1
|
Leitzke M, Roach DT, Hesse S, Schönknecht P, Becker GA, Rullmann M, Sattler B, Sabri O. Long COVID - a critical disruption of cholinergic neurotransmission? Bioelectron Med 2025; 11:5. [PMID: 40011942 DOI: 10.1186/s42234-025-00167-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 01/30/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Following the COVID-19 pandemic, there are many chronically ill Long COVID (LC) patients with different symptoms of varying degrees of severity. The pathological pathways of LC remain unclear until recently and make identification of path mechanisms and exploration of therapeutic options an urgent challenge. There is an apparent relationship between LC symptoms and impaired cholinergic neurotransmission. METHODS This paper reviews the current literature on the effects of blocked nicotinic acetylcholine receptors (nAChRs) on the main affected organ and cell systems and contrasts this with the unblocking effects of the alkaloid nicotine. In addition, mechanisms are presented that could explain the previously unexplained phenomenon of post-vaccination syndrome (PVS). The fact that not only SARS-CoV-2 but numerous other viruses can bind to nAChRs is discussed under the assumption that numerous other post-viral diseases and autoimmune diseases (ADs) may also be due to impaired cholinergic transmission. We also present a case report that demonstrates changes in cholinergic transmission, specifically, the availability of α4β2 nAChRs by using (-)-[18F]Flubatine whole-body positron emission tomography (PET) imaging of cholinergic dysfunction in a LC patient along with a significant neurological improvement before and after low-dose transcutaneous nicotine (LDTN) administration. Lastly, a descriptive analysis and evaluation were conducted on the results of a survey involving 231 users of LDTN. RESULTS A substantial body of research has emerged that offers a compelling explanation for the phenomenon of LC, suggesting that it can be plausibly explained because of impaired nAChR function in the human body. Following a ten-day course of transcutaneous nicotine administration, no enduring neuropathological manifestations were observed in the patient. This observation was accompanied by a significant increase in the number of free ligand binding sites (LBS) of nAChRs, as determined by (-)-[18F]Flubatine PET imaging. The analysis of the survey shows that the majority of patients (73.5%) report a significant improvement in the symptoms of their LC/MEF/CFS disease as a result of LDTN. CONCLUSIONS In conclusion, based on current knowledge, LDTN appears to be a promising and safe procedure to relieve LC symptoms with no expected long-term harm.
Collapse
Affiliation(s)
- Marco Leitzke
- Department of Nuclear Medicine, University of Leipzig Medical Centre, Leipzig, 04103, Germany.
- Department of Anesthesiology, Intensive Care Medicine, Pain- and Palliative Therapy Helios Clinics, Colditzer Straße 48, Leisnig, 04703, Germany.
| | - Donald Troy Roach
- School of Comillas University, Renegade Research, Madrid, 28015, Spain
| | - Swen Hesse
- Department of Nuclear Medicine, University of Leipzig Medical Centre, Leipzig, 04103, Germany
| | - Peter Schönknecht
- Department of Psychiatry and Neurology Altscherbitz, Schkeuditz, 04435, Germany
- Outpatient Department for Forensic-Psychiatric Research, University of Leipzig, Leipzig, 04103, Germany
| | - Georg-Alexander Becker
- Department of Nuclear Medicine, University of Leipzig Medical Centre, Leipzig, 04103, Germany
| | - Michael Rullmann
- Department of Nuclear Medicine, University of Leipzig Medical Centre, Leipzig, 04103, Germany
| | - Bernhardt Sattler
- Department of Nuclear Medicine, University of Leipzig Medical Centre, Leipzig, 04103, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig Medical Centre, Leipzig, 04103, Germany
| |
Collapse
|
2
|
Fekete M, Lehoczki A, Szappanos Á, Toth A, Mahdi M, Sótonyi P, Benyó Z, Yabluchanskiy A, Tarantini S, Ungvari Z. Cerebromicrovascular mechanisms contributing to long COVID: implications for neurocognitive health. GeroScience 2025; 47:745-779. [PMID: 39777702 PMCID: PMC11872997 DOI: 10.1007/s11357-024-01487-4] [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: 11/25/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Long COVID (also known as post-acute sequelae of SARS-CoV-2 infection [PASC] or post-COVID syndrome) is characterized by persistent symptoms that extend beyond the acute phase of SARS-CoV-2 infection, affecting approximately 10% to over 30% of those infected. It presents a significant clinical challenge, notably due to pronounced neurocognitive symptoms such as brain fog. The mechanisms underlying these effects are multifactorial, with mounting evidence pointing to a central role of cerebromicrovascular dysfunction. This review investigates key pathophysiological mechanisms contributing to cerebrovascular dysfunction in long COVID and their impacts on brain health. We discuss how endothelial tropism of SARS-CoV-2 and direct vascular infection trigger endothelial dysfunction, impaired neurovascular coupling, and blood-brain barrier disruption, resulting in compromised cerebral perfusion. Furthermore, the infection appears to induce mitochondrial dysfunction, enhancing oxidative stress and inflammation within cerebral endothelial cells. Autoantibody formation following infection also potentially exacerbates neurovascular injury, contributing to chronic vascular inflammation and ongoing blood-brain barrier compromise. These factors collectively contribute to the emergence of white matter hyperintensities, promote amyloid pathology, and may accelerate neurodegenerative processes, including Alzheimer's disease. This review also emphasizes the critical role of advanced imaging techniques in assessing cerebromicrovascular health and the need for targeted interventions to address these cerebrovascular complications. A deeper understanding of the cerebrovascular mechanisms of long COVID is essential to advance targeted treatments and mitigate its long-term neurocognitive consequences.
Collapse
Affiliation(s)
- Monika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Andrea Lehoczki
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary.
| | - Ágnes Szappanos
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Rheumatology and Clinical Immunology, Semmelweis University, Budapest, Hungary
| | - Attila Toth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, 4032, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, 4032, Hungary
| | - Mohamed Mahdi
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, University of Debrecen, 4032, Debrecen, Hungary
- Infectology Clinic, University of Debrecen Clinical Centre, 4031, Debrecen, Hungary
| | - Péter Sótonyi
- Department of Vascular and Endovascular Surgery, Heart and Vascular Centre, Semmelweis University, 1122, Budapest, Hungary
| | - Zoltán Benyó
- Institute of Translational Medicine, Semmelweis University, 1094, Budapest, Hungary
- Cerebrovascular and Neurocognitive Disorders Research Group, HUN-REN , Semmelweis University, 1094, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
3
|
Narayanan SN, Padiyath S, Chandrababu K, Raj L, P S BC, Ninan GA, Sivadasan A, Jacobs AR, Li YW, Bhaskar A. Neurological, psychological, psychosocial complications of long-COVID and their management. Neurol Sci 2025; 46:1-23. [PMID: 39516425 PMCID: PMC11698801 DOI: 10.1007/s10072-024-07854-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Since it first appeared, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has had a significant and lasting negative impact on the health and economies of millions of individuals all over the globe. At the level of individual health too, many patients are not recovering fully and experiencing a long-term condition now commonly termed 'long-COVID'. Long-COVID is a collection of symptoms which must last more than 12 weeks following initial COVID infection, and which cannot be adequately explained by alternate diagnoses. The neurological and psychosocial impact of long-COVID is itself now a global health crisis and therefore preventing, diagnosing, and managing these patients is of paramount importance. This review focuses primarily on: neurological functioning deficits; mental health impacts; long-term mood problems; and associated psychosocial issues, among patients suffering from long-COVID with an eye towards the neurological basis of these symptoms. A concise account of the clinical relevance of the neurological and psychosocial impacts of long-COVID, the effects on long-term morbidity, and varied approaches in managing patients with significant chronic neurological symptoms and conditions was extracted from the literature, analysed and reported. A comprehensive account of plausible pathophysiological mechanisms involved in the development of long-COVID, its management, and future research needs have been discussed.
Collapse
Affiliation(s)
- Sareesh Naduvil Narayanan
- Department of Physiology, School of Medicine and Dentistry, AUC-UK Track, University of Central Lancashire, Preston, UK.
| | - Sreeshma Padiyath
- Department of Microbiology, School of Medicine and Dentistry, AUC-UK Track, University of Central Lancashire, Preston, UK
| | - Krishnapriya Chandrababu
- Centre for Neuroscience, Department of Biotechnology, Cochin University of Science and Technology (CUSAT), Kochi, India
| | - Lima Raj
- Department of Psychology, Sree Sankaracharya University of Sanskrit, Kalady, India
| | - Baby Chakrapani P S
- Centre for Neuroscience, Department of Biotechnology, Cochin University of Science and Technology (CUSAT), Kochi, India
- Centre for Excellence in Neurodegeneration and Brain Health (CENABH), Cochin University of Science and Technology (CUSAT), Kochi, India
| | | | - Ajith Sivadasan
- Department of Neurology, Christian Medical College (CMC), Vellore, India
| | - Alexander Ryan Jacobs
- School of Medicine and Dentistry, AUC-UK Track, University of Central Lancashire, Preston, UK
| | - Yan Wa Li
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Anand Bhaskar
- Department of Physiology, Christian Medical College (CMC), Vellore, India
| |
Collapse
|
4
|
Russell SJ, Parker K, Lehoczki A, Lieberman D, Partha IS, Scott SJ, Phillips LR, Fain MJ, Nikolich JŽ. Post-acute sequelae of SARS-CoV-2 infection (Long COVID) in older adults. GeroScience 2024; 46:6563-6581. [PMID: 38874693 PMCID: PMC11493926 DOI: 10.1007/s11357-024-01227-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024] Open
Abstract
Long COVID, also known as PASC (post-acute sequelae of SARS-CoV-2), is a complex infection-associated chronic condition affecting tens of millions of people worldwide. Many aspects of this condition are incompletely understood. Among them is how this condition may manifest itself in older adults and how it might impact the older population. Here, we briefly review the current understanding of PASC in the adult population and examine what is known on its features with aging. Finally, we outline the major gaps and areas for research most germane to older adults.
Collapse
Affiliation(s)
- Samantha J Russell
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Karen Parker
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Andrea Lehoczki
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
- Department of Haematology and Stem Cell Transplantation, National Institute for Haematology and Infectious Diseases, South Pest Central Hospital, 1097, Budapest, Hungary
- Department of Public Health, Semmelweis University, Budapest, Hungary
| | - David Lieberman
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Indu S Partha
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Serena J Scott
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Banner University Medicine-Tucson, Tucson, AZ, USA
| | - Linda R Phillips
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
- College of Nursing, University of Arizona, Tucson, AZ, USA
| | - Mindy J Fain
- Division of General Internal Medicine, Geriatrics, and Palliative Medicine, Department of Medicine, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- Banner University Medicine-Tucson, Tucson, AZ, USA.
- College of Nursing, University of Arizona, Tucson, AZ, USA.
| | - Janko Ž Nikolich
- Arizona Center of Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA.
- The Aegis Consortium for Pandemic-Free Future, University of Arizona Health Sciences, Tucson, AZ, USA.
| |
Collapse
|
5
|
Tilikete C, Zamali I, Meddeb Z, Kharroubi G, Marzouki S, Dhaouadi T, Ben Hmid A, Samoud S, Galai Y, Charfeddine S, Abid L, Abdessalem S, Bettaieb J, Hamzaoui S, Bouslama K, Ben Ahmed M. Exploring the landscape of symptom-specific inflammatory cytokines in post-COVID syndrome patients. BMC Infect Dis 2024; 24:1337. [PMID: 39578766 PMCID: PMC11583569 DOI: 10.1186/s12879-024-10222-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024] Open
Abstract
INTRODUCTION Post-COVID syndrome (PCS) is characterized by a polymorphism of symptoms with hypothetical pathophysiological mechanisms. Here, we aimed to analyze the profile of inflammatory cytokines in patients with PCS and to study the relationship between this profile, the clinical symptoms as well as the endothelial function in PCS. METHODS Our analytical study involved all eligible patients (n = 66) with PCS included from April 2021 to December 2021. The serum concentration of cytokines IFN-γ, IL-1α, IL-1β, IL-6, IL-8, IL-10, IL-12p70, IL-27, IP-10, MCP-1 and TNF-α was quantified by flow cytometry. Endothelial function was explored by assessing microvascular flow and reactivity using thermal probes. A comparative study was carried out according to the presence of each PCS symptom. RESULTS The average age of our patients was 55.9 ± 16.2 years. The sex ratio was 0.69. Forty-one patients (62%) presented with a severe form of acute infection. The most frequently reported symptoms were dyspnea (67%), fatigue (50%), and memory problems (32%). Fifty-seven patients (86%) had endothelial dysfunction. The majority of patients had increased levels of IP-10 (100%), IL-8 (95%), IFN-γ (95%), MCP-1 (80%), and TNF-α (70%). The serum concentration of IL-10 was below the threshold of quantification in 89% of subjects. The severe form of acute infection was associated with elevated IL-10, MCP-1, and IL-27. Increased IL-6 and IL-27 levels were associated with fatigue while IL-8 concentrations were higher in patients who reported dyspnea. Elevation of IL-8 level was more common in patients with profound impairment of endothelial function. CONCLUSION Our results further support the presence of endothelial dysfunction in PCS and show an elevation of pro-inflammatory cytokines with a downmodulation of the IL-10- anti-inflammatory response. In addition, immuno-clinical phenotypes emerge, such as an inflammatory profile mediated by IL-6 and IL-27 in fatigue and IL-8 in dyspnea. The identification of immuno-clinical phenotypes would allow a better understanding of the pathophysiology of PCS symptoms.
Collapse
Affiliation(s)
- Chafik Tilikete
- Faculté de Médecine de Tunis, University Tunis El Manar, Tunis, Tunisia
| | - Imen Zamali
- Faculté de Médecine de Tunis, University Tunis El Manar, Tunis, Tunisia
- Department of Clinical Immunology, Institut Pasteur de Tunis, Tunis, Tunisia
- Laboratory of Transmission, Control and immunobiology of Infections (LR16IPT02), Institut Pasteur de Tunis, Tunis, Tunisia
| | - Zeineb Meddeb
- Faculté de Médecine de Tunis, University Tunis El Manar, Tunis, Tunisia
- Department of Internal Medicine, Mongi Slim La Marsa Hospital, Tunis, Tunisia
| | - Ghassen Kharroubi
- Faculté de Médecine de Tunis, University Tunis El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and immunobiology of Infections (LR16IPT02), Institut Pasteur de Tunis, Tunis, Tunisia
- Department of Medical Epidemiology, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Soumaya Marzouki
- Department of Clinical Immunology, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Tarak Dhaouadi
- Faculté de Médecine de Tunis, University Tunis El Manar, Tunis, Tunisia
- Department of Immunology, Charles Nicolle Hospital, Tunis, Tunisia
| | - Ahlem Ben Hmid
- Faculté de Médecine de Tunis, University Tunis El Manar, Tunis, Tunisia
- Department of Clinical Immunology, Institut Pasteur de Tunis, Tunis, Tunisia
- Laboratory of Transmission, Control and immunobiology of Infections (LR16IPT02), Institut Pasteur de Tunis, Tunis, Tunisia
| | - Samar Samoud
- Department of Clinical Immunology, Institut Pasteur de Tunis, Tunis, Tunisia
- Laboratory of Transmission, Control and immunobiology of Infections (LR16IPT02), Institut Pasteur de Tunis, Tunis, Tunisia
| | - Yousr Galai
- Department of Clinical Immunology, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Selma Charfeddine
- Department of Cardiology, Hedi Chaker University Hospital Sfax, Sfax, Tunisia
- Faculté de Médecine de Sfax, University of Sfax, Sfax, Tunisia
| | - Leila Abid
- Department of Cardiology, Hedi Chaker University Hospital Sfax, Sfax, Tunisia
- Faculté de Médecine de Sfax, University of Sfax, Sfax, Tunisia
| | | | - Jihène Bettaieb
- Department of Clinical Immunology, Institut Pasteur de Tunis, Tunis, Tunisia
- Laboratory of Transmission, Control and immunobiology of Infections (LR16IPT02), Institut Pasteur de Tunis, Tunis, Tunisia
- Department of Medical Epidemiology, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Saloua Hamzaoui
- Faculté de Médecine de Tunis, University Tunis El Manar, Tunis, Tunisia
- Department of Internal Medicine, Mongi Slim La Marsa Hospital, Tunis, Tunisia
| | - Kamel Bouslama
- Faculté de Médecine de Tunis, University Tunis El Manar, Tunis, Tunisia
- Department of Internal Medicine, Mongi Slim La Marsa Hospital, Tunis, Tunisia
| | - Mélika Ben Ahmed
- Faculté de Médecine de Tunis, University Tunis El Manar, Tunis, Tunisia.
- Department of Clinical Immunology, Institut Pasteur de Tunis, Tunis, Tunisia.
- Laboratory of Transmission, Control and immunobiology of Infections (LR16IPT02), Institut Pasteur de Tunis, Tunis, Tunisia.
| |
Collapse
|
6
|
Moulias A, Koros R, Papageorgiou A, Katechis S, Patrinos P, Trigka-Vasilakopoulou A, Papageorgiou A, Papaioannou O, Akinosoglou K, Leventopoulos G, Tsigkas G, Tzouvelekis A, Davlouros P. Assessment of Endothelial Function in Patients with COVID-19 Using Peripheral Arterial Tonometry. Life (Basel) 2024; 14:1512. [PMID: 39598310 PMCID: PMC11595729 DOI: 10.3390/life14111512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
There is increasing evidence that COVID-19 induces endothelial dysfunction that may precede thrombotic and cardiovascular complications. The aim of this study is to evaluate endothelial function using peripheral arterial tonometry (EndoPAT). The primary endpoint is the hyperemic vascular response index (LnRHI) at two months post-discharge. Secondary endpoints include the LnRHI during hospitalization and at six-month follow-up, the proportion of patients with endothelial dysfunction (LnRHI ≤ 0.51), and the incidence of thrombotic events, cardiovascular complications, and mortality during the follow-up period. The study included 23 COVID-19 patients and 22 COVID-19-negative, matched controls. The patients exhibited a significant reduction in the LnRHI at two months post-discharge compared to the controls (median = 0.55 [IQR: 0.49-0.68] vs. median = 0.70 [IQR: 0.62-0.83]; p = 0.012). The difference in the LnRHI between patients and controls was evident from hospitalization and persisted at two and six months without significant temporal changes. The proportion of COVID-19 patients with endothelial dysfunction (LnRHI ≤ 0.51) was 61% during hospitalization and 55% at six months. There was no significant difference in thrombotic or cardiovascular events, nor in mortality. This study demonstrates that COVID-19 adversely affects endothelial function, as evidenced by a reduction in the hyperemic vascular response index, and endothelial dysfunction may also persist.
Collapse
Affiliation(s)
- Athanasios Moulias
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Rafail Koros
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Angeliki Papageorgiou
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
- Department of Inherited and Rare Cardiovascular Diseases, Onassis Cardiac Surgery Center, 17674 Athens, Greece
| | - Spyridon Katechis
- Department of Rheumatology, General Hospital of Asklipieio Voulas, 16673 Athens, Greece;
| | - Panagiotis Patrinos
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Aikaterini Trigka-Vasilakopoulou
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Athanasios Papageorgiou
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Ourania Papaioannou
- Department of Pneumonology, General University Hospital of Patras, 26504 Patras, Greece; (O.P.); (A.T.)
| | - Karolina Akinosoglou
- Department of Internal Medicine, General University Hospital of Patras, 26504 Patras, Greece;
| | - Georgios Leventopoulos
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Grigorios Tsigkas
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| | - Argyrios Tzouvelekis
- Department of Pneumonology, General University Hospital of Patras, 26504 Patras, Greece; (O.P.); (A.T.)
| | - Periklis Davlouros
- Department of Cardiology, General University Hospital of Patras, 26504 Patras, Greece; (A.M.); (R.K.); (P.P.); (A.T.-V.); (A.P.); (G.L.); (G.T.); (P.D.)
| |
Collapse
|
7
|
Li B, Bai J, Xiong Y, Guo D, Fu B, Deng G, Wu H. Understanding the mechanisms and treatments of long COVID to address future public health risks. Life Sci 2024; 353:122938. [PMID: 39084516 DOI: 10.1016/j.lfs.2024.122938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/20/2024] [Accepted: 07/27/2024] [Indexed: 08/02/2024]
Abstract
The 2019 coronavirus disease (COVID-19), triggered by the severe acute respiratory syndrome coronavirus (SARS-CoV-2), has seen numerous individuals undergo and recover from it, drawing extensive attention to their health conditions. Extensive studies indicate that even after surpassing the acute phase of infection, patients continue to experience persistent symptoms such as fatigue, pain, depression, weakening, and anosmia. COVID-19 appears not to have concluded but rather to persist long-term in certain individuals, termed as "long COVID." This represents a heterogeneous ailment involving multiple organ systems, with a perceived complex and still elusive pathogenesis. Among patients with long COVID, observations reveal immune dysregulation, coagulation impairments, and microbial dysbiosis, considered potential mechanisms explaining sustained adverse outcomes post COVID-19. Based on the multifactorial nature, varied symptoms, and heterogeneity of long COVID, we have summarized several categories of current therapeutic approaches. Furthermore, the symptoms of long COVID resemble those of other viral illnesses, suggesting that existing knowledge may offer novel insights into long-term COVID implications. Here, we provide an overview of existing literature associated with long COVID and summarize potential mechanisms, treatment modalities, and other analogous conditions. Lastly, we underscore the inadequacies in long COVID treatment approaches and emphasize the significance of conducting further research and clinical trials.
Collapse
Affiliation(s)
- Bohao Li
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Junlu Bai
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Yan Xiong
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Dong Guo
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Beibei Fu
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Guohong Deng
- Department of Infectious Diseases, First Affiliated Hospital, Army Medical University, Chongqing, China.
| | - Haibo Wu
- School of Life Sciences, Chongqing University, Chongqing 401331, China.
| |
Collapse
|
8
|
Aljadah M, Khan N, Beyer AM, Chen Y, Blanker A, Widlansky ME. Clinical Implications of COVID-19-Related Endothelial Dysfunction. JACC. ADVANCES 2024; 3:101070. [PMID: 39055276 PMCID: PMC11269277 DOI: 10.1016/j.jacadv.2024.101070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/10/2024] [Accepted: 05/24/2024] [Indexed: 07/27/2024]
Abstract
Endothelial dysfunction represents a measurable and early manifestation of vascular disease. Emerging evidence suggests cardiovascular risk remains elevated after COVID-19 infection for at least 12 months, regardless of cardiovascular disease status prior to infection. We review the relationship between the severity of endothelial dysfunction and the severity of acute COVID-19 illness, the degree of impairment following recovery in both those with and without postacute sequalae SARS-CoV-2 infection, and current therapeutic efforts targeting endothelial function in patients following COVID-19 infection. We identify gaps in the literature to highlight specific areas where clinical research efforts hold promise for progress in understanding the connections between endothelial function, COVID-19, and clinical outcomes that will lead to beneficial therapeutics.
Collapse
Affiliation(s)
- Michael Aljadah
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Nabeel Khan
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Andreas M. Beyer
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Yiliang Chen
- Division of Endocrinology and Molecular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Versiti Blood Center of Wisconsin, Milwaukee, Wisconsin, USA
| | - Andrew Blanker
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Michael E. Widlansky
- Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
9
|
Chidambaram V, Kumar A, Sadaf MI, Lu E, Al’Aref SJ, Tarun T, Galiatsatos P, Gulati M, Blumenthal RS, Leucker TM, Karakousis PC, Mehta JL. COVID-19 in the Initiation and Progression of Atherosclerosis: Pathophysiology During and Beyond the Acute Phase. JACC. ADVANCES 2024; 3:101107. [PMID: 39113913 PMCID: PMC11304887 DOI: 10.1016/j.jacadv.2024.101107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 04/24/2024] [Accepted: 06/01/2024] [Indexed: 08/10/2024]
Abstract
The incidence of atherosclerotic cardiovascular disease is increasing globally, especially in low- and middle-income countries, despite significant efforts to reduce traditional risk factors. Premature subclinical atherosclerosis has been documented in association with several viral infections. The magnitude of the recent COVID-19 pandemic has highlighted the need to understand the association between SARS-CoV-2 and atherosclerosis. This review examines various pathophysiological mechanisms, including endothelial dysfunction, platelet activation, and inflammatory and immune hyperactivation triggered by SARS-CoV-2 infection, with specific attention on their roles in initiating and promoting the progression of atherosclerotic lesions. Additionally, it addresses the various pathogenic mechanisms by which COVID-19 in the post-acute phase may contribute to the development of vascular disease. Understanding the overlap of these syndromes may enable novel therapeutic strategies. We further explore the need for guidelines for closer follow-up for the often-overlooked evidence of atherosclerotic cardiovascular disease among patients with recent COVID-19, particularly those with cardiometabolic risk factors.
Collapse
Affiliation(s)
- Vignesh Chidambaram
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Amudha Kumar
- Division of Cardiology, Department of Medicine, Loyola University Medical Center, Maywood, Illinois, USA
| | - Murrium I. Sadaf
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Emily Lu
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Subhi J. Al’Aref
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Tushar Tarun
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Panagis Galiatsatos
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Martha Gulati
- Barbra Streisand Women's Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Roger S. Blumenthal
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thorsten M. Leucker
- Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Petros C. Karakousis
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jawahar L. Mehta
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Division of Cardiovascular Medicine, Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| |
Collapse
|
10
|
Calcaterra V, Zanelli S, Foppiani A, Verduci E, Benatti B, Bollina R, Bombaci F, Brucato A, Cammarata S, Calabrò E, Cirnigliaro G, Della Torre S, Dell’osso B, Moltrasio C, Marzano AV, Nostro C, Romagnuolo M, Trotta L, Savasi V, Smiroldo V, Zuccotti G. Long COVID in Children, Adults, and Vulnerable Populations: A Comprehensive Overview for an Integrated Approach. Diseases 2024; 12:95. [PMID: 38785750 PMCID: PMC11120262 DOI: 10.3390/diseases12050095] [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: 04/02/2024] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Long COVID affects both children and adults, including subjects who experienced severe, mild, or even asymptomatic SARS-CoV-2 infection. We have provided a comprehensive overview of the incidence, clinical characteristics, risk factors, and outcomes of persistent COVID-19 symptoms in both children and adults, encompassing vulnerable populations, such as pregnant women and oncological patients. Our objective is to emphasize the critical significance of adopting an integrated approach for the early detection and appropriate management of long COVID. The incidence and severity of long COVID symptoms can have a significant impact on the quality of life of patients and the course of disease in the case of pre-existing pathologies. Particularly, in fragile and vulnerable patients, the presence of PASC is related to significantly worse survival, independent from pre-existing vulnerabilities and treatment. It is important try to achieve an early recognition and management. Various mechanisms are implicated, resulting in a wide range of clinical presentations. Understanding the specific mechanisms and risk factors involved in long COVID is crucial for tailoring effective interventions and support strategies. Management approaches involve comprehensive biopsychosocial assessments and treatment of symptoms and comorbidities, such as autonomic dysfunction, as well as multidisciplinary rehabilitation. The overall course of long COVID is one of gradual improvement, with recovery observed in the majority, though not all, of patients. As the research on long-COVID continues to evolve, ongoing studies are likely to shed more light on the intricate relationship between chronic diseases, such as oncological status, cardiovascular diseases, psychiatric disorders, and the persistent effects of SARS-CoV-2 infection. This information could guide healthcare providers, researchers, and policymakers in developing targeted interventions.
Collapse
Affiliation(s)
- Valeria Calcaterra
- Department of Internal Medicine and Therapeutics, Università degli Sudi di Pavia, 27100 Pavia, Italy;
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (S.Z.); (E.V.)
| | - Sara Zanelli
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (S.Z.); (E.V.)
| | - Andrea Foppiani
- International Center for the Assessment of Nutritional Status and the Development of Dietary Intervention Strategies (ICANS-DIS), Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, 20157 Milano, Italy;
- IRCCS Istituto Auxologico Italiano, Department of Endocrine and Metabolic Medicine, Clinical Nutrition Unit, 20145 Milano, Italy
| | - Elvira Verduci
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (S.Z.); (E.V.)
- Department of Health Sciences, Università degli Studi di Milano, 20157 Milano, Italy
| | - Beatrice Benatti
- “Aldo Ravelli” Center for Nanotechnology and Neurostimulation, Università degli Studi di Milano, 20157 Milano, Italy; (B.B.); (B.D.)
- Department of Psychiatry, ASST Fatebenefratelli-Sacco, University of Milano, 20154 Milano, Italy; (G.C.); (C.N.)
| | - Roberto Bollina
- Department of Medical Oncology, ASST Rhodense, 20024 Milano, Italy; (R.B.); (S.D.T.); (V.S.)
| | - Francesco Bombaci
- Department of Radiology, ASST Fatebenefratelli Sacco, 20154 Milano, Italy;
| | - Antonio Brucato
- Department of Internal Medicine, ASST Fatebenefratelli-Sacco, 20154 Milano, Italy; (A.B.); (E.C.); (L.T.)
| | - Selene Cammarata
- Department of Woman, Mother and Neonate, Luigi Sacco Hospital, ASST Fatebenefratelli-Sacco, 20154 Milano, Italy; (S.C.); (V.S.)
| | - Elisa Calabrò
- Department of Internal Medicine, ASST Fatebenefratelli-Sacco, 20154 Milano, Italy; (A.B.); (E.C.); (L.T.)
| | - Giovanna Cirnigliaro
- Department of Psychiatry, ASST Fatebenefratelli-Sacco, University of Milano, 20154 Milano, Italy; (G.C.); (C.N.)
| | - Silvia Della Torre
- Department of Medical Oncology, ASST Rhodense, 20024 Milano, Italy; (R.B.); (S.D.T.); (V.S.)
| | - Bernardo Dell’osso
- “Aldo Ravelli” Center for Nanotechnology and Neurostimulation, Università degli Studi di Milano, 20157 Milano, Italy; (B.B.); (B.D.)
- Department of Psychiatry, ASST Fatebenefratelli-Sacco, University of Milano, 20154 Milano, Italy; (G.C.); (C.N.)
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
- Centro per lo Studio dei Meccanismi Molecolari alla Base delle Patologie Neuro-Psico-Geriatriche, Università degli Studi di Milano, 20157 Milano, Italy
| | - Chiara Moltrasio
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (C.M.); (A.V.M.); (M.R.)
| | - Angelo Valerio Marzano
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (C.M.); (A.V.M.); (M.R.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milano, Italy
| | - Chiara Nostro
- Department of Psychiatry, ASST Fatebenefratelli-Sacco, University of Milano, 20154 Milano, Italy; (G.C.); (C.N.)
| | - Maurizio Romagnuolo
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy; (C.M.); (A.V.M.); (M.R.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milano, Italy
| | - Lucia Trotta
- Department of Internal Medicine, ASST Fatebenefratelli-Sacco, 20154 Milano, Italy; (A.B.); (E.C.); (L.T.)
| | - Valeria Savasi
- Department of Woman, Mother and Neonate, Luigi Sacco Hospital, ASST Fatebenefratelli-Sacco, 20154 Milano, Italy; (S.C.); (V.S.)
- Department of Biomedical and Clinical Science, Università degli Studi di Milano, 20157 Milano, Italy
| | - Valeria Smiroldo
- Department of Medical Oncology, ASST Rhodense, 20024 Milano, Italy; (R.B.); (S.D.T.); (V.S.)
| | - Gianvincenzo Zuccotti
- Pediatric Department, Buzzi Children’s Hospital, 20154 Milano, Italy; (S.Z.); (E.V.)
- Department of Biomedical and Clinical Science, Università degli Studi di Milano, 20157 Milano, Italy
| |
Collapse
|
11
|
Nunes JM, Kell DB, Pretorius E. Herpesvirus Infection of Endothelial Cells as a Systemic Pathological Axis in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Viruses 2024; 16:572. [PMID: 38675914 PMCID: PMC11053605 DOI: 10.3390/v16040572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Understanding the pathophysiology of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is critical for advancing treatment options. This review explores the novel hypothesis that a herpesvirus infection of endothelial cells (ECs) may underlie ME/CFS symptomatology. We review evidence linking herpesviruses to persistent EC infection and the implications for endothelial dysfunction, encompassing blood flow regulation, coagulation, and cognitive impairment-symptoms consistent with ME/CFS and Long COVID. This paper provides a synthesis of current research on herpesvirus latency and reactivation, detailing the impact on ECs and subsequent systemic complications, including latent modulation and long-term maladaptation. We suggest that the chronicity of ME/CFS symptoms and the multisystemic nature of the disease may be partly attributable to herpesvirus-induced endothelial maladaptation. Our conclusions underscore the necessity for further investigation into the prevalence and load of herpesvirus infection within the ECs of ME/CFS patients. This review offers conceptual advances by proposing an endothelial infection model as a systemic mechanism contributing to ME/CFS, steering future research toward potentially unexplored avenues in understanding and treating this complex syndrome.
Collapse
Affiliation(s)
- Jean M. Nunes
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa;
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa;
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Building 220, Chemitorvet 200, 2800 Kongens Lyngby, Denmark
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa;
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK
| |
Collapse
|
12
|
Kitselman A K, Bédard-Matteau J, Rousseau S, Tabrizchi R, Daneshtalab N. Sex differences in vascular endothelial function related to acute and long COVID-19. Vascul Pharmacol 2024; 154:107250. [PMID: 38043758 DOI: 10.1016/j.vph.2023.107250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus has been at the forefront of health sciences research since its emergence in China in 2019 that quickly led to a global pandemic. As a result of this research, and the large numbers of infected patients globally, there were rapid enhancements made in our understanding of Coronavirus disease 2019 (COVID-19) pathology, including its role in the development of uncontrolled immune responses and its link to the development of endotheliitis and endothelial dysfunction. There were also some noted differences in the rate and severity of infection between males and females with acute COVID. Some individuals infected with SARS-CoV-2 also experience long-COVID, an important hallmark symptom of this being Myalgic Encephalomyelitis-Chronic Fatigue Syndrome (ME-CFS), also experienced differently between males and females. The purpose of this review is to discuss the impact of sex on the vasculature during acute and long COVID-19, present any link between ME-CFS and endothelial dysfunction, and provide evidence for the relationship between ME-CFS and the immune system. We also will delineate biological sex differences observed in other post viral infections and, assess if sex differences exist in how the immune system responds to viral infection causing ME-CFS.
Collapse
Affiliation(s)
- Kayla Kitselman A
- Faculty of Medicine, Division of Biomedical Sciences at Memorial University of Newfoundland and Labrador, Canada
| | - Jérôme Bédard-Matteau
- Faculty of Medicine, Department of Pharmacology and Therapeutics, McGill University, Québec, Canada; Meakins-Christie Laboratories, RI-MUHC, Block EOffice EM3.2244Lab E03.21371001 Decarie Blvd., Montreal, QC H4A 3J1, Canada
| | - Simon Rousseau
- Faculty of Medicine, Department of Pharmacology and Therapeutics, McGill University, Québec, Canada; Faculty of Medicine, Department of Experimental Medicine, McGill University, Québec, Canada; Meakins-Christie Laboratories, RI-MUHC, Block EOffice EM3.2244Lab E03.21371001 Decarie Blvd., Montreal, QC H4A 3J1, Canada
| | - Reza Tabrizchi
- Faculty of Medicine, Division of Biomedical Sciences at Memorial University of Newfoundland and Labrador, Canada
| | - Noriko Daneshtalab
- School of Pharmacy at Memorial University of Newfoundland and Labrador, Canada.
| |
Collapse
|
13
|
Cervia-Hasler C, Brüningk SC, Hoch T, Fan B, Muzio G, Thompson RC, Ceglarek L, Meledin R, Westermann P, Emmenegger M, Taeschler P, Zurbuchen Y, Pons M, Menges D, Ballouz T, Cervia-Hasler S, Adamo S, Merad M, Charney AW, Puhan M, Brodin P, Nilsson J, Aguzzi A, Raeber ME, Messner CB, Beckmann ND, Borgwardt K, Boyman O. Persistent complement dysregulation with signs of thromboinflammation in active Long Covid. Science 2024; 383:eadg7942. [PMID: 38236961 DOI: 10.1126/science.adg7942] [Citation(s) in RCA: 141] [Impact Index Per Article: 141.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 11/24/2023] [Indexed: 01/23/2024]
Abstract
Long Covid is a debilitating condition of unknown etiology. We performed multimodal proteomics analyses of blood serum from COVID-19 patients followed up to 12 months after confirmed severe acute respiratory syndrome coronavirus 2 infection. Analysis of >6500 proteins in 268 longitudinal samples revealed dysregulated activation of the complement system, an innate immune protection and homeostasis mechanism, in individuals experiencing Long Covid. Thus, active Long Covid was characterized by terminal complement system dysregulation and ongoing activation of the alternative and classical complement pathways, the latter associated with increased antibody titers against several herpesviruses possibly stimulating this pathway. Moreover, markers of hemolysis, tissue injury, platelet activation, and monocyte-platelet aggregates were increased in Long Covid. Machine learning confirmed complement and thromboinflammatory proteins as top biomarkers, warranting diagnostic and therapeutic interrogation of these systems.
Collapse
Affiliation(s)
- Carlo Cervia-Hasler
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Sarah C Brüningk
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Tobias Hoch
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Bowen Fan
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Giulia Muzio
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Ryan C Thompson
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Laura Ceglarek
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Roman Meledin
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Patrick Westermann
- Precision Proteomics Center, Swiss Institute of Allergy and Asthma Research, University of Zurich, 7265 Davos, Switzerland
| | - Marc Emmenegger
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Patrick Taeschler
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Yves Zurbuchen
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Michele Pons
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Dominik Menges
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, 8001 Zurich, Switzerland
| | - Tala Ballouz
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, 8001 Zurich, Switzerland
| | - Sara Cervia-Hasler
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Sarah Adamo
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Miriam Merad
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexander W Charney
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Milo Puhan
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, 8001 Zurich, Switzerland
| | - Petter Brodin
- Unit for Clinical Pediatrics, Department of Women's and Children's Health, Karolinska Institute, 17165 Solna, Sweden
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK
| | - Jakob Nilsson
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Miro E Raeber
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Christoph B Messner
- Precision Proteomics Center, Swiss Institute of Allergy and Asthma Research, University of Zurich, 7265 Davos, Switzerland
| | - Noam D Beckmann
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Division of Data Driven and Digital Medicine (D3M), Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Karsten Borgwardt
- Department of Biosystems Science and Engineering, ETH Zurich, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
- Faculty of Medicine and Faculty of Science, University of Zurich, 8006 Zurich, Switzerland
| |
Collapse
|
14
|
Miller JB, Rebman AW, de Flores MDV, Wang H, Darrah E, Aucott JN. Annexin A2 antibodies in post-treatment Lyme disease. Ther Adv Infect Dis 2024; 11:20499361241242971. [PMID: 38559699 PMCID: PMC10981857 DOI: 10.1177/20499361241242971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Background Anti-annexin A2 (AA2) antibodies have been described in Lyme arthritis and erythema migrans, although they have not been described in post-treatment Lyme disease (PTLD). Objectives Determine whether anti-AA2 antibodies are present among patients with PTLD and determine the clinical relevance of these antibodies. Design and methods Anti-AA2 levels were tested serially in a longitudinal cohort of 44 patients with acute Lyme disease, 22 with a return to health (EM RTH), and 22 with PTLD. Anti-AA2 antibodies were also assessed in a cross-sectional group of 281 patients with PTLD. Results Anti-AA2 antibodies were highest after antimicrobial therapy in both the EM RTH and PTLD cohorts. By 6 months, there was no difference between EM RTH and healthy controls. Anti-AA2 antibodies were higher in the cross-sectional PTLD group (79.69 versus 48.22 units, p < 0.0001), though with no difference in total symptom burden. Conclusion Anti-AA2 persists in PTLD, though did not identify a clinical phenotype.
Collapse
Affiliation(s)
- John B. Miller
- Division of Rheumatology, Johns Hopkins University School of Medicine, 5200 Eastern Avenue, Mason F Lord Building Center Tower, Suite 4100, Baltimore, MD 21224, USA
| | - Alison W. Rebman
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Hong Wang
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Erika Darrah
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John N. Aucott
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
15
|
Mroueh A, Fakih W, Carmona A, Trimaille A, Matsushita K, Marchandot B, Qureshi AW, Gong DS, Auger C, Sattler L, Reydel A, Hess S, Oulehri W, Vollmer O, Lessinger JM, Meyer N, Pieper MP, Jesel L, Bäck M, Schini-Kerth V, Morel O. COVID-19 promotes endothelial dysfunction and thrombogenicity: role of proinflammatory cytokines/SGLT2 prooxidant pathway. J Thromb Haemost 2024; 22:286-299. [PMID: 37797691 DOI: 10.1016/j.jtha.2023.09.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 09/18/2023] [Accepted: 09/21/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND COVID-19 is associated with an increased risk of cardiovascular complications. Although cytokines have a predominant role in endothelium damage, the precise molecular mechanisms are far from being elucidated. OBJECTIVES The present study hypothesized that inflammation in patients with COVID-19 contributes to endothelial dysfunction through redox-sensitive SGLT2 overexpression and investigated the protective effect of SGLT2 inhibition by empagliflozin. METHODS Human plasma samples were collected from patients with acute, subacute, and long COVID-19 (n = 100), patients with non-COVID-19 and cardiovascular risk factors (n = 50), and healthy volunteers (n = 25). Porcine coronary artery endothelial cells (ECs) were incubated with plasma (10%). Protein expression levels were determined using Western blot analyses and immunofluorescence staining, mRNA expression by quantitative reverse transcription-polymerase chain reaction, and the level of oxidative stress by dihydroethidium staining. Platelet adhesion, aggregation, and thrombin generation were determined. RESULTS Increased plasma levels of interleukin (IL)-1β, IL-6, tumor necrosis factor-α, monocyte chemoattractant protein-1, and soluble intercellular adhesion molecule-1 were observed in patients with COVID-19. Exposure of ECs to COVID-19 plasma with high cytokines levels induced redox-sensitive upregulation of SGLT2 expression via proinflammatory cytokines IL-1β, IL-6, and tumor necrosis factor-α which, in turn, fueled endothelial dysfunction, senescence, NF-κB activation, inflammation, platelet adhesion and aggregation, von Willebrand factor secretion, and thrombin generation. The stimulatory effect of COVID-19 plasma was blunted by neutralizing antibodies against proinflammatory cytokines and empagliflozin. CONCLUSION In patients with COVID-19, proinflammatory cytokines induced a redox-sensitive upregulation of SGLT2 expression in ECs, which in turn promoted endothelial injury, senescence, platelet adhesion, aggregation, and thrombin generation. SGLT2 inhibition with empagliflozin appeared as an attractive strategy to restore vascular homeostasis in COVID-19.
Collapse
Affiliation(s)
- Ali Mroueh
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, FMTS, Strasbourg, France
| | - Walaa Fakih
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, FMTS, Strasbourg, France
| | - Adrien Carmona
- Division of Cardiovascular Medicine, Strasbourg University Hospital, Strasbourg, France
| | - Antonin Trimaille
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, FMTS, Strasbourg, France; Division of Cardiovascular Medicine, Strasbourg University Hospital, Strasbourg, France. https://twitter.com/A_Trimaille
| | - Kensuke Matsushita
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, FMTS, Strasbourg, France; Division of Cardiovascular Medicine, Strasbourg University Hospital, Strasbourg, France
| | - Benjamin Marchandot
- Division of Cardiovascular Medicine, Strasbourg University Hospital, Strasbourg, France
| | - Abdul Wahid Qureshi
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, FMTS, Strasbourg, France
| | - Dal-Seong Gong
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, FMTS, Strasbourg, France
| | - Cyril Auger
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, FMTS, Strasbourg, France
| | - Laurent Sattler
- Department Laboratory Haematology, Centre for Thrombosis and Haemostasis, Strasbourg University Hospital, Strasbourg, France
| | - Antje Reydel
- Division of Cardiovascular Medicine, Strasbourg University Hospital, Strasbourg, France
| | - Sébastien Hess
- Division of Cardiovascular Medicine, Strasbourg University Hospital, Strasbourg, France
| | - Walid Oulehri
- Department of Critical Care, Strasbourg University Hospital, Strasbourg, France
| | - Olivier Vollmer
- Department of Immunology and Internal Medicine, Strasbourg University Hospital, Strasbourg, France
| | - Jean-Marc Lessinger
- Biochemistry and Molecular Biology Laboratory, Strasbourg University Hospital, Strasbourg, France
| | - Nicolas Meyer
- Department of Biostatistics, Strasbourg University Hospital, Strasbourg, France
| | | | - Laurence Jesel
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, FMTS, Strasbourg, France; Division of Cardiovascular Medicine, Strasbourg University Hospital, Strasbourg, France
| | - Magnus Bäck
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden; Section of Translational Cardiology, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden; Institut National de la Sante et de la Recherche Medicale U1116, Université de Lorraine, Nancy, France
| | - Valérie Schini-Kerth
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, FMTS, Strasbourg, France.
| | - Olivier Morel
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine, FMTS, Strasbourg, France; Division of Cardiovascular Medicine, Strasbourg University Hospital, Strasbourg, France; Department Laboratory Haematology, Centre for Thrombosis and Haemostasis, Strasbourg University Hospital, Strasbourg, France; Vietnam National Heart Institute, Bach Mai Hospital, Hanoi, Vietnam.
| |
Collapse
|
16
|
Zisis SN, Durieux JC, Mouchati C, Funderburg N, Ailstock K, Chong M, Labbato D, McComsey GA. Arterial Stiffness and Oxidized LDL Independently Associated With Post-Acute Sequalae of SARS-CoV-2. Pathog Immun 2023; 8:1-15. [PMID: 38156116 PMCID: PMC10753933 DOI: 10.20411/pai.v8i2.634] [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: 09/28/2023] [Accepted: 11/11/2023] [Indexed: 12/30/2023] Open
Abstract
OBJECTIVE COVID-19 survivors can experience lingering symptoms known as post-acute sequelae of SARS-CoV-2 (PASC) that appear in different phenotypes, and its etiology remains elusive. We assessed the relationship of endothelial dysfunction with having COVID and PASC. METHODS Data was collected from a prospectively enrolled cohort (n=379) of COVID-negative and COVID-positive participants with and without PASC. Primary outcomes, endothelial function (measured by reactive hyperemic index [RHI]), and arterial elasticity (measured by augmentation index standardized at 75 bpm [AI]), were measured using the FDA approved EndoPAT. Patient characteristics, labs, metabolic measures, markers of inflammation, and oxidized LDL (ox-LDL) were collected at each study visit, and PASC symptoms were categorized into 3 non-exclusive phenotypes: cardiopulmonary, neurocognitive, and general. COVID-negative controls were propensity score matched to COVID-negative-infected cases using the greedy nearest neighbor method. RESULTS There were 14.3% of participants who were fully recovered COVID positive and 28.5% who were COVID positive with PASC, averaging 8.64 ± 6.26 total number of symptoms. The mean RHI was similar across the cohort and having COVID or PASC was not associated with endothelial function (P=0.33). Age (P<0.0001), female sex (P<0.0001), and CRP P=0.04) were positively associated with arterial stiffness, and COVID positive PASC positive with neurological and/or cardiopulmonary phenotypes had the worst arterial elasticity (highest AI). Values for AI (P=0.002) and ox-LDL (P<0.0001) were independently and positively associated with an increased likelihood of having PASC. CONCLUSION There is evidence of an independent association between PASC, ox-LDL, and arterial stiffness with neurological and/or cardiopulmonary phenotypes having the worst arterial elasticity. Future studies should continue investigating the role of oxidative stress in the pathophysiology of PASC.
Collapse
Affiliation(s)
- Sokratis N Zisis
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Jared C Durieux
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | - Nicholas Funderburg
- School of Health and Rehabilitation Sciences, Ohio State University, Columbus, Ohio
| | - Kate Ailstock
- School of Health and Rehabilitation Sciences, Ohio State University, Columbus, Ohio
| | - Mary Chong
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Danielle Labbato
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Grace A McComsey
- School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Clinical Research Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| |
Collapse
|
17
|
Paris D, Palomba L, Albertini MC, Tramice A, Motta L, Giammattei E, Ambrosino P, Maniscalco M, Motta A. The biomarkers' landscape of post-COVID-19 patients can suggest selective clinical interventions. Sci Rep 2023; 13:22496. [PMID: 38110483 PMCID: PMC10728085 DOI: 10.1038/s41598-023-49601-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 12/10/2023] [Indexed: 12/20/2023] Open
Abstract
In COVID-19 clinical symptoms can persist even after negativization also in individuals who have had mild or moderate disease. We here investigated the biomarkers that define the post-COVID-19 clinical state analyzing the exhaled breath condensate (EBC) of 38 post COVID-19 patients and 38 sex and age-matched healthy controls via nuclear magnetic resonance (NMR)-based metabolomics. Predicted gene-modulated microRNAs (miRNAs) related to COVID-19 were quantified from EBC of 10 patients and 10 controls. Finally, clinical parameters from all post-COVID-19 patients were correlated with metabolomic data. Post-COVID-19 patients and controls showed different metabolic phenotype ("metabotype"). From the metabolites, by using enrichment analysis we identified miRNAs that resulted up-regulated (hsa-miR146a-5p) and down-regulated (hsa-miR-126-3p and hsa-miR-223-3p) in post-COVID-19. Taken together, our multiomics data indicate that post-COVID-19 patients before rehabilitation are characterized by persistent inflammation, dysregulation of liver, endovascular thrombotic and pulmonary processes, and physical impairment, which should be the primary clinical targets to contrast the post-acute sequelae of COVID-19.
Collapse
Affiliation(s)
- Debora Paris
- Institute of Biomolecular Chemistry, National Research Council, 80078, Pozzuoli (Naples), Italy
| | - Letizia Palomba
- Department of Biomolecular Sciences, "Carlo Bo" University, 61029, Urbino, Italy
| | | | - Annabella Tramice
- Institute of Biomolecular Chemistry, National Research Council, 80078, Pozzuoli (Naples), Italy
| | - Lorenzo Motta
- Neuroradiology Unit, Ospedale Santa Maria Della Misericordia, 45100, Rovigo, Italy
- IRCCS Istituto Delle Scienze Neurologiche (Padiglione G), via Altura 3, 40139, Bologna, Italy
| | - Eleonora Giammattei
- Department of Biomolecular Sciences, "Carlo Bo" University, 61029, Urbino, Italy
| | - Pasquale Ambrosino
- Directorate of Telese Terme Institute, Istituti Clinici Scientifici Maugeri IRCCS, 82037, Telese Terme (Benevento), Italy
| | - Mauro Maniscalco
- Pulmonary Rehabilitation Unit of the Telese Terme Institute, Istituti Clinici Scientifici Maugeri IRCCS, 82037, Telese Terme (Benevento), Italy.
- Department of Clinical Medicine and Surgery, Section of Respiratory Disease, University of Naples Federico II, 80131, Naples, Italy.
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council, 80078, Pozzuoli (Naples), Italy.
| |
Collapse
|
18
|
Astley C, Prado DMLD, Sieczkowska SM, Esteves GP, Suguita P, Fink T, Lindoso L, Matsuo O, Martins F, Bain V, Badue Pereira MF, Marques HH, Malluf A, Leal GN, Silva CA, Roschel H, Gualano B. Impaired cardiorespiratory fitness and endothelial function after SARS-CoV-2 infection in a sample of mainly immunocompromised youth. J Appl Physiol (1985) 2023; 135:1323-1329. [PMID: 37916270 DOI: 10.1152/japplphysiol.00213.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 11/03/2023] Open
Abstract
This study aimed to compare cardiopulmonary fitness and endothelial function 6 months after hospital diagnosis in a sample mainly comprising immunocompromised patients with confirmed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection versus noninfected controls. Youth (n = 30; age: 14 yr; 60% females) with confirmed SARS-CoV-2 seen in a tertiary hospital of Sao Paulo, Brazil, were matched by propensity score based on BMI, age, sex, and pre-existing diseases with a control group who had not been tested positive for SARS-CoV-2 infection (n = 30; age: 15 yr; 50% females). Cardiopulmonary fitness (by means of a cardiopulmonary exercise test: CPET) and brachial flow-mediated dilation (%b-FMD) were assessed 3-6 mo after diagnosis. Patients were matched by propensity score based on BMI, age, sex and pre-existing diseases, if any, with a control group who had not been tested positive for SARS-CoV-2. Compared with controls, patients with COVID-19 showed reduced ventilatory anaerobic threshold (VAT) and peak exercise time and minute ventilation/maximum voluntary ventilation (V̇e/MVV) (all P < 0.01). Brachial endothelial function variables were all adjusted for body surface area (BSA). Patients with COVID-19 had decreased %b-FMD (3.6 vs. 5.4; P = 0.03) mean and positive flow (P = 0.02 and P = 0.03, respectively) versus controls. Adjusted linear regression models exploring associations between CPET variables, %b-FMD and the potential predictors post-COVID-19 syndrome, number of symptoms, hospitalization, and COVID severity did not detect significant associations, except for total shear rate in hospitalization (coefficient: -65.07 [95%CI -119.5;-10.5], P = 0.02). Immunocompromised and previously healthy children and adolescents with COVID-19 presented with impaired exercise capacity and endothelial dysfunction when compared with their noninfected counterparts, but the mechanisms remain unknown.NEW & NOTEWORTHY COVID-19 appeared to impair recovery of exercise capacity and endothelial function in a sample mainly comprising immunocompromised patients, but the mechanisms are unknown. These findings support the need for preventive measures against COVID-19 in this vulnerable population and suggest the necessity of proper monitoring and treatment for these patients.
Collapse
Affiliation(s)
- Camilla Astley
- Center of Lifestyle Medicine, Applied Physiology & Nutrition Research Group, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
- Rheumatology Division, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Danilo Marcelo Leite do Prado
- Center of Lifestyle Medicine, Applied Physiology & Nutrition Research Group, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Sofia Mendes Sieczkowska
- Center of Lifestyle Medicine, Applied Physiology & Nutrition Research Group, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Gabriel P Esteves
- Center of Lifestyle Medicine, Applied Physiology & Nutrition Research Group, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Priscila Suguita
- Children and Adolescent Institute, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Thais Fink
- Children and Adolescent Institute, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Livia Lindoso
- Children and Adolescent Institute, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Olivia Matsuo
- Children and Adolescent Institute, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Fernanda Martins
- Children and Adolescent Institute, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Vera Bain
- Children and Adolescent Institute, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Maria Fernanda Badue Pereira
- Children and Adolescent Institute, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Heloisa Helena Marques
- Children and Adolescent Institute, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Adriana Malluf
- Children and Adolescent Institute, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Gabriela Nunes Leal
- Children and Adolescent Institute, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Clovis Artur Silva
- Children and Adolescent Institute, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Hamilton Roschel
- Center of Lifestyle Medicine, Applied Physiology & Nutrition Research Group, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
- Rheumatology Division, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Bruno Gualano
- Center of Lifestyle Medicine, Applied Physiology & Nutrition Research Group, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
- Rheumatology Division, Clinical Hospital, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
- Food Research Center, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
19
|
Hu Y, Liu Y, Zheng H, Liu L. Risk Factors for Long COVID in Older Adults. Biomedicines 2023; 11:3002. [PMID: 38002002 PMCID: PMC10669899 DOI: 10.3390/biomedicines11113002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
As time has passed following the COVID-19 pandemic, individuals infected with SARS-CoV-2 have gradually exhibited a variety of symptoms associated with long COVID in the postacute phase of infection. Simultaneously, in many countries worldwide, the process of population aging has been accelerating. Within this context, the elderly population has not only become susceptible and high-risk during the acute phase of COVID-19 but also has considerable risks when confronting long COVID. Elderly individuals possess specific immunological backgrounds, and during the process of aging, their immune systems can enter a state known as "immunosenescence". This further exacerbates "inflammaging" and the development of various comorbidities in elderly individuals, rendering them more susceptible to long COVID. Additionally, long COVID can inflict both physical and mental harm upon elderly people, thereby reducing their overall quality of life. Consequently, the impact of long COVID on elderly people should not be underestimated. This review seeks to summarize the infection characteristics and intrinsic factors of older adults during the COVID-19 pandemic, with a focus on the physical and mental impact of long COVID. Additionally, it aims to explore potential strategies to mitigate the risk of long COVID or other emerging infectious diseases among older adults in the future.
Collapse
Affiliation(s)
| | | | | | - Longding Liu
- Key Laboratory of Systemic Innovative Research on Virus Vaccines, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China; (Y.H.); (Y.L.); (H.Z.)
| |
Collapse
|
20
|
Ambrosino P, Di Minno MND, D'Anna SE, Formisano R, Pappone N, Mancusi C, Molino A, Motta A, Maniscalco M. Pulmonary rehabilitation and endothelial function in patients with chronic obstructive pulmonary disease: A prospective cohort study. Eur J Intern Med 2023; 116:96-105. [PMID: 37349204 DOI: 10.1016/j.ejim.2023.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/19/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is associated with subclinical atherosclerosis and endothelial dysfunction, thereby leading to increased cardiovascular risk. In the present study, we evaluated the changes in endothelium-dependent flow-mediated dilation (FMD) in a cohort of severe COPD patients undergoing pulmonary rehabilitation. METHODS Consecutive COPD patients referred to our Pulmonary Rehabilitation Unit were screened for inclusion. All study procedures were performed at hospital admission and discharge. RESULTS Of 78 patients screened for eligibility, a total of 40 participants (67.5% males, median age 72.5 years) were included. After pulmonary rehabilitation, a significant improvement in functional parameters, exercise capacity, and measures of disability and quality of life were documented. FMD changed from 3.25% (IQR: 2.31-4.26) to 4.95% (IQR: 3.57-6.02), corresponding to a 52.3% increase of its median value (P < 0.001). Significantly lower changes in FMD were documented in COPD patients with hypercholesterolemia as compared to those without (+0.33% ± 1.61 vs. +1.62% ± 1.59, P = 0.037). Changes in FMD (ΔFMD) were positively associated with changes in forced expiratory volume in 1 s (FEV1), when expressed both as absolute values (ΔFEV1) (r = 0.503, P = 0.002) and as percentages of predicted values (ΔFEV1%) (r = 0.608; P < 0.001). In multiple linear regressions, after adjusting for major cardiovascular risk factors, ΔFEV1 (β=0.342; P = 0.049) and ΔFEV1% (β=0.480; P = 0.015) were both confirmed as independent predictors of ΔFMD. CONCLUSIONS Results of our study suggest that endothelial function may improve in COPD after pulmonary rehabilitation. The potential beneficial effect in terms of cardiovascular risk prevention should be evaluated in ad hoc designed studies.
Collapse
Affiliation(s)
- Pasquale Ambrosino
- Istituti Clinici Scientifici Maugeri IRCCS, Directorate of Telese Terme Institute, Italy
| | | | - Silvestro Ennio D'Anna
- Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, Italy
| | - Roberto Formisano
- Istituti Clinici Scientifici Maugeri IRCCS, Cardiac Rehabilitation Unit of Telese Terme Institute, Italy
| | - Nicola Pappone
- Istituti Clinici Scientifici Maugeri IRCCS, Neuromotor Rehabilitation Unit of Telese Terme Institute, Italy
| | - Costantino Mancusi
- Department of Advanced Biomedical Science, Federico II University, Naples, Italy
| | - Antonio Molino
- Department of Clinical Medicine and Surgery, Federico II University, Naples, 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; Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, Italy.
| |
Collapse
|
21
|
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
|
22
|
Owens CD, Pinto CB, Detwiler S, Mukli P, Peterfi A, Szarvas Z, Hoffmeister JR, Galindo J, Noori J, Kirkpatrick AC, Dasari TW, James J, Tarantini S, Csiszar A, Ungvari Z, Prodan CI, Yabluchanskiy A. Cerebral small vessel disease pathology in COVID-19 patients: A systematic review. Ageing Res Rev 2023; 88:101962. [PMID: 37224885 PMCID: PMC10202464 DOI: 10.1016/j.arr.2023.101962] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/20/2023] [Accepted: 05/21/2023] [Indexed: 05/26/2023]
Abstract
Cerebral small vessel disease (CSVD) is the leading cause of vascular cognitive impairment and is associated with COVID-19. However, contributing factors that often accompany CSVD pathology in COVID-19 patients may influence the incidence of cerebrovascular complications. Thus, a mechanism linking COVID-19 and CSVD has yet to be uncovered and differentiated from age-related comorbidities (i.e., hypertension), and medical interventions during acute infection. We aimed to evaluate CSVD in acute and recovered COVID-19 patients and to differentiate COVID-19-related cerebrovascular pathology from the above-mentioned contributing factors by assessing the localization of microbleeds and ischemic lesions/infarctions in the cerebrum, cerebellum, and brainstem. A systematic search was performed in December 2022 on PubMed, Web of Science, and Embase using a pre-established search criterion related to history of, or active COVID-19 with CSVD pathology in adults. From a pool of 161 studies, 59 met eligibility criteria and were included. Microbleeds and ischemic lesions had a strong predilection for the corpus callosum and subcortical/deep white matter in COVID-19 patients, suggesting a distinct CSVD pathology. These findings have important implications for clinical practice and biomedical research as COVID-19 may independently, and through exacerbation of age-related mechanisms, contribute to increased incidence of CSVD.
Collapse
Affiliation(s)
- Cameron D Owens
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Camila Bonin Pinto
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sam Detwiler
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Anna Peterfi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Zsofia Szarvas
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Jordan R Hoffmeister
- Department of Psychiatry and Behavioral Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Juliette Galindo
- Department of Psychiatry and Behavioral Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jila Noori
- Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Angelia C Kirkpatrick
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Tarun W Dasari
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Judith James
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA; Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
23
|
Nunes JM, Kell DB, Pretorius E. Cardiovascular and haematological pathology in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS): A role for viruses. Blood Rev 2023; 60:101075. [PMID: 36963989 PMCID: PMC10027292 DOI: 10.1016/j.blre.2023.101075] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
ME/CFS is a debilitating chronic condition that often develops after viral or bacterial infection. Insight from the study of Long COVID/Post Acute Sequelae of COVID-19 (PASC), the post-viral syndrome associated with SARS-CoV-2 infection, might prove to be useful for understanding pathophysiological mechanisms of ME/CFS. Disease presentation is similar between the two conditions, and a subset of Long COVID patients meet the diagnostic criteria for ME/CFS. Since Long COVID is characterized by significant vascular pathology - including endothelial dysfunction, coagulopathy, and vascular dysregulation - the question of whether or not the same biological abnormalities are of significance in ME/CFS arises. Cardiac abnormalities have for a while now been documented in ME/CFS cohorts, with recent studies demonstrating major deficits in cerebral blood flow, and hence vascular dysregulation. A growing body of research is demonstrating that ME/CFS is accompanied by platelet hyperactivation, anomalous clotting, a procoagulant phenotype, and endothelial dysfunction. Endothelial damage and dysregulated clotting can impair substance exchange between blood and tissues, and result in hypoperfusion, which may contribute to the manifestation of certain ME/CFS symptoms. Here we review the ME/CFS literature to summarize cardiovascular and haematological findings documented in patients with the condition, and, in this context, briefly discuss the potential role of previously-implicated pathogens. Overall, cardiac and haematological abnormalities are present within ME/CFS cohorts. While atherosclerotic heart disease is not significantly associated with ME/CFS, suboptimal cardiovascular function defined by reduced cardiac output, impaired cerebral blood flow, and vascular dysregulation are, and these abnormalities do not appear to be influenced by deconditioning. Rather, these cardiac abnormalities may result from dysfunction in the (autonomic) nervous system. Plenty of recently published studies are demonstrating significant platelet hyperactivity and endothelial dysfunction in ME/CFS, as well as anomalous clotting processes. It is of particular importance to determine to what extent these cardiovascular and haematological abnormalities contribute to symptom severity, and if these two systems can be targeted for therapeutic purposes. Viral reservoirs of herpesviruses exist in ME/CFS, and most likely contribute to cardiovascular and haematological dysfunction directly or indirectly. This review highlights the potential of studying cardiac functioning, the vasculature, and coagulation system in ME/CFS.
Collapse
Affiliation(s)
- Jean M Nunes
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa.
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa; Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool L69 7ZB, UK; The Novo Nordisk Foundation Centre for Biosustainability, Building 220, Chemitorvet 200, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa; Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool L69 7ZB, UK.
| |
Collapse
|
24
|
Komaroff AL, Lipkin WI. ME/CFS and Long COVID share similar symptoms and biological abnormalities: road map to the literature. Front Med (Lausanne) 2023; 10:1187163. [PMID: 37342500 PMCID: PMC10278546 DOI: 10.3389/fmed.2023.1187163] [Citation(s) in RCA: 111] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Some patients remain unwell for months after "recovering" from acute COVID-19. They develop persistent fatigue, cognitive problems, headaches, disrupted sleep, myalgias and arthralgias, post-exertional malaise, orthostatic intolerance and other symptoms that greatly interfere with their ability to function and that can leave some people housebound and disabled. The illness (Long COVID) is similar to myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) as well as to persisting illnesses that can follow a wide variety of other infectious agents and following major traumatic injury. Together, these illnesses are projected to cost the U.S. trillions of dollars. In this review, we first compare the symptoms of ME/CFS and Long COVID, noting the considerable similarities and the few differences. We then compare in extensive detail the underlying pathophysiology of these two conditions, focusing on abnormalities of the central and autonomic nervous system, lungs, heart, vasculature, immune system, gut microbiome, energy metabolism and redox balance. This comparison highlights how strong the evidence is for each abnormality, in each illness, and helps to set priorities for future investigation. The review provides a current road map to the extensive literature on the underlying biology of both illnesses.
Collapse
Affiliation(s)
- Anthony L. Komaroff
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - W. Ian Lipkin
- Center for Infection and Immunity, Mailman School of Public Health, Vagelos College of Physicians and Surgeons of Columbia University, New York, NY, United States
| |
Collapse
|
25
|
Gyöngyösi M, Alcaide P, Asselbergs FW, Brundel BJJM, Camici GG, Martins PDC, Ferdinandy P, Fontana M, Girao H, Gnecchi M, Gollmann-Tepeköylü C, Kleinbongard P, Krieg T, Madonna R, Paillard M, Pantazis A, Perrino C, Pesce M, Schiattarella GG, Sluijter JPG, Steffens S, Tschöpe C, Van Linthout S, Davidson SM. Long COVID and the cardiovascular system-elucidating causes and cellular mechanisms in order to develop targeted diagnostic and therapeutic strategies: a joint Scientific Statement of the ESC Working Groups on Cellular Biology of the Heart and Myocardial and Pericardial Diseases. Cardiovasc Res 2023; 119:336-356. [PMID: 35875883 PMCID: PMC9384470 DOI: 10.1093/cvr/cvac115] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/27/2022] [Accepted: 07/01/2022] [Indexed: 02/07/2023] Open
Abstract
Long COVID has become a world-wide, non-communicable epidemic, caused by long-lasting multiorgan symptoms that endure for weeks or months after SARS-CoV-2 infection has already subsided. This scientific document aims to provide insight into the possible causes and therapeutic options available for the cardiovascular manifestations of long COVID. In addition to chronic fatigue, which is a common symptom of long COVID, patients may present with chest pain, ECG abnormalities, postural orthostatic tachycardia, or newly developed supraventricular or ventricular arrhythmias. Imaging of the heart and vessels has provided evidence of chronic, post-infectious perimyocarditis with consequent left or right ventricular failure, arterial wall inflammation, or microthrombosis in certain patient populations. Better understanding of the underlying cellular and molecular mechanisms of long COVID will aid in the development of effective treatment strategies for its cardiovascular manifestations. A number of mechanisms have been proposed, including those involving direct effects on the myocardium, microthrombotic damage to vessels or endothelium, or persistent inflammation. Unfortunately, existing circulating biomarkers, coagulation, and inflammatory markers, are not highly predictive for either the presence or outcome of long COVID when measured 3 months after SARS-CoV-2 infection. Further studies are needed to understand underlying mechanisms, identify specific biomarkers, and guide future preventive strategies or treatments to address long COVID and its cardiovascular sequelae.
Collapse
Affiliation(s)
- Mariann Gyöngyösi
- Division of Cardiology, 2nd Department of Internal Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Folkert W Asselbergs
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Health Data Research UK and Institute of Health Informatics, University College London, London, UK
| | - Bianca J J M Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital, Zurich, Switzerland
| | - Paula da Costa Martins
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Department of Molecular Genetics, Faculty of Sciences and Engineering, Maastricht University, Maastricht, The Netherlands
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Marianna Fontana
- Division of Medicine, Royal Free Hospital London, University College London, London, UK
| | - Henrique Girao
- Center for Innovative Biomedicine and Biotechnology (CIBB), Clinical Academic Centre of Coimbra (CACC), Faculty of Medicine, Univ Coimbra, Institute for Clinical and Biomedical Research (iCBR), Coimbra, Portugal
| | - Massimiliano Gnecchi
- Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Pavia, Italy
- Unit of Translational Cardiology, Division of Cardiology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Petra Kleinbongard
- Institut für Pathophysiologie, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Essen, Germany
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Rosalinda Madonna
- Department of Pathology, Institute of Cardiology, University of Pisa, Pisa, Italy
| | - Melanie Paillard
- Laboratoire CarMeN-équipe IRIS, INSERM, INRA, Université Claude Bernard Lyon-1, INSA-Lyon, Univ-Lyon, 69500 Bron, France
| | - Antonis Pantazis
- National Heart and Lung Institute, Imperial College London, London, UK
- Cardiovascular Research Centre at Royal Brompton and Harefield Hospitals, London, UK
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale cardiovascolare, Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Gabriele G Schiattarella
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
- Center for Cardiovascular Research (CCR), Department of Cardiology, Charité—Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Joost P G Sluijter
- Laboratory of Experimental Cardiology, Cardiology, UMC Utrecht Regenerative Medicine Center, Utrecht, The Netherlands
- Circulatory Health Laboratory, Utrecht University, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Munich, Germany
- Germany and Munich Heart Alliance, DZHK Partner Site Munich, Munich, Germany
| | - Carsten Tschöpe
- Berlin Institute of Health (BIH) at Charité, Universitätmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), German Center for Cardiovascular Research (DZHK), Partner site Berlin and Dept Cardiology (CVK), Charité, Berlin, Germany
| | - Sophie Van Linthout
- Berlin Institute of Health (BIH) at Charité, Universitätmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), German Center for Cardiovascular Research (DZHK), Partner site Berlin and Dept Cardiology (CVK), Charité, Berlin, Germany
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, WC1E 6HX London, UK
| |
Collapse
|
26
|
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: 3.0] [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
|
27
|
Conti V, Corbi G, Sabbatino F, De Pascale D, Sellitto C, Stefanelli B, Bertini N, De Simone M, Liguori L, Di Paola I, De Bernardo M, Tesse A, Rosa N, Pagliano P, Filippelli A. Long COVID: Clinical Framing, Biomarkers, and Therapeutic Approaches. J Pers Med 2023; 13:334. [PMID: 36836568 PMCID: PMC9959656 DOI: 10.3390/jpm13020334] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/09/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023] Open
Abstract
More than two years after the onset of the COVID-19 pandemic, healthcare providers are facing an emergency within an emergency, the so-called long COVID or post-COVID-19 syndrome (PCS). Patients diagnosed with PCS develop an extended range of persistent symptoms and/or complications from COVID-19. The risk factors and clinical manifestations are many and various. Advanced age, sex/gender, and pre-existing conditions certainly influence the pathogenesis and course of this syndrome. However, the absence of precise diagnostic and prognostic biomarkers may further complicate the clinical management of patients. This review aimed to summarize recent evidence on the factors influencing PCS, possible biomarkers, and therapeutic approaches. Older patients recovered approximately one month earlier than younger patients, with higher rates of symptoms. Fatigue during the acute phase of COVID-19 appears to be an important risk factor for symptom persistence. Female sex, older age, and active smoking are associated with a higher risk of developing PCS. The incidence of cognitive decline and the risk of death are higher in PCS patients than in controls. Complementary and alternative medicine appears to be associated with improvement in symptoms, particularly fatigue. The heterogeneous nature of post-COVID symptoms and the complexity of patients with PCS, who are often polytreated due to concomitant clinical conditions, suggest a holistic and integrated approach to provide useful guidance for the treatment and overall management of long COVID.
Collapse
Affiliation(s)
- Valeria Conti
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi, Italy
- Clinical Pharmacology and Pharmacogenetics Unit, University Hospital "San Giovanni di Dio e Ruggi, D'Aragona", 84131 Salerno, Italy
| | - Graziamaria Corbi
- Department of Translational Medical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Francesco Sabbatino
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Domenico De Pascale
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi, Italy
- Clinical Pharmacology and Pharmacogenetics Unit, University Hospital "San Giovanni di Dio e Ruggi, D'Aragona", 84131 Salerno, Italy
| | - Carmine Sellitto
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi, Italy
- Clinical Pharmacology and Pharmacogenetics Unit, University Hospital "San Giovanni di Dio e Ruggi, D'Aragona", 84131 Salerno, Italy
| | - Berenice Stefanelli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi, Italy
- Clinical Pharmacology and Pharmacogenetics Unit, University Hospital "San Giovanni di Dio e Ruggi, D'Aragona", 84131 Salerno, Italy
| | - Nicola Bertini
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi, Italy
- Clinical Pharmacology and Pharmacogenetics Unit, University Hospital "San Giovanni di Dio e Ruggi, D'Aragona", 84131 Salerno, Italy
| | - Matteo De Simone
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Luigi Liguori
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80131 Naples, Italy
| | - Ilenia Di Paola
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Maddalena De Bernardo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Angela Tesse
- CNRS, INSERM, L'institut du Thorax, Université de Nantes, F-44000 Nantes, France
| | - Nicola Rosa
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Pasquale Pagliano
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi, Italy
| | - Amelia Filippelli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Via S. Allende, 84081 Baronissi, Italy
- Clinical Pharmacology and Pharmacogenetics Unit, University Hospital "San Giovanni di Dio e Ruggi, D'Aragona", 84131 Salerno, Italy
| |
Collapse
|
28
|
Chen H, Peng J, Wang T, Wen J, Chen S, Huang Y, Zhang Y. Counter-regulatory renin-angiotensin system in hypertension: Review and update in the era of COVID-19 pandemic. Biochem Pharmacol 2023; 208:115370. [PMID: 36481346 PMCID: PMC9721294 DOI: 10.1016/j.bcp.2022.115370] [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: 10/12/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Cardiovascular disease is the major cause of mortality and disability, with hypertension being the most prevalent risk factor. Excessive activation of the renin-angiotensin system (RAS) under pathological conditions, leading to vascular remodeling and inflammation, is closely related to cardiovascular dysfunction. The counter-regulatory axis of the RAS consists of angiotensin-converting enzyme 2 (ACE2), angiotensin (1-7), angiotensin (1-9), alamandine, proto-oncogene Mas receptor, angiotensin II type-2 receptor and Mas-related G protein-coupled receptor member D. Each of these components has been shown to counteract the effects of the overactivated RAS. In this review, we summarize the latest insights into the complexity and interplay of the counter-regulatory RAS axis in hypertension, highlight the pathophysiological functions of ACE2, a multifunctional molecule linking hypertension and COVID-19, and discuss the function and therapeutic potential of targeting this counter-regulatory RAS axis to prevent and treat hypertension in the context of the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Hongyin Chen
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518000, Guangdong, China
| | - Jiangyun Peng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Tengyao Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Jielu Wen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Sifan Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, Guangdong, China,Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, Guangdong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China,Corresponding authors
| | - Yang Zhang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518000, Guangdong, China,Corresponding authors
| |
Collapse
|
29
|
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
|
30
|
Oikonomou E, Lampsas S, Theofilis P, Souvaliotis N, Papamikroulis GA, Katsarou O, Kalogeras K, Pantelidis P, Papaioannou TG, Tsatsaragkou A, Marinos G, Siasos G, Tousoulis D, Vavuranakis M. Impaired left ventricular deformation and ventricular-arterial coupling in post-COVID-19: association with autonomic dysregulation. Heart Vessels 2023; 38:381-393. [PMID: 36169708 PMCID: PMC9516516 DOI: 10.1007/s00380-022-02180-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 09/22/2022] [Indexed: 02/07/2023]
Abstract
Coronavirus disease-19 (COVID-19) has extended implications namely the long COVID-19 syndrome. We assessed over-time changes in left ventricular (LV) function, aortic stiffness, autonomic function, and ventricular-arterial coupling (VAC) in post-COVID-19 patients. We followed 34 post-COVID-19 subjects, up to 6 months post-hospital discharge. Subjects without COVID-19 served as control. We evaluated LV global longitudinal strain (LV-GLS), arterial stiffness [carotid-femoral pulse wave velocity (cf-PWV)], and heart rate variability -standard deviation of normal RR intervals (SDNN). VAC was estimated as the ratio of cf-PWV to LV-GLS. Post-COVID-19 individuals (1-month post-hospital discharge) presented with impaired LV-GLS [-18.4%(3.1) vs. -22.0%(2.7), P < 0.001], cf-PWV [12.1 m/s (3.2) vs. 9.6 m/s (1.9), P < 0.001], SDNN [111.3 ms (22.6) vs. 147.2 ms (14.0), P < 0.001], and VAC [-0.68 (0.22) vs. -0.44 (0.10), P < 0.001] compared to control. LV-GLS, SDNN, and VAC improved at the 6-month follow-up however they did not reach control levels. In post-COVID-19 subjects, SDNN and VAC were correlated at the 1-month (R = 0.499, P = 0.003) and 6-month (R = 0.372, P = 0.04) follow-up. Long COVID-19 syndrome was associated with impaired LV-GLS, SDNN, and VAC. Post-COVID-19 subjects presented with autonomic dysregulation associated with aortic stiffness, ventricular-arterial impairment, and LV dysfunction, even 6-months post-hospital discharge. These abnormalities may be related to the presence of long COVID-19 syndrome.
Collapse
Affiliation(s)
- Evangelos Oikonomou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Mesogeion 152, 11527 Athens, Greece ,1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Athens, Greece
| | - Stamatios Lampsas
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Mesogeion 152, 11527 Athens, Greece
| | - Panagiotis Theofilis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Athens, Greece
| | - Nektarios Souvaliotis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Mesogeion 152, 11527 Athens, Greece
| | - George Aggelos Papamikroulis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Mesogeion 152, 11527 Athens, Greece
| | - Ourania Katsarou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Mesogeion 152, 11527 Athens, Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Mesogeion 152, 11527 Athens, Greece ,1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Athens, Greece
| | - Panteleimon Pantelidis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Mesogeion 152, 11527 Athens, Greece
| | - Theodore G. Papaioannou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Mesogeion 152, 11527 Athens, Greece
| | - Aikaterini Tsatsaragkou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Mesogeion 152, 11527 Athens, Greece
| | - Georgios Marinos
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Mesogeion 152, 11527 Athens, Greece ,Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Dimitris Tousoulis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Athens, Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Mesogeion 152, 11527 Athens, Greece ,1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Athens, Greece
| |
Collapse
|
31
|
Ismail AMA. Erectile dysfunction: the non-utilized role of exercise rehabilitation for the most embarrassing forgotten post-COVID complication in men. Aging Male 2022; 25:217-218. [PMID: 35946582 DOI: 10.1080/13685538.2022.2108013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Ali Mohamed Ali Ismail
- Department of Physical Therapy for Cardiovascular/Respiratory Disorder and Geriatrics, Faculty of Physical Therapy, Cairo University, Giza, Egypt
| |
Collapse
|
32
|
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: 16] [Impact Index Per Article: 5.3] [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
|
33
|
Theofilis P, Lampsas S, Oikonomou E, Siasos G, Vavuranakis MA, Marinos G, Tsioufis K, Vavuranakis M, Tousoulis D. Endothelial dysfunction in convalescent COVID-19 patients: Systematic review and meta-analysis. Arch Cardiovasc Dis 2022; 115:675-677. [PMID: 36335024 PMCID: PMC9595447 DOI: 10.1016/j.acvd.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Panagiotis Theofilis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Vas. Sophias 114, 11527 Athens, Greece,Corresponding author
| | - Stamatios Lampsas
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Evangelos Oikonomou
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Vas. Sophias 114, 11527 Athens, Greece,3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece,Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, 02115 Boston, MA, USA
| | - Michael Andrew Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Georgios Marinos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Kostas Tsioufis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Vas. Sophias 114, 11527 Athens, Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, 11527 Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Vas. Sophias 114, 11527 Athens, Greece
| |
Collapse
|
34
|
Province VM, Szeghy RE, Stute NL, Augenreich MA, Behrens CE, Stickford JL, Stickford ASL, Ratchford S. Tracking peripheral vascular function for six months in young adults following SARS-CoV-2 infection. Physiol Rep 2022; 10:e15552. [PMID: 36541342 PMCID: PMC9768737 DOI: 10.14814/phy2.15552] [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: 11/09/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023] Open
Abstract
SARS-CoV-2 infection is known to instigate a range of physiologic perturbations, including vascular dysfunction. However, little work has concluded how long these effects may last, especially among young adults with mild symptoms. To determine potential recovery from acute vascular dysfunction in young adults (8 M/8F, 21 ± 1 yr, 23.5 ± 3.1 kg⋅m-2 ), we longitudinally tracked brachial artery flow-mediated dilation (FMD) and reactive hyperemia (RH) in the arm and hyperemic response to passive limb movement (PLM) in the leg, with Doppler ultrasound, as well as circulating biomarkers of inflammation (interleukin-6, C-reactive protein), oxidative stress (thiobarbituric acid reactive substances, protein carbonyl), antioxidant capacity (superoxide dismutase), and nitric oxide bioavailability (nitrite) monthly for a 6-month period post-SARS-CoV-2 infection. FMD, as a marker of macrovascular function, improved from month 1 (3.06 ± 1.39%) to month 6 (6.60 ± 2.07%; p < 0.001). FMD/Shear improved from month one (0.10 ± 0.06 AU) to month six (0.18 ± 0.70 AU; p = 0.002). RH in the arm and PLM in the leg, as markers of microvascular function, did not change during the 6 months (p > 0.05). Circulating markers of inflammation, oxidative stress, antioxidant capacity, and nitric oxide bioavailability did not change during the 6 months (p > 0.05). Together, these results suggest some improvements in macrovascular, but not microvascular function, over 6 months following SARS-CoV-2 infection. The data also suggest persistent ramifications for cardiovascular health among those recovering from mild illness and among young, otherwise healthy adults with SARS-CoV-2.
Collapse
Affiliation(s)
- Valesha M. Province
- Department of Health & Exercise ScienceAppalachian State UniversityBooneNorth CarolinaUSA
| | - Rachel E. Szeghy
- Department of Health & Exercise ScienceAppalachian State UniversityBooneNorth CarolinaUSA
| | - Nina L. Stute
- Department of Health & Exercise ScienceAppalachian State UniversityBooneNorth CarolinaUSA
| | - Marc A. Augenreich
- Department of Health & Exercise ScienceAppalachian State UniversityBooneNorth CarolinaUSA
| | - Christian E. Behrens
- Department of Health & Exercise ScienceAppalachian State UniversityBooneNorth CarolinaUSA
| | - Jonathon L. Stickford
- Department of Health & Exercise ScienceAppalachian State UniversityBooneNorth CarolinaUSA
| | | | - Stephen M. Ratchford
- Department of Health & Exercise ScienceAppalachian State UniversityBooneNorth CarolinaUSA
| |
Collapse
|
35
|
Sabioni L, De Lorenzo A, Castro-Faria-Neto HC, Estato V, Tibirica E. Long-term assessment of systemic microcirculatory function and plasma cytokines after coronavirus disease 2019 (COVID-19). Braz J Infect Dis 2022; 27:102719. [PMID: 36423696 PMCID: PMC9670297 DOI: 10.1016/j.bjid.2022.102719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/18/2022] [Accepted: 11/06/2022] [Indexed: 11/18/2022] Open
Abstract
Systemic microvascular dysfunction has been shown to be present in COVID-19, and serum cytokines are known to be involved in the regulation of vascular function. We sought to evaluate systemic microvascular endothelial function, with laser doppler perfusion monitoring (LDPM), and plasma levels of cytokines after acute COVID-19. Individuals admitted to a Cardiology hospital with acute COVID-19 and followed for 12-15 months after recovery underwent noninvasive evaluation of systemic endothelium-dependent microvascular reactivity by cutaneous LDPM with local thermal hyperemia (LTH). A multiplex biometric immunoassay panel was used to assess 48 serum cytokines and chemokines. Twenty patients and 14 control volunteers were enrolled. The areas under the curves of vasodilation induced by LTH were significantly increased after recovery (P=0.009) and were not different from values obtained in healthy volunteers (P = 0.85). The peak microvascular flow during LTH did also significantly increase (P = 0.02), and was not different form values obtained in healthy volunteers (P = 0.55). Several cytokines displayed significantly reduced serum concentrations after recovery from COVID-19. In conclusion, endothelium-dependent systemic microvascular reactivity improved after recovery from COVID-19 in patients with cardiovascular diseases, in parallel with a reduction in the levels of several serum cytokines and chemokines involved in the regulation of vascular function and inflammation.
Collapse
Affiliation(s)
- Letícia Sabioni
- Instituto Nacional de Cardiologia, Ministério da Saúde, Rio de Janeiro, RJ, Brazil
| | - Andrea De Lorenzo
- Instituto Nacional de Cardiologia, Ministério da Saúde, Rio de Janeiro, RJ, Brazil
| | | | - Vanessa Estato
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Eduardo Tibirica
- Instituto Nacional de Cardiologia, Ministério da Saúde, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
36
|
Mavraganis G, Dimopoulou MA, Delialis D, Bampatsias D, Patras R, Sianis A, Maneta E, Stamatelopoulos K, Georgiopoulos G. Clinical implications of vascular dysfunction in acute and convalescent COVID-19: A systematic review. Eur J Clin Invest 2022; 52:e13859. [PMID: 35986716 PMCID: PMC9539033 DOI: 10.1111/eci.13859] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/10/2022] [Accepted: 08/14/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Accumulating evidence suggests that endothelial dysfunction is implicated in the pathogenesis and severity of coronavirus disease 2019 (COVID-19). In this context, vascular impairment in COVID-19 might be associated with clinical manifestations and could refine risk stratification in these patients. METHODS This systematic review aims to synthesize current evidence on the frequency and the prognostic value of vascular dysfunction during acute and post-recovery COVID-19. After systematically searching the MEDLINE, clinicaltrials.gov and the Cochrane Library from 1 December 2019 until 05 March 2022, we identified 24 eligible studies with laboratory confirmed COVID-19 and a thorough examination of vascular function. Flow-mediated dilation (FMD) was assessed in 5 and 12 studies in acute and post-recovery phase respectively; pulse wave velocity (PWV) was the marker of interest in three studies in the acute and four studies in the post-recovery phase. RESULTS All studies except for one in the acute and in the post-recovery phase showed positive association between vascular dysfunction and COVID-19 infection. Endothelial dysfunction in two studies and increased arterial stiffness in three studies were related to inferior survival in COVID-19. DISCUSSION Overall, a detrimental effect of COVID-19 on markers of endothelial function and arterial stiffness that could persist even for months after the resolution of the infection and provide prognostic value was congruent across published studies. Further research is warranted to elucidate clinical implications of this association.
Collapse
Affiliation(s)
- Georgios Mavraganis
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Maria-Angeliki Dimopoulou
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Dimitrios Delialis
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Dimitrios Bampatsias
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Raphael Patras
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Alexandros Sianis
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Eleni Maneta
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Georgios Georgiopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece.,Department of Cardiovascular Imaging, School of Biomedical Engineering and Imaging Sciences, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
37
|
Is It All about Endothelial Dysfunction? Focusing on the Alteration in Endothelial Integrity as a Key Determinant of Different Pathological Mechanisms. Biomedicines 2022; 10:biomedicines10112757. [PMID: 36359277 PMCID: PMC9687329 DOI: 10.3390/biomedicines10112757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
|
38
|
Mild-to-Moderate COVID-19 Convalescents May Present Pro-Longed Endothelium Injury. J Clin Med 2022; 11:jcm11216461. [PMID: 36362687 PMCID: PMC9658558 DOI: 10.3390/jcm11216461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Background: The SARS-CoV-2 pandemic posed a great threat to public health, healthcare systems and the economy worldwide. It became clear that, in addition to COVID-19 and acute disease, the condition that develops after recovery may also negatively impact survivors’ health and quality of life. The damage inflicted by the viral infection on endothelial cells was identified quite early on as a possible mechanism underlying the so-called post-COVID syndrome. It became an urgent matter to establish whether convalescents present chronic endothelial impairment, which could result in an increased risk of cardiovascular and thrombotic complications. Methods: In this study, we measured the levels of CRP, ICAM-1, VCAM-1, E-selectin and syndecan-1 as markers of inflammation and endothelial injury in generally healthy convalescents selected from blood donors and compared these to a healthy control group. Results: We found higher concentrations of E-selectin and a lower level of syndecan-1 in convalescents in comparison to those of the control group. Conclusion: Based on our results, it can be concluded that, at least 6 months after infection, there is only slight evidence of endothelial dysfunction in COVID-19 convalescents who do not suffer from other comorbidities related to endothelial impairment.
Collapse
|
39
|
Pelle MC, Zaffina I, Lucà S, Forte V, Trapanese V, Melina M, Giofrè F, Arturi F. Endothelial Dysfunction in COVID-19: Potential Mechanisms and Possible Therapeutic Options. Life (Basel) 2022; 12:1605. [PMID: 36295042 PMCID: PMC9604693 DOI: 10.3390/life12101605] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/29/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
SARS-CoV-2, a novel coronavirus found in Wuhan (China) at the end of 2019, is the etiological agent of the current pandemic that is a heterogeneous disease, named coronavirus disease 2019 (COVID-19). SARS-CoV-2 affects primarily the lungs, but it can induce multi-organ involvement such as acute myocardial injury, myocarditis, thromboembolic eventsandrenal failure. Hypertension, chronic kidney disease, diabetes mellitus and obesity increase the risk of severe complications of COVID-19. There is no certain explanation for this systemic COVID-19 involvement, but it could be related to endothelial dysfunction, due to direct (endothelial cells are infected by the virus) and indirect damage (systemic inflammation) factors. Angiotensin-converting enzyme 2 (ACE2), expressed in human endothelium, has a fundamental role in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In fact, ACE2 is used as a receptor by SARS-CoV-2, leading to the downregulation of these receptors on endothelial cells; once inside, this virus reduces the integrity of endothelial tissue, with exposure of prothrombotic molecules, platelet adhesion, activation of coagulation cascades and, consequently, vascular damage. Systemic microangiopathy and thromboembolism can lead to multi-organ failure with an elevated risk of death. Considering the crucial role of the immunological response and endothelial damage in developing the severe form of COVID-19, in this review, we will attempt to clarify the underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Maria Chiara Pelle
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Isabella Zaffina
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Stefania Lucà
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Valentina Forte
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Vincenzo Trapanese
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Melania Melina
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Federica Giofrè
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| | - Franco Arturi
- Unit of Internal Medicine, Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
- Research Centre for the Prevention and Treatment of Metabolic Diseases (CR METDIS), University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy
| |
Collapse
|
40
|
Marcic M, Marcic L, Lovric Kojundzic S, Marinovic Guic M, Marcic B, Caljkusic K. Chronic Endothelial Dysfunction after COVID-19 Infection Shown by Transcranial Color-Coded Doppler: A Cross-Sectional Study. Biomedicines 2022; 10:2550. [PMID: 36289812 PMCID: PMC9599030 DOI: 10.3390/biomedicines10102550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
In addition to respiratory symptoms, COVID-19 often causes damage to many other organs, especially in severe forms of the disease. Long-term consequences after COVID-19 are common and often have neurological symptoms. Cerebral vasoreactivity may be impaired after acute COVID-19 and in our study, we wanted to show how constant and reversible are the changes in brain vasoreactivity after infection. This cross-sectional observational study included 49 patients diagnosed with COVID-19 and mild neurological symptoms 300 days after the onset of the disease. We used a transcranial color-coded Doppler (TCCD) and a breath-holding test (BHT) to examine cerebral vasoreactivity and brain endothelial function. We analyzed the parameters of the flow rate through the middle cerebral artery (MCA): peak systolic velocity (PSV), end-diastolic velocity (EDV), mean velocity (MV), resistance index (RI) and pulsatility index (PI), and we calculated the breath-holding index (BHI). Subjects after COVID-19 infection had lower measured velocity parameters through MCA at rest period and after BHT, lower relative increases of flow velocities after BHT, and lower BHI. We showed that subjects, 300 days after COVID-19, still have impaired cerebral vasoreactivity measured by TCCD and they have chronic endothelial dysfunction.
Collapse
Affiliation(s)
- Marino Marcic
- Department of Neurology, University Hospital Center Split, Spinciceva 1, 21000 Split, Croatia
| | - Ljiljana Marcic
- Department of Radiology, Polyclinic Medikol, Soltanska 1, 21000 Split, Croatia
- University Department of Health Studies, University of Split, Rudera Boskovica 35, 21000 Split, Croatia
| | - Sanja Lovric Kojundzic
- Department of Radiology, University Hospital Center Split, Spinciceva 1, 21000 Split, Croatia
| | - Maja Marinovic Guic
- University Department of Health Studies, University of Split, Rudera Boskovica 35, 21000 Split, Croatia
- Department of Radiology, University Hospital Center Split, Spinciceva 1, 21000 Split, Croatia
| | - Barbara Marcic
- Department of Medical Genetics, School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Kresimir Caljkusic
- Department of Neurology, University Hospital Center Split, Spinciceva 1, 21000 Split, Croatia
| |
Collapse
|
41
|
Batiha GES, Al-kuraishy HM, Al-Gareeb AI, Welson NN. Pathophysiology of Post-COVID syndromes: a new perspective. Virol J 2022; 19:158. [PMID: 36210445 PMCID: PMC9548310 DOI: 10.1186/s12985-022-01891-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022] Open
Abstract
Most COVID-19 patients recovered with low mortality; however, some patients experienced long-term symptoms described as "long-COVID" or "Post-COVID syndrome" (PCS). Patients may have persisting symptoms for weeks after acute SARS-CoV-2 infection, including dyspnea, fatigue, myalgia, insomnia, cognitive and olfactory disorders. These symptoms may last for months in some patients. PCS may progress in association with the development of mast cell activation syndrome (MCAS), which is a distinct kind of mast cell activation disorder, characterized by hyper-activation of mast cells with inappropriate and excessive release of chemical mediators. COVID-19 survivors, mainly women, and patients with persistent severe fatigue for 10 weeks after recovery with a history of neuropsychiatric disorders are more prone to develop PCS. High D-dimer levels and blood urea nitrogen were observed to be risk factors associated with pulmonary dysfunction in COVID-19 survivors 3 months post-hospital discharge with the development of PCS. PCS has systemic manifestations that resolve with time with no further complications. However, the final outcomes of PCS are chiefly unknown. Persistence of inflammatory reactions, autoimmune mimicry, and reactivation of pathogens together with host microbiome alterations may contribute to the development of PCS. The deregulated release of inflammatory mediators in MCAS produces extraordinary symptoms in patients with PCS. The development of MCAS during the course of SARS-CoV-2 infection is correlated to COVID-19 severity and the development of PCS. Therefore, MCAS is treated by antihistamines, inhibition of synthesis of mediators, inhibition of mediator release, and inhibition of degranulation of mast cells.
Collapse
Affiliation(s)
- Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Al Beheira, 22511 Egypt
| | - Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, ALmustansiriyia University, Baghdad, Iraq
| | - Nermeen N. Welson
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni Suef, 62511 Egypt
| |
Collapse
|
42
|
Haunhorst S, Bloch W, Wagner H, Ellert C, Krüger K, Vilser DC, Finke K, Reuken P, Pletz MW, Stallmach A, Puta C. Long COVID: a narrative review of the clinical aftermaths of COVID-19 with a focus on the putative pathophysiology and aspects of physical activity. OXFORD OPEN IMMUNOLOGY 2022; 3:iqac006. [PMID: 36846561 PMCID: PMC9494493 DOI: 10.1093/oxfimm/iqac006] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/30/2022] [Accepted: 09/13/2022] [Indexed: 11/14/2022] Open
Abstract
The pandemic coronavirus disease 2019 (COVID-19) can cause multi-systemic symptoms that can persist beyond the acute symptomatic phase. The post-acute sequelae of COVID-19 (PASC), also referred to as long COVID, describe the persistence of symptoms and/or long-term complications beyond 4 weeks from the onset of the acute symptoms and are estimated to affect at least 20% of the individuals infected with SARS-CoV-2 regardless of their acute disease severity. The multi-faceted clinical picture of long COVID encompasses a plethora of undulating clinical manifestations impacting various body systems such as fatigue, headache, attention disorder, hair loss and exercise intolerance. The physiological response to exercise testing is characterized by a reduced aerobic capacity, cardiocirculatory limitations, dysfunctional breathing patterns and an impaired ability to extract and use oxygen. Still, to this day, the causative pathophysiological mechanisms of long COVID remain to be elucidated, with long-term organ damage, immune system dysregulation and endotheliopathy being among the hypotheses discussed. Likewise, there is still a paucity of treatment options and evidence-based strategies for the management of the symptoms. In sum, this review explores different aspects of long COVID and maps the literature on what is known about its clinical manifestations, potential pathophysiological mechanisms, and treatment options.
Collapse
Affiliation(s)
- Simon Haunhorst
- Department of Sports Medicine and Health Promotion, Friedrich-Schiller-University Jena, Jena 07749, Germany
- Department of Movement Science, University of Münster, Münster 48149, Germany
| | - Wilhelm Bloch
- Department for Molecular and Cellular Sports Medicine, Institute for Cardiovascular Research and Sports Medicine, German Sport University Cologne, Cologne 50933, Germany
| | - Heiko Wagner
- Department of Movement Science, University of Münster, Münster 48149, Germany
| | - Claudia Ellert
- Department for Vascular Surgery, Lahn-Dill Clinics Wetzlar, Wetzlar 35578, Germany
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Giessen 35394, Germany
| | - Daniel C Vilser
- Hospital for Pediatrics and Adolescent Medicine, Jena University Hospital, Jena 07747, Germany
| | - Kathrin Finke
- Department of Neurology, Jena University Hospital, Jena 07747, Germany
| | - Philipp Reuken
- Clinic for Internal Medicine IV (Gastroenterology, Hepatology and Infectious Diseases), Jena University Hospital, Jena 07747, Germany
| | - Mathias W Pletz
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, Jena 07747, Germany
| | - Andreas Stallmach
- Clinic for Internal Medicine IV (Gastroenterology, Hepatology and Infectious Diseases), Jena University Hospital, Jena 07747, Germany
| | - Christian Puta
- Department of Sports Medicine and Health Promotion, Friedrich-Schiller-University Jena, Jena 07749, Germany
- Center for Interdisciplinary Prevention of Diseases related to Professional Activities, Jena 07749, Germany
| |
Collapse
|
43
|
Kanne JP, Little BP, Schulte JJ, Haramati A, Haramati LB. Long-term Lung Abnormalities Associated with COVID-19 Pneumonia. Radiology 2022; 306:e221806. [PMID: 36040336 PMCID: PMC9462591 DOI: 10.1148/radiol.221806] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
In the 3rd year of the SARS-CoV-2 pandemic, much has been learned about the long-term effects of COVID-19 pneumonia on the lungs. Approximately one-third of patients with moderate-to-severe pneumonia, especially those requiring intensive care therapy or mechanical ventilation, have residual abnormalities at chest CT 1 year after presentation. Abnormalities range from parenchymal bands to bronchial dilation to frank fibrosis. Less is known about the long-term pulmonary vascular sequelae, but there appears to be a persistent, increased risk of venothromboembolic events in a small cohort of patients. Finally, the associated histologic abnormalities resulting from SARS-CoV-2 infection are similar to those seen in patients with other causes of acute lung injury.
Collapse
|
44
|
Ismail AMA. Post-COVID Erectile Dysfunction: The Exercise May Be a Good Considered Complementary Choice. Am J Mens Health 2022; 16:15579883221114983. [PMID: 35927941 PMCID: PMC9358575 DOI: 10.1177/15579883221114983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Ali Mohamed Ali Ismail
- Department of Physical Therapy for Cardiovascular/Respiratory Disorder and Geriatrics, Faculty of Physical Therapy, Cairo University, Giza, Egypt
| |
Collapse
|
45
|
Szewczykowski C, Mardin C, Lucio M, Wallukat G, Hoffmanns J, Schröder T, Raith F, Rogge L, Heltmann F, Moritz M, Beitlich L, Schottenhamml J, Herrmann M, Harrer T, Ganslmayer M, Kruse FE, Kräter M, Guck J, Lämmer R, Zenkel M, Gießl A, Hohberger B. Long COVID: Association of Functional Autoantibodies against G-Protein-Coupled Receptors with an Impaired Retinal Microcirculation. Int J Mol Sci 2022; 23:7209. [PMID: 35806214 PMCID: PMC9266742 DOI: 10.3390/ijms23137209] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 02/05/2023] Open
Abstract
Long COVID (LC) describes the clinical phenotype of symptoms after infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Diagnostic and therapeutic options are limited, as the pathomechanism of LC is elusive. As the number of acute SARS-CoV-2 infections was and is large, LC will be a challenge for the healthcare system. Previous studies revealed an impaired blood flow, the formation of microclots, and autoimmune mechanisms as potential factors in this complex interplay. Since functionally active autoantibodies against G-protein-coupled receptors (GPCR-AAbs) were observed in patients after SARS-CoV-2 infection, this study aimed to correlate the appearance of GPCR-AAbs with capillary microcirculation. The seropositivity of GPCR-AAbs was measured by an established cardiomyocyte bioassay in 42 patients with LC and 6 controls. Retinal microcirculation was measured by OCT-angiography and quantified as macula and peripapillary vessel density (VD) by the Erlangen-Angio Tool. A statistical analysis yielded impaired VD in patients with LC compared to the controls, which was accentuated in female persons. A significant decrease in macula and peripapillary VD for AAbs targeting adrenergic β2-receptor, MAS-receptor angiotensin-II-type-1 receptor, and adrenergic α1-receptor were observed. The present study might suggest that a seropositivity of GPCR-AAbs can be linked to an impaired retinal capillary microcirculation, potentially mirroring the systemic microcirculation with consecutive clinical symptoms.
Collapse
Affiliation(s)
- Charlotte Szewczykowski
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Christian Mardin
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Marianna Lucio
- Research Unit Analytical BioGeoChemistry, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany;
| | | | - Jakob Hoffmanns
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Thora Schröder
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Franziska Raith
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Lennart Rogge
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Felix Heltmann
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Michael Moritz
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Lorenz Beitlich
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Julia Schottenhamml
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Martin Herrmann
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.H.); (T.H.)
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Thomas Harrer
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (M.H.); (T.H.)
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
| | - Marion Ganslmayer
- Department of Internal Medicine 1, Universität of Erlangen-Nürnberg, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Friedrich E. Kruse
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany; (M.K.); (J.G.)
| | - Jochen Guck
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, 91058 Erlangen, Germany; (M.K.); (J.G.)
| | - Robert Lämmer
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Matthias Zenkel
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Andreas Gießl
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| | - Bettina Hohberger
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; (C.S.); (C.M.); (J.H.); (T.S.); (F.R.); (L.R.); (F.H.); (M.M.); (L.B.); (J.S.); (F.E.K.); (R.L.); (M.Z.); (A.G.)
| |
Collapse
|
46
|
Battaglini D, Lopes-Pacheco M, Castro-Faria-Neto HC, Pelosi P, Rocco PRM. Laboratory Biomarkers for Diagnosis and Prognosis in COVID-19. Front Immunol 2022; 13:857573. [PMID: 35572561 PMCID: PMC9091347 DOI: 10.3389/fimmu.2022.857573] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/31/2022] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) causes a wide spectrum of clinical manifestations, with progression to multiorgan failure in the most severe cases. Several biomarkers can be altered in coronavirus disease 2019 (COVID-19), and they can be associated with diagnosis, prognosis, and outcomes. The most used biomarkers in COVID-19 include several proinflammatory cytokines, neuron-specific enolase (NSE), lactate dehydrogenase (LDH), aspartate transaminase (AST), neutrophil count, neutrophils-to-lymphocytes ratio, troponins, creatine kinase (MB), myoglobin, D-dimer, brain natriuretic peptide (BNP), and its N-terminal pro-hormone (NT-proBNP). Some of these biomarkers can be readily used to predict disease severity, hospitalization, intensive care unit (ICU) admission, and mortality, while others, such as metabolomic and proteomic analysis, have not yet translated to clinical practice. This narrative review aims to identify laboratory biomarkers that have shown significant diagnostic and prognostic value for risk stratification in COVID-19 and discuss the possible clinical application of novel analytic strategies, like metabolomics and proteomics. Future research should focus on identifying a limited but essential number of laboratory biomarkers to easily predict prognosis and outcome in severe COVID-19.
Collapse
Affiliation(s)
- Denise Battaglini
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) for Oncology and Neuroscience, Genoa, Italy.,Department of Surgical Science and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy.,Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Paolo Pelosi
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, Instituto di Ricovero e Cura a Carattere Scientifico (IRCCS) for Oncology and Neuroscience, Genoa, Italy.,Department of Surgical Science and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,COVID-19 Virus Network from Brazilian Council for Scientific and Technological Development, Brasília, Brazil.,COVID-19 Virus Network from Foundation Carlos Chagas Filho Research Support of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
47
|
Persistent viral RNA shedding of SARS-CoV-2 is associated with delirium incidence and six-month mortality in hospitalized COVID-19 patients. GeroScience 2022; 44:1241-1254. [PMID: 35538386 PMCID: PMC9090540 DOI: 10.1007/s11357-022-00561-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/17/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Persistent viral RNA shedding of SARS-CoV-2 following COVID-19 has increasingly been recognized, with limited understanding of its implications on outcomes in hospitalized COVID-19 patients. METHODS We retrospectively assessed for persistent viral shedding across Northwestern Medicine Healthcare (NMHC) patients between March and August 2020. We assessed for predictors of persistent viral shedding, in-hospital delirium, and six-month mortality using binary logistic regression. RESULTS Of the 2,518 hospitalized patients with an RT-PCR-confirmed diagnosis of COVID-19, 959 underwent repeat SARS-CoV-2 RT-PCR at least fourteen days from initial positive testing. Of those, 405 (42.2%) patients were found to have persistent viral shedding. Persistent viral shedding was associated with male sex, increased BMI, diabetes mellitus, chronic kidney disease, and exposure to corticosteroids during initial COVID-19 hospitalization. Persistent viral shedding was independently associated with incidence of in-hospital delirium after adjusting for factors including severity of respiratory dysfunction (OR 2.45; 95% CI 1.75, 3.45). Even after adjusting for age, severity of respiratory dysfunction, and occurrence of in-hospital delirium, persistent viral shedding remained significantly associated with increased six-month mortality (OR 2.43; 95% CI 1.42, 4.29). CONCLUSIONS Persistent viral shedding occurs frequently in hospitalized COVID-19 patients and is associated with in-hospital delirium and increased six-month mortality.
Collapse
|
48
|
Long-term cardio-vascular risk assessment in chronic kidney disease and kidney transplanted patients following SARS-COV-2 disease: protocol for multi-center observational match controlled trial. BMC Nephrol 2022; 23:176. [PMID: 35524223 PMCID: PMC9077937 DOI: 10.1186/s12882-022-02809-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The coronavirus disease (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) produced a pandemic since March 2020 by affecting more than 243 million people with more than 5 million deaths globally. SARS-CoV-2 infection is produced by binding to angiotensin-converting enzyme, which among other sites is highly expressed in the endothelial cells of the blood vessels, pericytes and the heart, as well as in renal podocytes and proximal tubular epithelial cells. SARS-CoV-2 and cardiovascular disease (CVD) are interconnected by risk factors association with an increased incidence of the disease and by determining de novo cardiac complications. At the same time, COVID-19 disease can lead to acute kidney injury directly, or due to sepsis, multi-organ failure and shock. Therefore, the pre-existence of both CVD and chronic kidney disease (CKD) is linked with a higher risk of severe disease and worse prognosis. METHODS The main aim of this study is to assess the CV risk in a CKD (stage 3 to 5), dialysis and kidney transplanted population, following SARS-CoV-2 infection, with focus on the endothelial dysfunction as compared to a control group of matched patients. By using clinical evaluation, flow-mediated dilatation, carotid-femoral pulse wave velocity, intima-media thickness, echocardiographic parameters, lung ultrasound, bioimpedance spectroscopy and a series of novel biomarkers, the investigators will determine the long-term impact of this disease on CV and renal outcomes. DISCUSSION This study will address the challenges and implications in long-term CV sequeale of COVID-19 and focus on a better understanding of the underlying mechanisms and possible therapeutic options. TRIAL REGISTRATION Patient enrolment in the trial started in January 2021 and is expected to finish at the end of 2022. The study can be found on ClinicalTrials.gov database with NCT05125913 identifier. Registered on 18 November 2021 - Retrospectively registered.
Collapse
|
49
|
Wang C, Yu C, Jing H, Wu X, Novakovic VA, Xie R, Shi J. Long COVID: The Nature of Thrombotic Sequelae Determines the Necessity of Early Anticoagulation. Front Cell Infect Microbiol 2022; 12:861703. [PMID: 35449732 PMCID: PMC9016198 DOI: 10.3389/fcimb.2022.861703] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/15/2022] [Indexed: 12/24/2022] Open
Abstract
Many discharged COVID-19 patients affected by sequelae experience reduced quality of life leading to an increased burden on the healthcare system, their families and society at large. Possible pathophysiological mechanisms of long COVID include: persistent viral replication, chronic hypoxia and inflammation. Ongoing vascular endothelial damage promotes platelet adhesion and coagulation, resulting in the impairment of various organ functions. Meanwhile, thrombosis will further aggravate vasculitis contributing to further deterioration. Thus, long COVID is essentially a thrombotic sequela. Unfortunately, there is currently no effective treatment for long COVID. This article summarizes the evidence for coagulation abnormalities in long COVID, with a focus on the pathophysiological mechanisms of thrombosis. Extracellular vesicles (EVs) released by various types of cells can carry SARS-CoV-2 through the circulation and attack distant tissues and organs. Furthermore, EVs express tissue factor and phosphatidylserine (PS) which aggravate thrombosis. Given the persistence of the virus, chronic inflammation and endothelial damage are inevitable. Pulmonary structural changes such as hypertension, embolism and fibrosis are common in long COVID. The resulting impaired lung function and chronic hypoxia again aggravates vascular inflammation and coagulation abnormalities. In this article, we also summarize recent research on antithrombotic therapy in COVID-19. There is increasing evidence that early anticoagulation can be effective in improving outcomes. In fact, persistent systemic vascular inflammation and dysfunction caused by thrombosis are key factors driving various complications of long COVID. Early prophylactic anticoagulation can prevent the release of or remove procoagulant substances, thereby protecting the vascular endothelium from damage, reducing thrombotic sequelae, and improving quality of life for long-COVID patients.
Collapse
Affiliation(s)
- Chengyue Wang
- Department of Hematology, The First Hospital of Harbin, Harbin Medical University, Harbin, China
- Department of Nephrology, The First Hospital of Harbin, Harbin Medical University, Harbin, China
| | - Chengyuan Yu
- Department of Hematology, The First Hospital of Harbin, Harbin Medical University, Harbin, China
- Department of Geriatric, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Haijiao Jing
- Department of Hematology, The First Hospital of Harbin, Harbin Medical University, Harbin, China
| | - Xiaoming Wu
- Department of Hematology, The First Hospital of Harbin, Harbin Medical University, Harbin, China
| | - Valerie A. Novakovic
- Department of Research, Veterans Affairs (VA) Boston Healthcare System, Harvard Medical School, Boston, MA, United States
| | - Rujuan Xie
- Department of Nephrology, The First Hospital of Harbin, Harbin Medical University, Harbin, China
- *Correspondence: Rujuan Xie, ; Jialan Shi,
| | - Jialan Shi
- Department of Hematology, The First Hospital of Harbin, Harbin Medical University, Harbin, China
- Department of Research, Veterans Affairs (VA) Boston Healthcare System, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
- *Correspondence: Rujuan Xie, ; Jialan Shi,
| |
Collapse
|
50
|
Ambrosino P, Calcaterra IL, Mosella M, Formisano R, D’Anna SE, Bachetti T, Marcuccio G, Galloway B, Mancini FP, Papa A, Motta A, Di Minno MND, Maniscalco M. Endothelial Dysfunction in COVID-19: A Unifying Mechanism and a Potential Therapeutic Target. Biomedicines 2022; 10:biomedicines10040812. [PMID: 35453563 PMCID: PMC9029464 DOI: 10.3390/biomedicines10040812] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 02/07/2023] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) generated a worldwide emergency, until the declaration of the pandemic in March 2020. SARS-CoV-2 could be responsible for coronavirus disease 2019 (COVID-19), which goes from a flu-like illness to a potentially fatal condition that needs intensive care. Furthermore, the persistence of functional disability and long-term cardiovascular sequelae in COVID-19 survivors suggests that convalescent patients may suffer from post-acute COVID-19 syndrome, requiring long-term care and personalized rehabilitation. However, the pathophysiology of acute and post-acute manifestations of COVID-19 is still under study, as a better comprehension of these mechanisms would ensure more effective personalized therapies. To date, mounting evidence suggests a crucial endothelial contribution to the clinical manifestations of COVID-19, as endothelial cells appear to be a direct or indirect preferential target of the virus. Thus, the dysregulation of many of the homeostatic pathways of the endothelium has emerged as a hallmark of severity in COVID-19. The aim of this review is to summarize the pathophysiology of endothelial dysfunction in COVID-19, with a focus on personalized pharmacological and rehabilitation strategies targeting endothelial dysfunction as an attractive therapeutic option in this clinical setting.
Collapse
Affiliation(s)
- Pasquale Ambrosino
- Istituti Clinici Scientifici Maugeri IRCCS, Cardiac Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy; (R.F.); (F.P.M.); (A.P.)
- Correspondence: (P.A.); (M.M.)
| | | | - Marco Mosella
- Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy; (M.M.); (S.E.D.)
| | - Roberto Formisano
- Istituti Clinici Scientifici Maugeri IRCCS, Cardiac Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy; (R.F.); (F.P.M.); (A.P.)
| | - Silvestro Ennio D’Anna
- Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy; (M.M.); (S.E.D.)
| | - Tiziana Bachetti
- Istituti Clinici Scientifici Maugeri IRCCS, Scientific Direction, 27100 Pavia, Italy;
| | - Giuseppina Marcuccio
- Università della Campania Luigi Vanvitelli, 81100 Caserta, Italy; (G.M.); (B.G.)
| | - Brurya Galloway
- Università della Campania Luigi Vanvitelli, 81100 Caserta, Italy; (G.M.); (B.G.)
| | - Francesco Paolo Mancini
- Istituti Clinici Scientifici Maugeri IRCCS, Cardiac Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy; (R.F.); (F.P.M.); (A.P.)
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| | - Antimo Papa
- Istituti Clinici Scientifici Maugeri IRCCS, Cardiac Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy; (R.F.); (F.P.M.); (A.P.)
| | - Andrea Motta
- Institute of Biomolecular Chemistry, National Research Council, 80078 Pozzuoli, Italy;
| | | | - Mauro Maniscalco
- Istituti Clinici Scientifici Maugeri IRCCS, Pulmonary Rehabilitation Unit of Telese Terme Institute, 82037 Telese Terme, Italy; (M.M.); (S.E.D.)
- Correspondence: (P.A.); (M.M.)
| |
Collapse
|