaccination Followed by COVID-19 Infection. Brain Sci 2022; 12:brainsci12070902. [PMID: 35884709 PMCID: PMC9313130 DOI: 10.3390/brainsci12070902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 02/05/2023] Open
Abstract
The presence of neurological symptoms within the clinical range of COVID-19 disease infection has increased. This paper presents the situation of a 45-year-old man having the medical antecedent diabetes mellitus, who presented to the emergency department with fever, headache, and respiratory symptoms, nine days following vaccination with the Ad26.COV2-S COVID-19 vaccine. The patient tested positive for SARS-CoV-2 based on nasal polymerase chain reaction (RT-PCR). Two weeks after the presentation, he developed Tolosa–Hunt Syndrome, an autoimmune phenomenon, with painful left ophthalmoplegia. Significant improvement was seen in terms of his discomfort; however, ptosis and ocular mobility improved only moderately after treatment with intravenous methylprednisolone, and the patient was discharged on a new insulin regimen. The patient returned after four weeks and the neurological exam results showed significant signs of right hemiparesis, mixed aphasia, incomplete left ophthalmoplegia, severe headache, and agitation; after a few days, the patient experienced a depressed level of consciousness and coma. The patient’s clinical condition worsened and, unfortunately, he died. MRI brain images revealed multiple ischemic strokes, meningitis, infectious vasculitis, and hemorrhagic encephalitis, which are all serious complications of COVID-19.
Collapse
Affiliation(s)
- Anca Elena Gogu
- Department of Neurology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (A.E.G.); (D.C.J.)
- Centre for Cognitive Research in Neuropsychiatric Pathology (Neuropsy-Cog), Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Andrei Gheorghe Motoc
- Department of Anatomy and Embryology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Any Docu Axelerad
- Department of Neurology, General Medicine Faculty, “Ovidius” University, 900470 Constanta, Romania;
| | - Alina Zorina Stroe
- Department of Neurology, General Medicine Faculty, “Ovidius” University, 900470 Constanta, Romania;
- Correspondence: ; Tel.: +40-727-987-950
| | - Andreea Alexandra Gogu
- “Victor Babes”, Medicine Faculty, University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Dragos Catalin Jianu
- Department of Neurology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (A.E.G.); (D.C.J.)
- Centre for Cognitive Research in Neuropsychiatric Pathology (Neuropsy-Cog), Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| |
Collapse
|
31
|
Yang S, Tong Y, Chen L, Yu W. Human Identical Sequences, hyaluronan, and hymecromone ─ the new mechanism and management of COVID-19. MOLECULAR BIOMEDICINE 2022; 3:15. [PMID: 35593963 PMCID: PMC9120813 DOI: 10.1186/s43556-022-00077-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/04/2022] [Indexed: 02/08/2023] Open
Abstract
COVID-19 caused by SARS-CoV-2 has created formidable damage to public health and market economy. Currently, SARS-CoV-2 variants has exacerbated the transmission from person-to-person. Even after a great deal of investigation on COVID-19, SARS-CoV-2 is still rampaging globally, emphasizing the urgent need to reformulate effective prevention and treatment strategies. Here, we review the latest research progress of COVID-19 and provide distinct perspectives on the mechanism and management of COVID-19. Specially, we highlight the significance of Human Identical Sequences (HIS), hyaluronan, and hymecromone ("Three-H") for the understanding and intervention of COVID-19. Firstly, HIS activate inflammation-related genes to influence COVID-19 progress through NamiRNA-Enhancer network. Accumulation of hyaluronan induced by HIS-mediated HAS2 upregulation is a substantial basis for clinical manifestations of COVID-19, especially in lymphocytopenia and pulmonary ground-glass opacity. Secondly, detection of plasma hyaluronan can be effective for evaluating the progression and severity of COVID-19. Thirdly, spike glycoprotein of SARS-CoV-2 may bind to hyaluronan and further serve as an allergen to stimulate allergic reaction, causing sudden adverse effects after vaccination or the aggravation of COVID-19. Finally, antisense oligonucleotides of HIS or inhibitors of hyaluronan synthesis (hymecromone) or antiallergic agents could be promising therapeutic agents for COVID-19. Collectively, Three-H could hold the key to understand the pathogenic mechanism and create effective therapeutic strategies for COVID-19.
Collapse
Affiliation(s)
- Shuai Yang
- Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Shanghai Public Health Clinical Center & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Key Laboratory of Medical Epigenetics, Shanghai, 200032, People's Republic of China
| | - Ying Tong
- Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Shanghai Public Health Clinical Center & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Key Laboratory of Medical Epigenetics, Shanghai, 200032, People's Republic of China
| | - Lu Chen
- Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Shanghai Public Health Clinical Center & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Key Laboratory of Medical Epigenetics, Shanghai, 200032, People's Republic of China
| | - Wenqiang Yu
- Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Shanghai Public Health Clinical Center & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Key Laboratory of Medical Epigenetics, Shanghai, 200032, People's Republic of China.
| |
Collapse
|