1
|
van der Knaap N, Klinkhammer S, Postma AA, Visser-Meily JMA, Horn J, van Heugten CM, Voorter PHM, van der Thiel MM, Drenthen GS, Backes WH, van Rosmalen F, van Santen S, van Bussel BCT, van der Horst ICC, Linden DEJ, Ariës MJH, Jansen JFA. Post-COVID microvascular dysfunction in hospitalized COVID-19 survivors is associated with acute disease severity and persistent cognitive complaints. J Neurol Sci 2025; 472:123464. [PMID: 40088612 DOI: 10.1016/j.jns.2025.123464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/05/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
OBJECTIVE Coronavirus disease 2019 (COVID-19) is known to have adverse effects on the brain's vasculature in some patients. After recovery of the infection, vascular abnormalities may persist, but it remains unclear which pathological pathways play a role in post-COVID vascular and cognitive dysfunction and may contribute to post-COVID cognitive complaints. METHODS In this observational cohort study, 108 previously hospitalized COVID-19 survivors (general ward: n = 53; intensive care unit (ICU): n = 55) were compared. Cerebral microvascular properties in the cortical gray matter (cGM), normal-appearing white matter (NAWM), and white matter hyperintensities (WMH) were assessed using multi-b-value diffusion MRI around 9 months post-infection and related to acute systemic blood markers and post-COVID cognitive performance and complaints. RESULTS A lower microvascular perfusion volume fraction (fmv) and blood flow-related measure (fmv·Dmv) were found in ICU compared to general ward patients in the cGM (p = .032; p = .021), NAWM (p = .008; p = .006), and WMH (p = .014; p = .035). No associations were found between diffusion/perfusion metrics and cognitive performance, but a lower fmv in the NAWM was found in those with more cognitive complaints (p = .047). In ICU survivors, higher median blood lactate levels during ICU admission were associated with lower fmv (p = .031) and fmv·Dmv (p = .044) in the NAWM. INTERPRETATION Significantly more widespread post-COVID cerebral microvascular dysfunction was found in COVID-19 ICU compared to general ward survivors. Post-COVID microvascular dysfunction in the NAWM may be due to more severe cerebral tissue hypoxia at time of the infection and is associated with persisting subjective cognitive complaints, even in absence of objective cognitive problems.
Collapse
Affiliation(s)
- Noa van der Knaap
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, the Netherlands; Department of Intensive Care, Maastricht University Medical Center, the Netherlands; Mental Health & Neuroscience Research Institute (MHeNs), Maastricht University, the Netherlands
| | - Simona Klinkhammer
- Mental Health & Neuroscience Research Institute (MHeNs), Maastricht University, the Netherlands; Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, the Netherlands; Limburg Brain Injury Center, Maastricht University, the Netherlands
| | - Alida A Postma
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, the Netherlands; Mental Health & Neuroscience Research Institute (MHeNs), Maastricht University, the Netherlands
| | - Johanna M A Visser-Meily
- Department of Rehabilitation, Physical Therapy Science and Sports, University Medical Center Utrecht, the Netherlands; UMC Utrecht Brain Center, UMC Utrecht, the Netherlands
| | - Janneke Horn
- Deparment of Intensive Care, University Medical Center Amsterdam, the Netherlands; Amsterdam Neuroscience, University Medical Center Amsterdam, the Netherlands
| | - Caroline M van Heugten
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, the Netherlands; Limburg Brain Injury Center, Maastricht University, the Netherlands; Department of Neuropsychology and Psychopharmacology, Maastricht University, the Netherlands
| | - Paulien H M Voorter
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, the Netherlands; Mental Health & Neuroscience Research Institute (MHeNs), Maastricht University, the Netherlands
| | - Merel M van der Thiel
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, the Netherlands; Mental Health & Neuroscience Research Institute (MHeNs), Maastricht University, the Netherlands
| | - Gerhard S Drenthen
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, the Netherlands; Mental Health & Neuroscience Research Institute (MHeNs), Maastricht University, the Netherlands
| | - Walter H Backes
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, the Netherlands; Mental Health & Neuroscience Research Institute (MHeNs), Maastricht University, the Netherlands; Cardiovascular Disease Research Institute (CARIM), Maastricht University, the Netherlands
| | - Frank van Rosmalen
- Department of Intensive Care, Maastricht University Medical Center, the Netherlands; Cardiovascular Disease Research Institute (CARIM), Maastricht University, the Netherlands
| | - Susanne van Santen
- Department of Intensive Care, Maastricht University Medical Center, the Netherlands
| | - Bas C T van Bussel
- Department of Intensive Care, Maastricht University Medical Center, the Netherlands; Cardiovascular Disease Research Institute (CARIM), Maastricht University, the Netherlands; Care and Public Health Research Institute (CAPHRI), Maastricht University, the Netherlands
| | - Iwan C C van der Horst
- Department of Intensive Care, Maastricht University Medical Center, the Netherlands; Cardiovascular Disease Research Institute (CARIM), Maastricht University, the Netherlands
| | - David E J Linden
- Mental Health & Neuroscience Research Institute (MHeNs), Maastricht University, the Netherlands
| | - Marcel J H Ariës
- Department of Intensive Care, Maastricht University Medical Center, the Netherlands; Mental Health & Neuroscience Research Institute (MHeNs), Maastricht University, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, the Netherlands; Mental Health & Neuroscience Research Institute (MHeNs), Maastricht University, the Netherlands; Department of Electrical Engineering, Eindhoven University of Technology, the Netherlands.
| |
Collapse
|
2
|
Haddox S, Wu P, Singh S, Qin F, Engel J, Kian A, Ahmad S, Li H, Wu P. Landscape of chimeric RNAs in COVID-19 patient blood. Genes Dis 2025; 12:101348. [PMID: 40110491 PMCID: PMC11919593 DOI: 10.1016/j.gendis.2024.101348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/12/2024] [Accepted: 04/24/2024] [Indexed: 03/22/2025] Open
Abstract
Despite the availability of efficacious vaccines, COVID-19 persists and our knowledge of how SARS-CoV-2 infection affects host transcriptomics remains incomplete. Transcriptome analysis, which has progressed our understanding of the patient response to SARS-CoV-2 infection, can be enhanced by considering chimeric transcript expression. Here we assess and characterize chimeric RNAs found in the whole blood of 178 COVID-19 patients. STAR-Fusion, SOAPfuse, and EricScript were used to detect chimeric RNAs resulting in over 30,000 predictions with approximately 500 high-confidence predictions that were found by more than one software and filtered based on exon annotations around the chimeric splice junction. GO term enrichment performed on the 5' and 3' parental genes of chimeric RNAs found in severe and critical patients resulted in pathways known to be affected in these patients, such as erythroid differentiation. Motif enrichment analysis of sequences proximal to chimeric splice junctions found in COVID-19 patients versus those found in GTEx whole blood revealed two RNA binding proteins previously implicated with coronavirus infection, PTBP1 and SFPQ. We discovered a chimeric RNA that correlated with COVID-19 disease status and appeared to be dependent upon a loss of PTBP1's function as a splicing repressor. Overall, we found over 350 novel COVID-19-specific chimeric RNAs not detectable in GTEx whole blood that may also serve as biomarkers for viral infection.
Collapse
Affiliation(s)
- Samuel Haddox
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Ping Wu
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- National Clinical Research Center for Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Sandeep Singh
- Computational Toxicology Facility, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh 226001, India
| | - Fujun Qin
- School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Jack Engel
- Department of Pathology, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Andrea Kian
- Department of Pathology, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Syed Ahmad
- Department of Pathology, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Hui Li
- Department of Biochemistry and Molecular Genetics, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Department of Pathology, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Peng Wu
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- National Clinical Research Center for Gynecology and Obstetrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
3
|
de Melo BP, da Silva JAM, Rodrigues MA, Palmeira JDF, Saldanha-Araujo F, Argañaraz GA, Argañaraz ER. SARS-CoV-2 Spike Protein and Long COVID-Part 1: Impact of Spike Protein in Pathophysiological Mechanisms of Long COVID Syndrome. Viruses 2025; 17:617. [PMID: 40431629 PMCID: PMC12115690 DOI: 10.3390/v17050617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/06/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
SARS-CoV-2 infection has resulted in more than 700 million cases and nearly 7 million deaths worldwide. Although vaccination efforts have effectively reduced mortality and transmission rates, a significant proportion of recovered patients-up to 40%-develop long COVID syndrome (LC) or post-acute sequelae of COVID-19 infection (PASC). LC is characterized by the persistence or emergence of new symptoms following initial SARS-CoV-2 infection, affecting the cardiovascular, neurological, respiratory, gastrointestinal, reproductive, and immune systems. Despite the broad range of clinical symptoms that have been described, the risk factors and pathogenic mechanisms behind LC remain unclear. This review, the first of a two-part series, is distinguished by the discussion of the role of the SARS-CoV-2 spike protein in the primary mechanisms underlying the pathophysiology of LC.
Collapse
Affiliation(s)
- Bruno Pereira de Melo
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasilia 70910-900, DF, Brazil
| | - Jhéssica Adriane Mello da Silva
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasilia 70910-900, DF, Brazil
| | - Mariana Alves Rodrigues
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasilia 70910-900, DF, Brazil
| | - Julys da Fonseca Palmeira
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasilia 70910-900, DF, Brazil
| | - Felipe Saldanha-Araujo
- Laboratory of Hematology and Stem Cells (LHCT), Faculty of Health Sciences, University of Brasília, Brasilia 70910-900, DF, Brazil
| | - Gustavo Adolfo Argañaraz
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasilia 70910-900, DF, Brazil
| | - Enrique Roberto Argañaraz
- Laboratory of Molecular Neurovirology, Department of Pharmacy, Faculty of Health Science, University of Brasília, Brasilia 70910-900, DF, Brazil
| |
Collapse
|
4
|
Erdik B. Driving under viral impairment: Linking acute SARS-CoV-2 infections to elevated car crash risks. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0004420. [PMID: 40198595 PMCID: PMC11978055 DOI: 10.1371/journal.pgph.0004420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/20/2025] [Indexed: 04/10/2025]
Abstract
This study explores the linkage between acute SARS-CoV-2 and car crashes across U.S. states, correlating with COVID-19 mitigation strategies, vaccination rates, and Long COVID prevalence. This investigation analyzed aggregate COVID-19 and car crash data spanning 2020-2023, with data collection occurring between March and May 2024. Analysis was done via a Poisson regression model, adjusted for population. Key variables included vaccination status, month-specific effects relating to initial pandemic shutdowns, and Long COVID rates. Results demonstrated a significant association between acute COVID-19 infections and an increase in car crashes, independent of Long COVID status to the tune of an OR of 1.25 [1.23-1.26]. This association was observed despite varying mitigation efforts and vaccination rates across states. The study found no protective effect of vaccination against car crashes, challenging prior assumptions about the benefits of vaccination. Notably, the risk associated with COVID-19 was found to be analogous to driving impairments seen with alcohol consumption at legal limits. Findings suggest significant implications for public health policies, especially in assessing the readiness of individuals recovering from COVID-19 to engage in high-risk activities such as pilots or nuclear plant employees. Further research is necessary to establish causation and explore the exact effects of COVID-19 within the CNS affecting cognition and behavior.
Collapse
Affiliation(s)
- Baran Erdik
- Department of Healthcare Administration, American Vision University, Anaheim, California, United States of America
- Hygia Health, Miami, Florida, United States of America
| |
Collapse
|
5
|
Liu Y, Wang N, Ru Q, Fan K, Sun N, Sun P, Li H, Yin W. Assay of cardiopulmonary bypass system for porcine alveolar macrophages removing GFP- E. coli from erythrocyte surfaces. PeerJ 2025; 13:e18934. [PMID: 40061231 PMCID: PMC11887565 DOI: 10.7717/peerj.18934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/14/2025] [Indexed: 05/13/2025] Open
Abstract
While it is established that complement receptor molecules on the surface of erythrocytes are crucial for the clearance of immune complexes in the body, the molecular mechanisms underlying the interaction between macrophages and erythrocytes in pigs remain inadequately understood. Consequently, we built a detection system with a closed-circulation flow chamber and a constant flow pump. Additionally, we optimized parameters including system flow velocity and fluid shear force. In the circulatory system, our study measured the fluorescence intensity of erythrocyte and pulmonary alveolar macrophages (PAMs) surfaces before and after the blockade of complement receptor 1 (CR1)-like receptors and Fc receptors. The results indicated that porcine erythrocytes and PAMs exhibited a diminished rate of change in fluorescence intensity under the blocked condition. Through transmission electron microscopy, it was observed that PAMs effectively removed sensitized GFP-E. coli adhering immunologically to porcine erythrocytes. The findings indicate that PAMs effectively removed sensitized GFP-E. coli from the surface immunoadhesion of porcine erythrocytes, facilitated by the mediation of surface CR1-like receptors and Fc receptors.
Collapse
Affiliation(s)
- Yongqiang Liu
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong City, Shanxi Province, China
| | - Nan Wang
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong City, Shanxi Province, China
| | - Qing Ru
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong City, Shanxi Province, China
| | - Kuohai Fan
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong City, Shanxi Province, China
| | - Na Sun
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong City, Shanxi Province, China
| | - Panpan Sun
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong City, Shanxi Province, China
| | - Hongquan Li
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong City, Shanxi Province, China
| | - Wei Yin
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong City, Shanxi Province, China
| |
Collapse
|
6
|
Gáspár Z, Szabó BG, Ceglédi A, Lakatos B. Human herpesvirus reactivation and its potential role in the pathogenesis of post-acute sequelae of SARS-CoV-2 infection. GeroScience 2025; 47:167-187. [PMID: 39207648 PMCID: PMC11872864 DOI: 10.1007/s11357-024-01323-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
The emergence of SARS-CoV-2 has precipitated a global pandemic with substantial long-term health implications, including the condition known as post-acute sequelae of SARS-CoV-2 infection (PASC), commonly referred to as Long COVID. PASC is marked by persistent symptoms such as fatigue, neurological issues, and autonomic dysfunction that persist for months beyond the acute phase of COVID-19. This review examines the potential role of herpesvirus reactivation, specifically Epstein-Barr virus (EBV) and cytomegalovirus (CMV), in the pathogenesis of PASC. Elevated antibody titers and specific T cell responses suggest recent herpesvirus reactivation in some PASC patients, although viremia is not consistently detected. SARS-CoV-2 exhibits endothelial trophism, directly affecting the vascular endothelium and contributing to microvascular pathologies. These pathologies are significant in PASC, where microvascular dysfunction may underlie various chronic symptoms. Similarly, herpesviruses like CMV also exhibit endothelial trophism, which may exacerbate endothelial damage when reactivated. Evidence suggests that EBV and CMV reactivation could indirectly contribute to the immune dysregulation, immunosenescence, and autoimmune responses observed in PASC. Additionally, EBV may play a role in the genesis of neurological symptoms through creating mitochondrial dysfunction, though direct confirmation remains elusive. The reviewed evidence suggests that while herpesviruses may not play a direct role in the pathogenesis of PASC, their potential indirect effects, especially in the context of endothelial involvement, warrant further investigation.
Collapse
Affiliation(s)
- Zsófia Gáspár
- School of PhD Studies, Semmelweis University, Üllői Street 26, 1085, Budapest, Hungary
- South Pest Central Hospital, National Institute of Haematology and Infectious Diseases, Albert Flórián Street 5-7, 1097, Budapest, Hungary
| | - Bálint Gergely Szabó
- School of PhD Studies, Semmelweis University, Üllői Street 26, 1085, Budapest, Hungary.
- South Pest Central Hospital, National Institute of Haematology and Infectious Diseases, Albert Flórián Street 5-7, 1097, Budapest, Hungary.
- Departmental Group of Infectious Diseases, Department of Internal Medicine and Haematology, Semmelweis University, Albert Flórián Street 5-7, 1097, Budapest, Hungary.
| | - Andrea Ceglédi
- South Pest Central Hospital, National Institute of Haematology and Infectious Diseases, Albert Flórián Street 5-7, 1097, Budapest, Hungary
| | - Botond Lakatos
- School of PhD Studies, Semmelweis University, Üllői Street 26, 1085, Budapest, Hungary
- South Pest Central Hospital, National Institute of Haematology and Infectious Diseases, Albert Flórián Street 5-7, 1097, Budapest, Hungary
- Departmental Group of Infectious Diseases, Department of Internal Medicine and Haematology, Semmelweis University, Albert Flórián Street 5-7, 1097, Budapest, Hungary
| |
Collapse
|
7
|
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
|
8
|
Mielcarska MB, Rouse BT. Viruses and the Brain-A Relationship Prone to Trouble. Viruses 2025; 17:203. [PMID: 40006958 PMCID: PMC11860391 DOI: 10.3390/v17020203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Neurological disorders, some of which are associated with viral infections, are growing due to the aging and expanding population. Despite strong defenses of the central nervous system, some viruses have evolved ways to breach them, which often result in dire consequences. In this review, we recount the various ways by which different viruses can enter the CNS, and we describe the consequences of such invasions. Consequences may manifest as acute disease, such as encephalitis, meningitis, or result in long-term effects, such as neuromuscular dysfunction, as occurs in poliomyelitis. We discuss evidence for viral involvement in the causation of well-known chronic neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, as well as vascular dementia in the elderly. We also describe the approaches currently available to control a few of the neural viral infections. These include antivirals that are effective against human immunodeficiency virus and herpes simplex virus, as well as vaccines valuable for controlling rabies virus, poliomyelitis virus, and some flavivirus infections. There is an urgent need to better understand, at a molecular level, how viruses contribute to acute and, especially, chronic neurological diseases and to develop more precise and effective vaccines and therapies.
Collapse
Affiliation(s)
- Matylda Barbara Mielcarska
- Department of Preclinical Sciences, Institute of Veterinary Sciences, Warsaw University of Life Sciences–SGGW, Jana Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Barry T. Rouse
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
9
|
Chang CC, Wang YH, Yen JC, Liaw CC, Tsai KC, Wei WC, Chiou WF, Chiou CT, Liou KT, Shen YC, Su YC. NRICM101 in combatting COVID-19 induced brain fog: Neuroprotective effects and neurovascular integrity preservation in hACE2 mice. J Tradit Complement Med 2025; 15:36-50. [PMID: 39807266 PMCID: PMC11725119 DOI: 10.1016/j.jtcme.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 01/16/2025] Open
Abstract
Amidst growing concerns over COVID-19 aftereffects like fatigue and cognitive issues, NRICM101, a traditional Chinese medicine, has shown promise. Used by over 2 million people globally, it notably reduces hospitalizations and intubations in COVID-19 patients. To explore whether NRICM101 could combat COVID-19 brain fog, we tested NRICM101 on hACE2 transgenic mice administered the S1 protein of SARS-CoV-2, aiming to mitigate S1-induced cognitive issues by measuring animal behaviors, immunohistochemistry (IHC) staining, and next-generation sequencing (NGS) analysis. The study revealed that S1 protein-administered mice displayed marked signs of brain fog, characterized by reduced learning, memory, and nesting abilities. However, NRICM101 treatment in these animals ameliorated all these cognitive functions. S1 protein administration in mice induced notable inflammation, leading to the death of neurons (NeuN+) and neural stem cells (DCX+) in hACE2 transgenic mice. This was accompanied by heightened microglia activation (IBA1+/CD68+), increased cytokine production (IL1β, IL6), induction of neutrophil extracellular traps (NET), inflammation (NLRP3, CD11b), and platelet (CD31, vWF) and complement (C3) activation, ultimately damaging neurovasculature and disrupting the blood-brain barrier (B.B.B.). Administration of NRICM101 effectively alleviated all these pathological changes. In conclusion, NRICM101 has the potential to prevent COVID-19-associated brain fog by bolstering neurovascular integrity and protecting neurons and neural stem cells. This is achieved by the inhibition of S1 protein-induced complement activation, which in turn leads to the prevention of damage to the neurovasculature and the subsequent death of neurons.
Collapse
Affiliation(s)
- Cher-Chia Chang
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei City, 112304, Taiwan
| | - Yea-Hwey Wang
- National Taipei University of Nursing and Health Science, Taipei City, 112303, Taiwan
| | - Jiin-Cherng Yen
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei City, 112304, Taiwan
| | - Chia-Ching Liaw
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, 112026, Taiwan
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807378, Taiwan
| | - Keng-Chang Tsai
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, 112026, Taiwan
- Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei City, 110301, Taiwan
| | - Wen-Chi Wei
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, 112026, Taiwan
| | - Wen-Fei Chiou
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, 112026, Taiwan
| | - Chun-Tang Chiou
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, 112026, Taiwan
| | - Kuo-Tong Liou
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, 112026, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, 25245, Taiwan
- Department of Chinese Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei City, 114202, Taiwan
| | - Yuh-Chiang Shen
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, 112026, Taiwan
- National Taipei University of Nursing and Health Science, Taipei City, 112303, Taiwan
| | - Yi-Chang Su
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei City, 112026, Taiwan
| |
Collapse
|
10
|
Zhang-James Y, Clay JWS, Aber RB, Gamble HM, Faraone SV. Post-COVID-19 Mental Health Distress in 13 Million Youth: A Retrospective Cohort Study of Electronic Health Records. J Am Acad Child Adolesc Psychiatry 2025; 64:65-76. [PMID: 38815620 DOI: 10.1016/j.jaac.2024.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 03/24/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVE To investigate the impact of the SARS-CoV-2 infection on the rates of mental disorders in youth. METHOD The study involved 7,519,465 children and 5,338,496 adolescents from the TriNetX Research Network, all without prior mental disorder histories. Among them, 290,145 children and 223,667 adolescents had SARS-CoV-2-positive tests or confirmed COVID-19 diagnoses. Kaplan-Meier survival analysis was used to evaluate the probability of developing new mental disorders (any codes in International Classification of Diseases, Tenth Revision, Clinical Modification (ICD-10-CM) F01-F99 category and suicidal behaviors) within 2 years post infection, compared to the propensity score-matched youth who were never infected. RESULTS Within 2 years post SARS-CoV-2 infection, children had a probability of 0.15 in acquiring new psychiatric diagnoses, compared to 0.026 for matched non-infected children; adolescents had a 0.19 probability against 0.05 for their non-infected counterparts. The hazard ratio (HR) was 6.0 (95% CI = 5.8-6.3) for children and 4.2 for adolescents (95% CI = 4.1-4.4), with children vs adolescents HR of 1.4 (95% CI = 1.36-1.51). Elevated HRs were observed for almost all subcategories of mental disorders and suicidal behaviors, with variations based on sex, severity of SARS-CoV-2 infection, and viral variants. COVID-19 was similar to other respiratory infections and was associated with a similarly increased rate of mental disorders in adolescents, but had a significantly higher effect on children (HR = 1.57, 95% CI =1.53-1.61). CONCLUSION This study revealed significant mental health distress following SARS-CoV-2 infection in youth, which was more pronounced in children than in adolescents. These findings underscore the urgent need to support at-risk youth, particularly those who contracted SARS-CoV-2 at younger ages and had more severe infections. PLAIN LANGUAGE SUMMARY The authors of this study conducted analyses using electronic medical records from the TriNetX Research Network to investigate the impact of SARS-CoV-2 infection on the rates of mental health disorders in youth. Within 2 years after SARS-CoV-2 infection, children and adolescents were 6.0 and 4.2 times more likely to acquire a new psychiatric diagnosis, respectively. While the risk of being diagnosed with a mental health disorder following SARS-CoV-2 infection in adolescents was comparable to other respiratory infections, it had a more pronounced effect on children.
Collapse
Affiliation(s)
- Yanli Zhang-James
- Norton College of Medicine at SUNY Upstate Medical University, Syracuse, New York.
| | - John W S Clay
- Norton College of Medicine at SUNY Upstate Medical University, Syracuse, New York
| | - Rachel B Aber
- Norton College of Medicine at SUNY Upstate Medical University, Syracuse, New York
| | - Hilary M Gamble
- Norton College of Medicine at SUNY Upstate Medical University, Syracuse, New York
| | - Stephen V Faraone
- Norton College of Medicine at SUNY Upstate Medical University, Syracuse, New York
| |
Collapse
|
11
|
Morse BA, Motovilov K, Michael Brode W, Michael Tee F, Melamed E. A review of intravenous immunoglobulin in the treatment of neuroimmune conditions, acute COVID-19 infection, and post-acute sequelae of COVID-19 Syndrome. Brain Behav Immun 2025; 123:725-738. [PMID: 39389388 DOI: 10.1016/j.bbi.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/12/2024] [Accepted: 10/05/2024] [Indexed: 10/12/2024] Open
Abstract
Intravenous immunoglobulin (IVIG) is an immunomodulatory therapy that has been studied in several neuroimmune conditions, such as Guillain-Barré Syndrome, chronic inflammatory demyelinating polyneuropathy, multifocal motor neuropathy, and multiple sclerosis. It has also been proposed as a potential treatment option for acute COVID-19 infection and post-acute sequelae of SARS-CoV-2 infection (PASC). IVIG is thought to function by providing the recipient with a pool of antibodies, which can, in turn, modulate immune responses through multiple mechanisms including neutralization of cytokines and autoantibodies, saturation of neonatal fragment crystallizable receptors, inhibition of complement activation, and regulation of T and B cell mediated inflammation. In acute COVID-19, studies have shown that early administration of IVIG and plasmapheresis in severe cases can reduce the need for mechanical ventilation, shorten ICU and hospital stays, and lower mortality. Similarly, in PASC, while research is still in early stages, IVIG has been shown to alleviate persistent symptoms in small patient cohorts. Furthermore, IVIG has shown benefits in another condition which has symptomatic overlap with PASC, myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), though studies have yielded mixed results. It is important to note that IVIG can be associated with several potential adverse effects, such as anaphylaxis, headaches, thrombosis, liver enzyme elevations and renal complications. In addition, the high cost of IVIG can be a deterrent for payers and patients. This review provides a comprehensive update on the use of IVIG in multiple neuroimmune conditions, ME/CFS, acute COVID-19, and PASC, as well as covers its history, production, pricing, and mechanisms of action. We also identify key areas of future research, including the need to optimize the use of Ig product dosing, timing, and patient selection across conditions, particularly in the context of COVID-19 and PASC.
Collapse
Affiliation(s)
- Brinkley A Morse
- Department of Neurology, Dell Medical School at the University of Texas, Austin, USA
| | - Katherine Motovilov
- Department of Neurology, Dell Medical School at the University of Texas, Austin, USA
| | - W Michael Brode
- Department of Internal Medicine, Dell Medical School at the University of Texas, Austin, USA
| | - Francis Michael Tee
- Department of Internal Medicine, Dell Medical School at the University of Texas, Austin, USA.
| | - Esther Melamed
- Department of Neurology, Dell Medical School at the University of Texas, Austin, USA.
| |
Collapse
|
12
|
Bremner JD, Russo SJ, Gallagher R, Simon NM. Acute and long-term effects of COVID-19 on brain and mental health: A narrative review. Brain Behav Immun 2025; 123:928-945. [PMID: 39500417 PMCID: PMC11974614 DOI: 10.1016/j.bbi.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/16/2024] [Accepted: 11/02/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND COVID infection has been associated with long term sequalae (Long COVID) which include neurological and behavioral effects in thousands of patients, but the etiology and scope of symptoms is not well understood. This paper reviews long term sequelae of COVID on brain and mental health in patients with the Long COVID syndrome. METHODS This was a literature review which queried databases for Pubmed, Psychinfo, and Medline for the following topics for January 1, 2020-July 15, 2023: Long COVID, PASC, brain, brain imaging, neurological, neurobiology, mental health, anxiety, depression. RESULTS Tens of thousands of patients have developed Long COVID, with the most common neurobehavioral symptoms anosmia (loss of smell) and fatigue. Anxiety and mood disorders are elevated and seen in about 25% of Long COVID patients. Neuropsychological testing studies show a correlation between symptom severity and cognitive dysfunction, while brain imaging studies show global decreases in gray matter and alterations in olfactory and other brain areas. CONCLUSIONS Studies to date show an increase in neurobehavioral disturbances in patients with Long COVID. Future research is needed to determine mechanisms.
Collapse
Affiliation(s)
- J Douglas Bremner
- Departments of Psychiatry & Behavioral Sciences and Radiology, Emory University School of Medicine, Atlanta Georgia, and the Atlanta VA Medical Center, Decatur, GA, USA; Nash Family Department Neuroscience and Brain-Body Research Center, Icahn School of Medicine at Mt. Sinai, New York, NY, USA; Department of Child and Adolescent Psychiatry, New York University (NYU) Langone Health, New York, NY, USA.
| | - Scott J Russo
- Nash Family Department Neuroscience and Brain-Body Research Center, Icahn School of Medicine at Mt. Sinai, New York, NY, USA
| | - Richard Gallagher
- Department of Child and Adolescent Psychiatry, New York University (NYU) Langone Health, New York, NY, USA; Department of Psychiatry, New York University (NYU) Langone Health, New York, NY, USA
| | - Naomi M Simon
- Department of Psychiatry, New York University (NYU) Langone Health, New York, NY, USA
| |
Collapse
|
13
|
Guedj E, Cionca A, Péron JA, Ayubcha C, Assal F, Horowitz T, Alavi A. Long Coronavirus Disease and the Brain: Molecular Neuroimaging Insights into Neurologic and Psychiatric Sequelae. PET Clin 2025; 20:39-55. [PMID: 39482218 DOI: 10.1016/j.cpet.2024.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has led to a variety of health challenges, with "long COVID" emerging as a widespread and debilitating post-acute syndrome among a considerable number of infected patients. This PET review synthesizes current evidence of the neurologic and psychiatric sequelae of COVID. This review also explores the pathophysiological mechanisms of these results, including astrocyte dysfunction and glutamate dysregulation, as well as the multimodal comparison to MR imaging findings. The findings underscore the potential for long-term brain injury. Additionally, the authors discuss the role of advanced imaging multimodal techniques in diagnosing, monitoring, and guiding treatment strategies for long COVID.
Collapse
Affiliation(s)
- Eric Guedj
- Biophysics and Nuclear Medicine, Aix Marseille University, Marseille, France; APHM, CNRS, Centrale Marseille, Institut Fresnel, Timone Hospital, Marseille, France; Nuclear Medicine Department, CERIMED, Marseille, France.
| | - Alexandre Cionca
- Neuro-X Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Julie A Péron
- Clinical and Experimental Neuropsychology Laboratory, Faculty of Psychology, University of Geneva, Geneva, Switzerland; Neurology Division, Geneva University Hospitals, Geneva, Switzerland
| | - Cyrus Ayubcha
- Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Frédéric Assal
- Neurology Division, Geneva University Hospitals, Geneva, Switzerland; Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Tatiana Horowitz
- Biophysics and Nuclear Medicine, Aix Marseille University, Marseille, France; APHM, CNRS, Centrale Marseille, Institut Fresnel, Timone Hospital, Marseille, France; Nuclear Medicine Department, CERIMED, Marseille, France
| | - Abass Alavi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
14
|
Hawley HB. Long COVID: Clinical Findings, Pathology, and Endothelial Molecular Mechanisms. Am J Med 2025; 138:91-97. [PMID: 37704072 DOI: 10.1016/j.amjmed.2023.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/29/2023] [Accepted: 08/08/2023] [Indexed: 09/15/2023]
Abstract
Persistence of COVID-19 symptoms may follow severe acute respiratory syndrome coronavirus 2 infection. The incidence of long COVID increases with the severity of acute disease, but even mild disease can be associated with sequelae. The symptoms vary widely, with fatigue, shortness of breath, and cognitive dysfunction the most common. Abnormalities of multiple organs have been documented, and histopathology has revealed widespread microthrombi. Elevated levels of complement are present in acute COVID-19 patients and may persist at lower levels in long COVID. Evidence supports complement activation, with endotheliopathy-associated disease as the molecular mechanism causing both acute and long COVID.
Collapse
Affiliation(s)
- H Bradford Hawley
- Department of Medicine, Wright State University Boonshoft School of Medicine, Dayton, Ohio.
| |
Collapse
|
15
|
Daskoulidou N, Carpanini SM, Zelek WM, Morgan BP. Involvement of Complement in Alzheimer's Disease: From Genetics Through Pathology to Therapeutic Strategies. Curr Top Behav Neurosci 2025; 69:3-24. [PMID: 39455500 DOI: 10.1007/7854_2024_524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
Complement is a critical component of innate immunity, evolved to defend against pathogens and clear toxic debris ranging from dead and dying cells to immune complexes. These roles make complement a key player in homeostasis; however, complement has a dark side. When the rigid control mechanisms fail, complement becomes dysregulated, acting as a driver of inflammation and resultant pathology in numerous diseases. Roles of complement in Alzheimer's disease (AD) and other dementias have emerged in recent years, supported by genetic, biomarker and pathological evidence and animal model studies. Numerous questions remain regarding the precise roles of complement in the brain in health and disease, including where and when complement is expressed, how it contributes to immune defence and garbage disposal in the healthy brain, and exactly how complement contributes to pathology in dementias. In this brief review, we will summarise current knowledge on complement roles in brain, present the evidence implicating complement in AD and explore whether complement represents an attractive therapeutic target for AD.
Collapse
Affiliation(s)
| | - Sarah M Carpanini
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff, UK
| | - Wioleta M Zelek
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff, UK
| | - B Paul Morgan
- UK Dementia Research Institute Cardiff, Cardiff University, Cardiff, UK.
| |
Collapse
|
16
|
Nath A, Kolson DL. Reemerging Infectious Diseases and Neuroimmunologic Complications. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200356. [PMID: 39693583 PMCID: PMC11658811 DOI: 10.1212/nxi.0000000000200356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 11/07/2024] [Indexed: 12/20/2024]
Abstract
During the past decade (and beyond), neurologists have become aware of the emergence, persistence, and consequences of some familiar and new infections affecting the nervous system. Even among the familiar CNS infections, such as herpes virus, polyoma virus/JC, influenza, arbovirus, and hepatitis, challenges remain in developing effective antiviral treatments and treatments of postinfection sequelae. With the changing environment and increased global travel, arthropod vectors that mediate zoonotic disease transmission have spread unfamiliar viruses such as West Nile virus, dengue, chikungunya, equine encephalitis, and Zika, among others. Although the global health impact of these diseases has not risen to that of COVID-19 and HIV, it is likely to dramatically increase with continued spread of transmission vectors and the emergence of new zoonotic animal-to-human diseases mediated by those transmission vectors. Furthermore, specific virus-targeting treatments or effective vaccines for arboviral infections are not yet available, and this represents a major challenge in limiting the morbidity of these infections. By contrast, HIV-1, a disease that originated by direct transmission from nonhuman primates to humans (as early as the 1930s), after many years of intense study, is now targeted by highly specific and effective antiviral drugs that can limit the spread of infection and extend human life and health in all populations. Even with these dramatic therapeutic effects of suppressing HIV replication, neurologic dysfunction (primarily cognitive impairment) affects significant numbers of persons living with HIV. This emphasizes not only the importance of treating the underlying infection but also developing treatments for legacy effects of the initial infection even after antiviral therapy. Notably, the rapid emergence of SARS-CoV-2 infection was met with rapid implementation of highly effective and specific antiviral therapies. This resulted in early and dramatic lowering of the morbidity and mortality of SARS-CoV-2 infection. Nonetheless, the postinfectious complications of SARS-CoV-2 infection (long COVID) are now among the more costly consequences of emerging zoonotic infections worldwide. Developing new antiviral therapies that can penetrate the CNS, vaccines, and therapies that target host immune responses and metabolic dysfunction will be necessary for management of infectious and postinfectious complications of established and emerging infections.
Collapse
Affiliation(s)
- Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD; and
| | - Dennis L Kolson
- Department of Neurology, University of Pennsylvania, Philadelphia
| |
Collapse
|
17
|
Nersesjan V, Boldingh MI, Paulsen EQ, Argren M, Høgestøl E, Aamodt AH, Popperud TH, Kondziella D, Jørgensen CS, Jensen VVS, Benros ME. Antibodies against SARS-CoV-2 spike protein in the cerebrospinal fluid of COVID-19 patients and vaccinated controls: a multicentre study. J Neurol 2024; 272:60. [PMID: 39680178 DOI: 10.1007/s00415-024-12769-4] [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: 09/09/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 12/17/2024]
Abstract
INTRODUCTION SARS-CoV-2 antibodies in the cerebrospinal fluid (CSF) of COVID-19 patients possibly reflect blood-cerebrospinal fluid barrier (BCB) disruption due to systemic inflammation. However, some studies indicate that CSF antibodies signal a neurotropic infection. Currently, larger studies are needed to clarify this, and it is unknown if CSF antibodies appear solely after infection or also after COVID-19 vaccination. Therefore, we aimed to investigate the CSF dynamics of SARS-CoV-2 antibodies in a multicenter study of COVID-19 patients and vaccinated controls. METHODS A cohort study of Danish and Norwegian COVID-19 patients and controls investigated with a lumbar puncture (April 2020-December 2022). Serum and CSF were analysed locally for routine investigations, and centrally at Statens Serum Institut (Danish governmental public health institute) for SARS-CoV-2 IgG antibodies against the spike protein using the Euroimmun (quantitative) and Wantai (qualitative) assays. Primary outcome was the quantity of CSF SARS-CoV-2 antibodies post-COVID versus post-vaccination. Secondary outcomes included regression models examining the relationship between CSF antibodies and serum levels, albumin ratio, CSF pleocytosis, COVID-19 severity, and temporal antibody dynamics. RESULTS We included 124 individuals (Mean [SD] age 47.2 [16.6]; 59.7% males surviving COVID-19 and controls. Of these, 86 had paired CSF-serum testing. Antibody-index calculations did not support a SARS-CoV-2 brain infection. Multi-variate regression revealed that CSF SARS-CoV-2 antibodies were most strongly influenced by serum antibody levels and BCB permeability, as measured by increasing albumin ratio. CSF antibody levels displayed a dose-response relationship (p < 0.0001) influenced by preceding vaccinations or infections. CSF antibody levels (median [IQR]) were highest among those both previously infected and vaccinated, 100.0 [25.0-174.0], and those vaccinated without prior infection, 85.0 [12.0-142.0], and lowest among previously infected individuals without preceding vaccination, 5.9 [2.7-55.1], (p = 0.003). SARS-CoV-2 antibodies in CSF were also detected via qualitative assays in the COVID-19 (46.8%) and vaccinated (78.6%) groups, p = 0.03. CONCLUSION SARS-CoV-2 antibodies detected in CSF can be derived following both infection and vaccination for COVID-19. CSF antibody levels increase in a dose-response relationship with the number of prior infections and vaccinations and are most strongly influenced by serum antibody levels and BCB permeability. These findings stress the importance of carefully interpreting CSF antibody results when assessing neurological complications following infections not categorized as neurotropic.
Collapse
Affiliation(s)
- Vardan Nersesjan
- Copenhagen Research Center for Biological and Precision Psychiatry, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | | | | | - Maria Argren
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Einar Høgestøl
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Department of Psychology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne Hege Aamodt
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- Department of Neuromedicine and Movement Science, The Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Daniel Kondziella
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Charlotte Sværke Jørgensen
- Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen, Denmark
| | | | - Michael E Benros
- Copenhagen Research Center for Biological and Precision Psychiatry, Mental Health Centre Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
18
|
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
|
19
|
Gaudry C, Dhersin R, Dubée V. [Mechanisms of prolonged symptoms following acute COVID-19: Some pathophysiological pathways]. Rev Mal Respir 2024; 41:660-668. [PMID: 39426876 DOI: 10.1016/j.rmr.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/30/2024] [Indexed: 10/21/2024]
Abstract
INTRODUCTION Following the Omicron wave in early 2022, an estimated 60-70% of the French population was infected with the SARS-CoV-2 virus. One out of ten infected subjects could have persistent symptoms three months after infection, representing a public health challenge. CURRENT STATE OF KNOWLEDGE The persistent symptoms may be secondary to diverse entities with distinct mechanisms. While organic infection sequelae occur mainly after severe COVID-19, some symptoms appear to be essentially psychological in origin; in addition, many subjects present stereotyped symptoms of fluctuating intensity with no identified anatomical or psychic substratum, often in the aftermath of a benign infection. The most frequent complaints are fatigue, pain, dyspnea and difficulty concentrating. PERSPECTIVES The hypotheses explored to explain these symptoms include: persistent immune dysfunction, inducted autoimmunity, and microbiome disturbances. Persistent viral antigens may lie at the crossroads of these mechanisms. To date, these different etiological avenues have yet to lead to the development of diagnostic tests or specific therapeutic strategies. CONCLUSION Prolonged symptoms after COVID-19 correspond to heterogeneous nosological entities with poorly understood mechanisms.
Collapse
Affiliation(s)
- C Gaudry
- Service des maladies infectieuses et tropicales, CHU d'Angers, 4, rue Larrey, 49100 Angers, France
| | - R Dhersin
- Service des maladies infectieuses et tropicales, CHU d'Angers, 4, rue Larrey, 49100 Angers, France
| | - V Dubée
- Service des maladies infectieuses et tropicales, CHU d'Angers, 4, rue Larrey, 49100 Angers, France.
| |
Collapse
|
20
|
Tavee J. Current concepts in long COVID-19 brain fog and postural orthostatic tachycardia syndrome. Ann Allergy Asthma Immunol 2024; 133:522-530. [PMID: 39154907 DOI: 10.1016/j.anai.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/20/2024]
Abstract
Neurologic complications of long COVID-19 syndrome are one of the leading causes of global disability. In particular, post-COVID-19 cognitive dysfunction and dysautonomia in the form of postural orthostatic tachycardia syndrome (POTS) markedly affect patient quality of life and ability to return to work. The underlying pathophysiology of post-COVID-19 neurologic complications is unknown but is likely multifactorial with immune dysregulation and microvascular dysfunction playing central roles. Specific pathogenic factors with supportive evidence to date include cytokine-mediated inflammation, autoantibodies, immune exhaustion, disruption of the renin-angiotensin system, reduced serotonin levels, and microglial activation. The prevalence of post-COVID-19 cognitive dysfunction ranges from 10% to 88% and is affected by viral variant and hospitalization status among other factors, whereas that of long COVID-19 POTS is unknown due to referral bias and varying definitions. Treatment is largely supportive and often incorporates combined modalities. Marginal benefits with cognitive behavioral therapy, hyperbaric oxygen therapy, and supplements have been found for post-COVID-19 brain fog, whereas established POTS therapies aimed at improving venous return and reducing heart rate may reduce symptoms of long COVID-19 POTS. Although significant recovery has been noted for many cases of post-COVID-19 brain fog and POTS, prospective studies have revealed evidence of persistent symptoms and neurologic deficits a year after infection in some patients. Further studies that provide insight into the underlying pathophysiology of long COVID-19 are needed for development of target directed therapy.
Collapse
Affiliation(s)
- Jinny Tavee
- Division of Neurology, National Jewish Health, Denver, Colorado.
| |
Collapse
|
21
|
Lu J, Zuo X, Cai A, Xiao F, Xu Z, Wang R, Miao C, Yang C, Zheng X, Wang J, Ding X, Xiong W. Cerebral small vessel injury in mice with damage to ACE2-expressing cerebral vascular endothelial cells and post COVID-19 patients. Alzheimers Dement 2024; 20:7971-7988. [PMID: 39352003 PMCID: PMC11567838 DOI: 10.1002/alz.14279] [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: 02/26/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 10/03/2024]
Abstract
INTRODUCTION The angiotensin-converting enzyme 2 (ACE2), which is expressed in cerebral vascular endothelial cells (CVECs), has been currently identified as a functional receptor for SARS-CoV-2. METHODS We specifically induced injury to ACE2-expressing CVECs in mice and evaluated the effects of such targeted damage through magnetic resonance imaging (MRI) and cognitive behavioral tests. In parallel, we recruited a single-center cohort of COVID-19 survivors and further assessed their brain microvascular injury based on cognition and emotional scales, cranial MRI scans, and blood proteomic measurements. RESULTS Here, we show an array of pathological and behavioral alterations characteristic of cerebral small vessel disease (CSVD) in mice that targeted damage to ACE2-expressing CVECs, and COVID-19 survivors. These CSVD-like manifestations persist for at least 7 months post-recovery from COVID-19. DISCUSSION Our findings suggest that SARS-CoV-2 may induce cerebral small vessel damage with persistent sequelae, underscoring the imperative for heightened clinical vigilance in mitigating or treating SARS-CoV-2-mediated cerebral endothelial injury throughout infection and convalescence. HIGHLIGHTS Cerebral small vessel disease-associated changes were observed after targeted damage to angiotensin-converting enzyme 2-expressing cerebral vascular endothelial cells. SARS-CoV-2 may induce cerebral small vessel damage with persistent sequelae. Clinical vigilance is needed in preventing SARS-CoV-2-induced cerebral endothelial damage during infection and recovery.
Collapse
Affiliation(s)
- Jieping Lu
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Xin Zuo
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial IntelligenceHefei Comprehensive National Science CenterHefeiChina
| | - Aoling Cai
- Key Laboratory of Magnetic Resonance in Biological SystemsState Key Laboratory of Magnetic Resonance and Atomic and Molecular PhysicsNational Center for Magnetic Resonance in WuhanWuhan Institute of Physics and MathematicsInnovation Academy for Precision Measurement Science and TechnologyChinese Academy of Sciences‐Wuhan National Laboratory for OptoelectronicsWuhanChina
- The Affiliated Changzhou Second People's Hospital of Nanjing Medical UniversityChangzhou Second People's HospitalChangzhou Medical CenterNanjing Medical UniversityChangzhouChina
| | - Fang Xiao
- Department of RadiologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Zhenyu Xu
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Rui Wang
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Chenjian Miao
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Chen Yang
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Xingxing Zheng
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Jie Wang
- Key Laboratory of Magnetic Resonance in Biological SystemsState Key Laboratory of Magnetic Resonance and Atomic and Molecular PhysicsNational Center for Magnetic Resonance in WuhanWuhan Institute of Physics and MathematicsInnovation Academy for Precision Measurement Science and TechnologyChinese Academy of Sciences‐Wuhan National Laboratory for OptoelectronicsWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Xiaoling Ding
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Wei Xiong
- Department of NeurologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial IntelligenceHefei Comprehensive National Science CenterHefeiChina
- Anhui Province Key Laboratory of Biomedical Aging ResearchHefeiChina
- CAS Key Laboratory of Brain Function and DiseaseHefeiChina
| |
Collapse
|
22
|
Dunai C, Hetherington C, Boardman SA, Clark JJ, Sharma P, Subramaniam K, Tharmaratnam K, Needham EJ, Williams R, Huang Y, Wood GK, Collie C, Fower A, Fox H, Ellul MA, Held M, Egbe FN, Griffiths M, Solomon T, Breen G, Kipar A, Cavanagh J, Irani SR, Vincent A, Stewart JP, Taams LS, Menon DK, Michael BD. Pulmonary SARS-CoV-2 infection leads to para-infectious immune activation in the brain. Front Immunol 2024; 15:1440324. [PMID: 39474424 PMCID: PMC11519853 DOI: 10.3389/fimmu.2024.1440324] [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: 05/29/2024] [Accepted: 09/04/2024] [Indexed: 11/02/2024] Open
Abstract
Neurological complications, including encephalopathy and stroke, occur in a significant proportion of COVID-19 cases but viral protein is seldom detected in the brain parenchyma. To model this situation, we developed a novel low-inoculum K18-hACE2 mouse model of SARS-CoV-2 infection during which active viral replication was consistently seen in mouse lungs but not in the brain. We found that several mediators previously associated with encephalopathy in clinical samples were upregulated in the lung, including CCL2, and IL-6. In addition, several inflammatory mediations, including CCL4, IFNγ, IL-17A, were upregulated in the brain, associated with microglial reactivity. Parallel in vitro experiments demonstrated that the filtered supernatant from SARS-CoV-2 virion exposed brain endothelial cells induced activation of uninfected microglia. This model successfully recreates SARS-CoV-2 virus-associated para-infectious brain inflammation which can be used to study the pathophysiology of the neurological complications and the identification of potential immune targets for treatment.
Collapse
Affiliation(s)
- Cordelia Dunai
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, United Kingdom
- Clinical Infection Microbiology and Immunology, Institute of Infection Ecology and Veterinary Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Claire Hetherington
- Clinical Infection Microbiology and Immunology, Institute of Infection Ecology and Veterinary Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Sarah A. Boardman
- Clinical Infection Microbiology and Immunology, Institute of Infection Ecology and Veterinary Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Jordan J. Clark
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Parul Sharma
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Krishanthi Subramaniam
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Kukatharmini Tharmaratnam
- Department of Health Data Science, Institute of Population Health, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Edward J. Needham
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Robyn Williams
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Yun Huang
- Clinical Infection Microbiology and Immunology, Institute of Infection Ecology and Veterinary Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Greta K. Wood
- Clinical Infection Microbiology and Immunology, Institute of Infection Ecology and Veterinary Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Ceryce Collie
- Clinical Infection Microbiology and Immunology, Institute of Infection Ecology and Veterinary Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Andrew Fower
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Hannah Fox
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Mark A. Ellul
- Clinical Infection Microbiology and Immunology, Institute of Infection Ecology and Veterinary Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Marie Held
- Centre for Cell Imaging, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Franklyn N. Egbe
- Clinical Infection Microbiology and Immunology, Institute of Infection Ecology and Veterinary Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Michael Griffiths
- Clinical Infection Microbiology and Immunology, Institute of Infection Ecology and Veterinary Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Tom Solomon
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, United Kingdom
- Clinical Infection Microbiology and Immunology, Institute of Infection Ecology and Veterinary Sciences, University of Liverpool, Liverpool, United Kingdom
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
| | - Gerome Breen
- Department of Social, Genetic & Developmental Psychiatry Centre, School of Mental Health & Psychological Sciences, King’s College London, London, United Kingdom
- NIHR Maudsley Biomedical Research Centre, King’s College London, London, United Kingdom
| | - Anja Kipar
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Laboratory for Animal Model Pathology, Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Jonathan Cavanagh
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sarosh R. Irani
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
| | - Angela Vincent
- Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - James P. Stewart
- Department of Infection Biology and Microbiomes, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Leonie S. Taams
- Centre for Inflammation Biology and Cancer Immunology, Department of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - David K. Menon
- Division of Anaesthesia, Addenbrooke’s Hospital, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Benedict D. Michael
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, United Kingdom
- Clinical Infection Microbiology and Immunology, Institute of Infection Ecology and Veterinary Sciences, University of Liverpool, Liverpool, United Kingdom
- Department of Neurology, The Walton Centre NHS Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
23
|
He X, Zhang X, Zhong W. Emerging small-molecule antiviral agents in long COVID prevention. Front Pharmacol 2024; 15:1457672. [PMID: 39444602 PMCID: PMC11496125 DOI: 10.3389/fphar.2024.1457672] [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: 07/01/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Long COVID, or Post-Acute Sequelae of COVID-19 (PASC), was characterized by persistent symptoms such as fatigue, shortness of breath, and cognitive impairments. These symptoms, emerging one to 2 months post-infection and persisting for several months, cannot be attributed to other diagnoses. The pathophysiology of long COVID remained elusive; however, emerging studies suggested multiple potential mechanisms, including the reactivation of Epstein-Barr virus, persistent SARS-CoV-2 reservoirs, neuroinflammation, and vascular damage, which may contribute to its development. Long COVID affected multiple organ systems, including respiratory, circulatory, and nervous systems, leading to a range of functional impairments. Additionally, it showed a profound impact on mental health, manifesting as anxiety and depression, which significantly degraded the quality of life. The absence of definitive treatments underscored the importance of prevention. Recent evidence indicated that early antiviral intervention-particularly with small-molecule drugs such as Metformin, Ensitrelvir, Molnupiravir, and Nirmatrelvir-may effectively reduce the incidence of long COVID. This underscored the promising role of small-molecule compounds in mitigating long-term COVID-19 consequences, offering a novel preventive strategy against long COVID and its extensive impacts on patients.
Collapse
Affiliation(s)
- Xiaomeng He
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiang Zhang
- Department of Blood Transfusion Medicine, The 940th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Lanzhou, China
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
24
|
Fitzgerald ML, Cohen AK, Jaudon TW, Vogel JM, Koppes AN, Santos L, Robles R, Lin J, Davids JD, McWilliams C, Redfield S, Banks KP, Richardson M, Tindle Akintonwa TT, Pollack B, Spier E, Weiss A, Assaf G, Davis H, McCorkell L. A call from patient-researchers to advance research on long COVID. Cell 2024; 187:5490-5496. [PMID: 39366339 DOI: 10.1016/j.cell.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 10/06/2024]
Abstract
Long COVID is a chronic and often disabling illness with long-term consequences. Although progress has been made in the clinical characterization of long COVID, no approved treatments exist and disconnects between patients and researchers threaten to hinder future progress. Incorporating patients as active collaborators in long COVID research can bridge the gap and accelerate progress toward treatments and cures.
Collapse
Affiliation(s)
| | - Alison K Cohen
- Patient-Led Research Collaborative, Washington, DC, USA; University of California, San Francisco, San Francisco, CA, USA
| | - Toni Wall Jaudon
- Patient-Led Research Collaborative, Washington, DC, USA; University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Julia Moore Vogel
- Patient-Led Research Collaborative, Washington, DC, USA; Scripps Research, La Jolla, CA, USA
| | - Abigail N Koppes
- Patient-Led Research Collaborative, Washington, DC, USA; Northeastern University, Department of Chemical Engineering, Boston, MA, USA
| | - Lucia Santos
- Patient-Led Research Collaborative, Washington, DC, USA
| | - Rachel Robles
- Patient-Led Research Collaborative, Washington, DC, USA; Universität Bern, Bern, Switzerland
| | - Jerry Lin
- Patient-Led Research Collaborative, Washington, DC, USA
| | - J D Davids
- Patient-Led Research Collaborative, Washington, DC, USA; Strategies for High Impact/Long COVID Justice, Detroit, MI, USA
| | - Chris McWilliams
- Patient-Led Research Collaborative, Washington, DC, USA; University of Bristol, Bristol, UK
| | | | - Kathleen P Banks
- Patient-Led Research Collaborative, Washington, DC, USA; Boston University School of Public Health, Boston, MA, USA
| | - Maria Richardson
- Patient-Led Research Collaborative, Washington, DC, USA; Millions Missing México, Mexico City, Mexico
| | - Teresa T Tindle Akintonwa
- Patient-Led Research Collaborative, Washington, DC, USA; Black COVID-19 Survivors Alliance, Marietta, GA, USA
| | - Beth Pollack
- Patient-Led Research Collaborative, Washington, DC, USA; Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ezra Spier
- Patient-Led Research Collaborative, Washington, DC, USA
| | - Aimee Weiss
- Patient-Led Research Collaborative, Washington, DC, USA
| | - Gina Assaf
- Patient-Led Research Collaborative, Washington, DC, USA
| | - Hannah Davis
- Patient-Led Research Collaborative, Washington, DC, USA
| | | |
Collapse
|
25
|
Sapio MR, Staedtler ES, King DM, Maric D, Jahanipour J, Ghetti A, Jacobson KA, Mannes AJ, Iadarola MJ. Analgesic candidate adenosine A 3 receptors are expressed by perineuronal peripheral macrophages in human dorsal root ganglion and spinal cord microglia. Pain 2024; 165:2323-2343. [PMID: 38691673 PMCID: PMC11408117 DOI: 10.1097/j.pain.0000000000003242] [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: 11/09/2023] [Accepted: 02/22/2024] [Indexed: 05/03/2024]
Abstract
ABSTRACT Adenosine receptors are a family of purinergic G protein-coupled receptors that are widely distributed in bodily organs and in the peripheral and central nervous systems. Recently, antihyperalgesic actions have been suggested for the adenosine A 3 receptor, and its agonists have been proposed as new neuropathic pain treatments. We hypothesized that these receptors may be expressed in nociceptive primary afferent neurons. However, RNA sequencing across species, eg, rat, mouse, dog, and human, suggests that dorsal root ganglion (DRG) expression of ADORA3 is inconsistent. In rat and mouse, Adora3 shows very weak to no expression in DRG, whereas it is well expressed in human DRG. However, the cell types in human DRG that express ADORA3 have not been delineated. An examination of DRG cell types using in situ hybridization clearly detected ADORA3 transcripts in peripheral macrophages that are in close apposition to the neuronal perikarya but not in peripheral sensory neurons. By contrast, ADORA1 was found primarily in neurons, where it is broadly expressed at low levels. These results suggest that a more complex or indirect mechanism involving modulation of macrophage and/or microglial cells may underlie the potential analgesic action of adenosine A 3 receptor agonism.
Collapse
Affiliation(s)
- Matthew R Sapio
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Ellen S Staedtler
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Diana M King
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD, United States
| | - Jahandar Jahanipour
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, Bethesda, MD, United States
| | - Andre Ghetti
- AnaBios Corporation, San Diego, CA, United States
| | - Kenneth A Jacobson
- National Institute of Diabetes and Digestive and Kidney Diseases, Molecular Recognition Section, Laboratory of Bioorganic Chemistry, Bethesda, MD, United States
| | - Andrew J Mannes
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Michael J Iadarola
- Department of Perioperative Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
26
|
Desai R, Mellacheruvu SP, Akella SA, Mohammed AS, Hussain M, Mohammed AA, Saketha P, Sunkara P, Gummadi J, Ghantasala P. Recurrent stroke admissions with vs without COVID-19 and associated in-hospital mortality: A United States nationwide analysis, 2020. World J Virol 2024; 13:96453. [PMID: 39323442 PMCID: PMC11401001 DOI: 10.5501/wjv.v13.i3.96453] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/12/2024] [Accepted: 07/10/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has been shown to increase the risk of stroke. However, the prevalence and risk of recurrent stroke in COVID-19 patients with prior stroke/transient ischemic attack (TIA), as well as its impact on mortality, are not established. AIM To evaluate the impact of COVID-19 on in-hospital mortality, length of stay, and healthcare costs in patients with recurrent strokes. METHODS We identified admissions of recurrent stroke (current acute ischemic stroke admissions with at least one prior TIA or stroke) in patients with and without COVID-19 using ICD-10-CM codes using the National Inpatient Sample (2020). We analyzed the impact of COVID-19 on mortality following recurrent stroke admissions by subgroups. RESULTS Of 97455 admissions with recurrent stroke, 2140 (2.2%) belonged to the COVID-19-positive group. The COVID-19-positive group had a higher prevalence of diabetes and chronic kidney disease vs the COVID-19 negative group (P < 0.001). Among the subgroups, patients aged > 65 years, patients aged 45-64 years, Asians, Hispanics, whites, and blacks in the COVID-19 positive group had higher rates of all-cause mortality than the COVID-19 negative group (P < 0.01). Higher odds of in-hospital mortality were seen in the group aged 45-64 (OR: 8.40, 95%CI: 4.18-16.91) vs the group aged > 65 (OR: 7.04, 95%CI: 5.24-9.44), males (OR: 7.82, 95%CI: 5.38-11.35) compared to females (OR: 6.15, 95%CI: 4.12-9.18), and in Hispanics (OR: 15.47, 95%CI: 7.61-31.44) and Asians/Pacific Islanders (OR: 14.93, 95%CI: 7.22-30.87) compared to blacks (OR: 5.73, 95%CI: 3.08-10.68), and whites (OR: 5.54, 95%CI: 3.79-8.09). CONCLUSION The study highlights the increased risk of all-cause in-hospital mortality in recurrent stroke patients with COVID-19, with a more pronounced increase in middle-aged patients, males, Hispanics, or Asians.
Collapse
Affiliation(s)
- Rupak Desai
- Outcomes Research, Independent Researcher, Atlanta, GA 30033, United States
| | | | - Sai Anusha Akella
- Department of Internal Medicine, One Brooklyn Health- Interfaith Medical Center, Brooklyn, NY 11213, United States
| | - Adil Sarvar Mohammed
- Department of Internal Medicine, Central Michigan University College of Medicine, Saginaw, MI 48602, United States
| | - Mushfequa Hussain
- Department of Internal Medicine, Kamineni Institute of Medical Sciences, Narketpally 508254, India
| | - Abdul Aziz Mohammed
- Department of Internal Medicine, Kamineni Institute of Medical Sciences, Narketpally 508254, India
| | - Pakhal Saketha
- Department of Internal Medicine, Bhaskar Medical College, Moinabad 500075, Hyderabad, India
| | - Praveena Sunkara
- Department of Internal Medicine, MedStar Medical Group, Charlotte Hall, MD 20622, United States
| | - Jyotsna Gummadi
- Department of Medicine, Medstar Franklin Square Medical Center, Baltimore, MD 21237, United States
| | - Paritharsh Ghantasala
- Department of Internal Medicine, Central Michigan University College of Medicine, Saginaw, MI 48602, United States
| |
Collapse
|
27
|
Liu P, Ernst T, Liang H, Jiang D, Cunningham E, Ryan M, Lu H, Kottilil S, Chang L. Elevated cerebral oxygen extraction in patients with post-COVID conditions. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2024; 3:169-174. [PMID: 39959306 PMCID: PMC11823640 DOI: 10.1515/nipt-2024-0014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/11/2024] [Indexed: 02/18/2025]
Abstract
Objectives Dysfunction of cerebral microcirculation due to SARS-CoV-2 infection has been postulated to be a plausible mechanism for the neurological symptoms of post-COVID-19 conditions (neuro-PCC), affecting oxygen homeostasis in the brain. In this study, we aimed to investigate the balance between cerebral oxygen delivery and consumption, measured by oxygen extraction fraction (OEF), in patients with neuro-PCC. Methods 25 participants with neuro-PCC (8 previously hospitalized and 17 not hospitalized) and 59 age-matched healthy controls were studied. Global OEF was quantified using TRUST MRI and compared across the three groups. Associations between OEF and neurobehavioral measures were also evaluated in participants with neuro-PCC. Results OEF was significantly different (one-way ANCOVA-p=0.046) among the three groups, after accounting for age and sex. On post-hoc analyses, previously hospitalized neuro-PCC participants had significantly higher OEF (42.40 ± 5.40 %) than both uninfected controls (37.70 ± 5.09 %, p=0.032) and neuro-PCC participants without hospitalization (37.02 ± 5.05 %, p=0.015). Within the participants with neuro-PCC, OEF was significantly associated with locomotor function assessed with the 4-m walk gait speed score (β=-0.03, r=0.34, p=0.003). Conclusions Participants with neuro-PCC had altered cerebral OEF, which is also associated with slower locomotion. OEF is a promising marker for studying neuro-PCC.
Collapse
Affiliation(s)
- Peiying Liu
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Thomas Ernst
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Huajun Liang
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dengrong Jiang
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Eric Cunningham
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Meghann Ryan
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hanzhang Lu
- Department of Radiology, Johns Hopkins University, Baltimore, MD, USA
| | - Shyamasundaran Kottilil
- Department of Medicine, Institutue of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Linda Chang
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
28
|
Ryu JK, Yan Z, Montano M, Sozmen EG, Dixit K, Suryawanshi RK, Matsui Y, Helmy E, Kaushal P, Makanani SK, Deerinck TJ, Meyer-Franke A, Rios Coronado PE, Trevino TN, Shin MG, Tognatta R, Liu Y, Schuck R, Le L, Miyajima H, Mendiola AS, Arun N, Guo B, Taha TY, Agrawal A, MacDonald E, Aries O, Yan A, Weaver O, Petersen MA, Meza Acevedo R, Alzamora MDPS, Thomas R, Traglia M, Kouznetsova VL, Tsigelny IF, Pico AR, Red-Horse K, Ellisman MH, Krogan NJ, Bouhaddou M, Ott M, Greene WC, Akassoglou K. Fibrin drives thromboinflammation and neuropathology in COVID-19. Nature 2024; 633:905-913. [PMID: 39198643 PMCID: PMC11424477 DOI: 10.1038/s41586-024-07873-4] [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: 02/13/2023] [Accepted: 07/24/2024] [Indexed: 09/01/2024]
Abstract
Life-threatening thrombotic events and neurological symptoms are prevalent in COVID-19 and are persistent in patients with long COVID experiencing post-acute sequelae of SARS-CoV-2 infection1-4. Despite the clinical evidence1,5-7, the underlying mechanisms of coagulopathy in COVID-19 and its consequences in inflammation and neuropathology remain poorly understood and treatment options are insufficient. Fibrinogen, the central structural component of blood clots, is abundantly deposited in the lungs and brains of patients with COVID-19, correlates with disease severity and is a predictive biomarker for post-COVID-19 cognitive deficits1,5,8-10. Here we show that fibrin binds to the SARS-CoV-2 spike protein, forming proinflammatory blood clots that drive systemic thromboinflammation and neuropathology in COVID-19. Fibrin, acting through its inflammatory domain, is required for oxidative stress and macrophage activation in the lungs, whereas it suppresses natural killer cells, after SARS-CoV-2 infection. Fibrin promotes neuroinflammation and neuronal loss after infection, as well as innate immune activation in the brain and lungs independently of active infection. A monoclonal antibody targeting the inflammatory fibrin domain provides protection from microglial activation and neuronal injury, as well as from thromboinflammation in the lung after infection. Thus, fibrin drives inflammation and neuropathology in SARS-CoV-2 infection, and fibrin-targeting immunotherapy may represent a therapeutic intervention for patients with acute COVID-19 and long COVID.
Collapse
Affiliation(s)
- Jae Kyu Ryu
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Zhaoqi Yan
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Mauricio Montano
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Elif G Sozmen
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Karuna Dixit
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | | | - Yusuke Matsui
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Ekram Helmy
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Prashant Kaushal
- Department of Microbiology, Immunology and Molecular Genetics (MIMG), University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences (QCBio), University of California Los Angeles, Los Angeles, CA, USA
| | - Sara K Makanani
- Department of Microbiology, Immunology and Molecular Genetics (MIMG), University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences (QCBio), University of California Los Angeles, Los Angeles, CA, USA
| | - Thomas J Deerinck
- National Center for Microscopy and Imaging Research, Center for Research on Biological Systems, University of California San Diego, La Jolla, CA, USA
| | | | | | - Troy N Trevino
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Min-Gyoung Shin
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Reshmi Tognatta
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Yixin Liu
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Renaud Schuck
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Lucas Le
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Hisao Miyajima
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Andrew S Mendiola
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Nikhita Arun
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Brandon Guo
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Taha Y Taha
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
| | - Ayushi Agrawal
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Eilidh MacDonald
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Oliver Aries
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Aaron Yan
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Olivia Weaver
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Mark A Petersen
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Rosa Meza Acevedo
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Maria Del Pilar S Alzamora
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Reuben Thomas
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Michela Traglia
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Valentina L Kouznetsova
- San Diego Supercomputer Center, University of California San Diego, La Jolla, CA, USA
- CureScience Institute, San Diego, CA, USA
| | - Igor F Tsigelny
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA
- San Diego Supercomputer Center, University of California San Diego, La Jolla, CA, USA
- CureScience Institute, San Diego, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Alexander R Pico
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Center for Research on Biological Systems, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Nevan J Krogan
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
- Quantitative Biosciences Institute (QBI), University of California San Francisco, San Francisco, CA, USA
- COVID-19 Research Group (QCRG), University of California San Francisco, San Francisco, CA, USA
| | - Mehdi Bouhaddou
- Department of Microbiology, Immunology and Molecular Genetics (MIMG), University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences (QCBio), University of California Los Angeles, Los Angeles, CA, USA
| | - Melanie Ott
- Gladstone Institute of Virology, San Francisco, CA, USA
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA
- COVID-19 Research Group (QCRG), University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Warner C Greene
- Gladstone Institute of Virology, San Francisco, CA, USA.
- Michael Hulton Center for HIV Cure Research at Gladstone, San Francisco, CA, USA.
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA.
| | - Katerina Akassoglou
- Center for Neurovascular Brain Immunology at Gladstone and UCSF, San Francisco, CA, USA.
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
29
|
Strong MJ, McLellan C, Kaplanis B, Droppelmann CA, Junop M. Phase Separation of SARS-CoV-2 Nucleocapsid Protein with TDP-43 Is Dependent on C-Terminus Domains. Int J Mol Sci 2024; 25:8779. [PMID: 39201466 PMCID: PMC11354357 DOI: 10.3390/ijms25168779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
The SARS-CoV-2 nucleocapsid protein (N protein) is critical in viral replication by undergoing liquid-liquid phase separation to seed the formation of a ribonucleoprotein (RNP) complex to drive viral genomic RNA (gRNA) translation and in suppressing both stress granules and processing bodies, which is postulated to increase uncoated gRNA availability. The N protein can also form biomolecular condensates with a broad range of host endogenous proteins including RNA binding proteins (RBPs). Amongst these RBPs are proteins that are associated with pathological, neuronal, and glial cytoplasmic inclusions across several adult-onset neurodegenerative disorders, including TAR DNA binding protein 43 kDa (TDP-43) which forms pathological inclusions in over 95% of amyotrophic lateral sclerosis cases. In this study, we demonstrate that the N protein can form biomolecular condensates with TDP-43 and that this is dependent on the N protein C-terminus domain (N-CTD) and the intrinsically disordered C-terminus domain of TDP-43. This process is markedly accelerated in the presence of RNA. In silico modeling suggests that the biomolecular condensate that forms in the presence of RNA is composed of an N protein quadriplex in which the intrinsically disordered TDP-43 C terminus domain is incorporated.
Collapse
Affiliation(s)
- Michael J. Strong
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada; (C.M.); (C.A.D.)
- Department of Clinical Neurological Sciences, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Crystal McLellan
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada; (C.M.); (C.A.D.)
| | - Brianna Kaplanis
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada; (B.K.); (M.J.)
| | - Cristian A. Droppelmann
- Molecular Medicine Group, Robarts Research Institute, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada; (C.M.); (C.A.D.)
| | - Murray Junop
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada; (B.K.); (M.J.)
| |
Collapse
|
30
|
Mortazavi S, Rashedi V, Cheraghian B, Pourshams F, Saeidimehr S, Dehghan B, Pourshams M. Coronavirus disease 2019 and its impact on the cognition of older adults: Unraveling the role of inflammation. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2024; 19:100238. [PMID: 38779343 PMCID: PMC11109741 DOI: 10.1016/j.cpnec.2024.100238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND The Coronavirus Disease 2019 (COVID-19) pandemic significantly impacted the older adult population globally. This study aimed to investigate cognitive function and its relationship with inflammation in older COVID-19 survivors over a three-month follow-up to address concerns about cognitive impairment and its risk factors. METHODS In this descriptive-analytical study, 177 hospitalized COVID-19 patients aged >60 were assessed from July 2021 to February 2022. Psychiatric, global cognitive assessments and activities of daily living were conducted at discharge, 1 month, and 3 months post-discharge. Statistical analyses were conducted using SPSS Version 24. The evolution of cognitive status over time was evaluated using the Repeated Measures Test. The study probed into the association between inflammatory markers and cognitive function through the Pearson correlation test and the Mann-Whitney U test. Additionally, the link between anxiety/depression and cognitive performance was examined using the Pearson correlation. RESULTS Results indicated that higher levels of C-reactive protein (CRP), D-dimer, and Lactate Dehydrogenase (LDH) were correlated to reduced cognitive performance. Conversely, Erythrocyte Sedimentation Rate (ESR) and Creatine Phosphokinase (CPK) did not exhibit a significant relationship with cognitive scores. A positive correlation was observed between improved cognitive function (reflected by higher GPCOG scores) and lower levels of anxiety and depression (indicated by lower scores on the Hospital Anxiety and Depression Scale). Over the study period, cognitive function and anxiety scores showed an upward trend, whereas symptoms of depression and challenges in daily activities remained consistent. CONCLUSIONS The study highlights the enduring effects and detrimental role of inflammation on overall cognitive abilities among older survivors of COVID-19. It underscores the urgent need for specialized interventions and rehabilitative strategies to facilitate sustained cognitive recuperation among these individuals.
Collapse
Affiliation(s)
- Shahrzad Mortazavi
- Department of Psychiatry, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vahid Rashedi
- Iranian Research Center on Aging, Department of Aging, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Bahman Cheraghian
- Department of Biostatistics and Epidemiology, School of Public Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Pourshams
- Department of Neurology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Saeidimehr
- Family Health Research Center, Petroleum Industry Health Organization, Iran
| | - Bahram Dehghan
- Family Health Research Center, Petroleum Industry Health Organization, Iran
| | - Maryam Pourshams
- Department of Psychiatry, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
31
|
McClelland AC, Benitez SJ, Burns J. COVID-19 Neuroimaging Update: Pathophysiology, Acute Findings, and Post-Acute Developments. Semin Ultrasound CT MR 2024; 45:318-331. [PMID: 38518814 DOI: 10.1053/j.sult.2024.03.003] [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: 03/24/2024]
Abstract
COVID-19 has prominent effects on the nervous system with important manifestations on neuroimaging. In this review, we discuss the neuroimaging appearance of acute COVID-19 that became evident during the early stages of the pandemic. We highlight the underlying pathophysiology mediating nervous system effects and neuroimaging appearances including systemic inflammatory response such as cytokine storm, coagulopathy, and para/post-infections immune mediated phenomena. We also discuss the nervous system manifestations of COVID-19 and the role of imaging as the pandemic has evolved over time, including related to the development of vaccines and the emergence of post-acute sequalae such as long COVID.
Collapse
Affiliation(s)
| | - Steven J Benitez
- Department of Radiology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY
| | - Judah Burns
- Department of Radiology, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
32
|
Barros-Aragão FG, Pinto TP, Carregari VC, Rezende NB, Pinheiro TL, Reis-de-Oliveira G, Cabral-Castro MJ, Queiroz DC, Fonseca PL, Gonçalves AL, de Freitas GR, Sudo FK, Mattos P, Bozza FA, Rodrigues EC, Aguiar RS, Rodrigues RS, Brandão CO, Souza AS, Martins-de-Souza D, De Felice FG, Tovar-Moll F. Changes in neuroinflammatory biomarkers correlate with disease severity and neuroimaging alterations in patients with COVID-19 neurological complications. Brain Behav Immun Health 2024; 39:100805. [PMID: 39022627 PMCID: PMC11253226 DOI: 10.1016/j.bbih.2024.100805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/15/2024] [Accepted: 06/10/2024] [Indexed: 07/20/2024] Open
Abstract
COVID-19 induces acute and persistent neurological symptoms in mild and severe cases. Proposed concomitant mechanisms include direct viral infection and strain, coagulopathy, hypoxia, and neuroinflammation. However, underlying molecular alterations associated with multiple neurological outcomes in both mild and severe cases are majorly unexplored. To illuminate possible mechanisms leading to COVID-19 neurological disease, we retrospectively investigated in detail a cohort of 35 COVID-19 mild and severe hospitalized patients presenting neurological alterations subject to clinically indicated cerebrospinal fluid (CSF) sampling. Clinical and neurological investigation, brain imaging, viral sequencing, and cerebrospinal CSF analyses were carried out. We found that COVID-19 patients presented heterogeneous neurological symptoms dissociated from lung burden. Nasal swab viral sequencing revealed a dominant strain at the time of the study, and we could not detect traces of SARS-CoV-2's spike protein in patients' CSF by multiple reaction monitoring analysis. Patients presented ubiquitous systemic hyper-inflammation and broad alterations in CSF proteomics related to inflammation, innate immunity, and hemostasis, irrespective of COVID-19 severity or neuroimaging alterations. Elevated CSF interleukin-6 (IL6) correlated with disease severity (sex-, age-, and comorbidity-adjusted mean Severe 24.5 pg/ml, 95% confidence interval (CI) 9.62-62.23 vs. Mild 3.91 pg/mL CI 1.5-10.3 patients, p = 0.019). CSF tumor necrosis factor-alpha (TNFα) and IL6 levels were higher in patients presenting pronounced neuroimaging alterations compared to those who did not (sex-, age-, and comorbidity-adjusted mean TNFα Pronounced 3.4, CI 2.4-4.4 vs. Non-Pronounced 2.0, CI 1.4-2.5, p = 0.022; IL6 Pronounced 33.11, CI 8.89-123.31 vs Non-Pronounced 6.22, CI 2.9-13.34, p = 0.046). Collectively, our findings put neuroinflammation as a possible driver of COVID-19 acute neurological disease in mild and severe cases.
Collapse
Affiliation(s)
- Fernanda G.Q. Barros-Aragão
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Institute of Medical Biochemistry Leopoldo De Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil, 21941-902
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences & Department of Psychiatry, Queen's University, Kingston, Ontario, Canada, K7L 3N6
| | - Talita P. Pinto
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Victor C. Carregari
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil, 13083-862
| | - Nathane B.S. Rezende
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Thaís L. Pinheiro
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Institute of Medical Biochemistry Leopoldo De Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil, 21941-902
| | - Guilherme Reis-de-Oliveira
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil, 13083-862
| | - Mauro J. Cabral-Castro
- Institute of Microbiology Paulo de Goés, UFRJ, Rio de Janeiro, Brazil, 21941-902
- Department of Pathology, Faculty of Medicine, Universidade Federal Fluminense, Niterói, RJ, Brazil, 24210-346
| | - Daniel C. Queiroz
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil, 31270-901
| | - Paula L.C. Fonseca
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil, 31270-901
| | - Alessandro L. Gonçalves
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil, 31270-901
| | | | - Felipe K. Sudo
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Paulo Mattos
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Fernando A. Bozza
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Erika C. Rodrigues
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Renato S. Aguiar
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Department of Genetics, Ecology, and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil, 31270-901
| | - Rosana S. Rodrigues
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | | | - Andrea S. Souza
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| | - Daniel Martins-de-Souza
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil, 13083-862
| | - Fernanda G. De Felice
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
- Institute of Medical Biochemistry Leopoldo De Meis (IBqM), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil, 21941-902
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences & Department of Psychiatry, Queen's University, Kingston, Ontario, Canada, K7L 3N6
| | - Fernanda Tovar-Moll
- D’Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil, 22281-100
| |
Collapse
|
33
|
Gupta A, Comfort B, Young K, Montgomery R. A pilot study to assess blood-brain barrier permeability in long COVID. Brain Imaging Behav 2024; 18:830-834. [PMID: 38520594 DOI: 10.1007/s11682-024-00877-8] [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] [Accepted: 03/15/2024] [Indexed: 03/25/2024]
Abstract
The etiology of brain fog associated with long COVID is not clear. Based on some preliminary work, disruption of the blood-brain barrier has been hypothesized, but has not been tested in patients with long COVID. In this case-control pilot study, we evaluated blood-brain barrier permeability in patients with long COVID and subjective memory loss or brain fog. We used 99 m Technetium diethylenetriaminepentaacetic acid single-photon emission computed tomography (SPECT) to measure blood-brain barrier permeability and a telephone assessment (T-cog) to measure cognitive function. The blood-brain barrier permeability was quantified via SPECT standard uptake value (SUV). We assessed the blood-brain barrier permeability in 14 long COVID patients and 10 control participants without subjective cognitive impairment or brain fog. Participants in the two groups were similar in age. The long COVID group had more comorbidities compared to the control group. There was no difference in the SUVs in the long COVID (0.22 ± 0.12) vs the control (0.17 ± 0.04) group. There was no difference in the T-cog results in the two groups either. We found no evidence of a difference in blood-brain permeability in patients with long COVID when compared to controls without a known history of COVID-19 infection. Larger studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Aditi Gupta
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
- Department of Neurology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| | - Branden Comfort
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Kate Young
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| | - Robert Montgomery
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
34
|
Datta G, Rezagholizadeh N, Hasler WA, Khan N, Chen X. SLC38A9 regulates SARS-CoV-2 viral entry. iScience 2024; 27:110387. [PMID: 39071889 PMCID: PMC11277692 DOI: 10.1016/j.isci.2024.110387] [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: 03/04/2024] [Revised: 05/13/2024] [Accepted: 06/24/2024] [Indexed: 07/30/2024] Open
Abstract
SARS-CoV-2 viral entry into host cells depends on the cleavage of spike (S) protein into S1 and S2 proteins. Such proteolytic cleavage by furin results in the exposure of a multibasic motif on S1, which is critical for SARS-CoV-2 viral infection and transmission; however, how such a multibasic motif contributes to the infection of SARS-CoV-2 remains elusive. Here, we demonstrate that the multibasic motif on S1 is critical for its interaction with SLC38A9, an endolysosome-resident arginine sensor. SLC38A9 knockdown prevents S1-induced endolysosome de-acidification and blocks the S protein-mediated entry of pseudo-SARS-CoV-2 in Calu-3, U87MG, Caco-2, and A549 cells. Our findings provide a novel mechanism in regulating SARS-CoV-2 viral entry; S1 present in endolysosome lumen could interact with SLC38A9, which mediates S1-induced endolysosome de-acidification and dysfunction, facilitating the escape of SARS-CoV-2 from endolysosomes and enhancing viral entry.
Collapse
Affiliation(s)
- Gaurav Datta
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Neda Rezagholizadeh
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Wendie A. Hasler
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Nabab Khan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| |
Collapse
|
35
|
Krishna VD, Chang A, Korthas H, Var SR, Seelig DM, Low WC, Li L, Cheeran MCJ. Impact of age and sex on neuroinflammation following SARS-CoV-2 infection in a murine model. Front Microbiol 2024; 15:1404312. [PMID: 39077737 PMCID: PMC11284165 DOI: 10.3389/fmicb.2024.1404312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/24/2024] [Indexed: 07/31/2024] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the etiological agent of COVID-19, is known to infect people of all ages and both sexes. Senior populations have the greatest risk of severe COVID-19, and sexual dimorphism in clinical outcomes has been reported. Neurological symptoms are widely observed in COVID-19 patients, with many survivors exhibiting persistent neurological and cognitive impairment. The present study aims to investigate the impact of age and sex on the neuroinflammatory response to SARS-CoV-2 infection using a mouse model. Wild-type C57BL/6J mice were intranasally inoculated with SARS-CoV-2 lineage B.1.351, a variant known to infect mice. Older male mice exhibited a significantly greater weight loss and higher viral loads in the lung at 3 days post infection. Notably, no viral RNA was detected in the brains of infected mice. Nevertheless, expression of IL-6, TNF-α, and CCL-2 in the lung and brain increased with viral infection. RNA-seq transcriptomic analysis of brains showed that SARS-CoV-2 infection caused significant changes in gene expression profiles, implicating innate immunity, defense response to virus, and cerebrovascular and neuronal functions. These findings demonstrate that SARS-CoV-2 infection triggers a neuroinflammatory response, despite the lack of detectable virus in the brain. Aberrant activation of innate immune response, disruption of blood-brain barrier and endothelial cell integrity, and suppression of neuronal activity and axonogenesis underlie the impact of SARS-CoV-2 infection on the brain. Understanding the role of these affected pathways in SARS-CoV-2 pathogenesis helps identify appropriate points of therapeutic interventions to alleviate neurological dysfunction observed during COVID-19.
Collapse
Affiliation(s)
- Venkatramana D. Krishna
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, United States
| | - Allison Chang
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Holly Korthas
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Susanna R. Var
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Davis M. Seelig
- Comparative Pathology Shared Resource, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Walter C. Low
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Ling Li
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Maxim C. -J. Cheeran
- Department of Veterinary Population Medicine, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, United States
| |
Collapse
|
36
|
Vanderheiden A, Hill JD, Jiang X, Deppen B, Bamunuarachchi G, Soudani N, Joshi A, Cain MD, Boon ACM, Klein RS. Vaccination reduces central nervous system IL-1β and memory deficits after COVID-19 in mice. Nat Immunol 2024; 25:1158-1171. [PMID: 38902519 DOI: 10.1038/s41590-024-01868-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 05/13/2024] [Indexed: 06/22/2024]
Abstract
Up to 25% of individuals infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exhibit postacute cognitive sequelae. Although millions of cases of coronavirus disease 2019 (COVID-19)-mediated memory dysfunction are accumulating worldwide, the underlying mechanisms and how vaccination lowers risk are unknown. Interleukin-1 (IL-1), a key component of innate immune defense against SARS-CoV-2 infection, is elevated in the hippocampi of individuals with COVID-19. Here we show that intranasal infection of C57BL/6J mice with SARS-CoV-2 Beta variant leads to central nervous system infiltration of Ly6Chi monocytes and microglial activation. Accordingly, SARS-CoV-2, but not H1N1 influenza virus, increases levels of brain IL-1β and induces persistent IL-1R1-mediated loss of hippocampal neurogenesis, which promotes postacute cognitive deficits. Vaccination with a low dose of adenoviral-vectored spike protein prevents hippocampal production of IL-1β during breakthrough SARS-CoV-2 infection, loss of neurogenesis and subsequent memory deficits. Our study identifies IL-1β as one potential mechanism driving SARS-CoV-2-induced cognitive impairment in a new mouse model that is prevented by vaccination.
Collapse
Affiliation(s)
- Abigail Vanderheiden
- Center for Neuroimmunology and Neuroinfectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeremy D Hill
- Center for Neuroimmunology and Neuroinfectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoping Jiang
- Center for Neuroimmunology and Neuroinfectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ben Deppen
- Center for Neuroimmunology and Neuroinfectious Diseases, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Gayan Bamunuarachchi
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nadia Soudani
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Astha Joshi
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Matthew D Cain
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Adrianus C M Boon
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Robyn S Klein
- Schulich School of Medicine and Dentistry, Department of Microbiology and Immunology, Western University, London, Ontario, Canada.
- Schulich School of Medicine and Dentistry, Western Institute of Neuroscience, Western University, London, Ontario, Canada.
| |
Collapse
|
37
|
Trevino TN, Fogel AB, Otkiran G, Niladhuri SB, Sanborn MA, Class J, Almousawi AA, Vanhollebeke B, Tai LM, Rehman J, Richner JM, Lutz SE. Engineered Wnt7a ligands rescue blood-brain barrier and cognitive deficits in a COVID-19 mouse model. Brain 2024; 147:1636-1643. [PMID: 38306655 PMCID: PMC11068107 DOI: 10.1093/brain/awae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/18/2023] [Accepted: 01/19/2024] [Indexed: 02/04/2024] Open
Abstract
Respiratory infection with SARS-CoV-2 causes systemic vascular inflammation and cognitive impairment. We sought to identify the underlying mechanisms mediating cerebrovascular dysfunction and inflammation following mild respiratory SARS-CoV-2 infection. To this end, we performed unbiased transcriptional analysis to identify brain endothelial cell signalling pathways dysregulated by mouse adapted SARS-CoV-2 MA10 in aged immunocompetent C57Bl/6 mice in vivo. This analysis revealed significant suppression of Wnt/β-catenin signalling, a critical regulator of blood-brain barrier (BBB) integrity. We therefore hypothesized that enhancing cerebrovascular Wnt/β-catenin activity would offer protection against BBB permeability, neuroinflammation, and neurological signs in acute infection. Indeed, we found that delivery of cerebrovascular-targeted, engineered Wnt7a ligands protected BBB integrity, reduced T-cell infiltration of the brain, and reduced microglial activation in SARS-CoV-2 infection. Importantly, this strategy also mitigated SARS-CoV-2 induced deficits in the novel object recognition assay for learning and memory and the pole descent task for bradykinesia. These observations suggest that enhancement of Wnt/β-catenin signalling or its downstream effectors could be potential interventional strategies for restoring cognitive health following viral infections.
Collapse
Affiliation(s)
- Troy N Trevino
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | - Avital B Fogel
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | - Guliz Otkiran
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | - Seshadri B Niladhuri
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | - Mark A Sanborn
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | - Jacob Class
- Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | - Ali A Almousawi
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles (ULB), Gosselies B-6041, Belgium
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | - Jalees Rehman
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | - Justin M Richner
- Department of Microbiology and Immunology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| | - Sarah E Lutz
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
38
|
Chaganti J, Poudel G, Cysique LA, Dore GJ, Kelleher A, Matthews G, Darley D, Byrne A, Jakabek D, Zhang X, Lewis M, Jha N, Brew BJ. Blood brain barrier disruption and glutamatergic excitotoxicity in post-acute sequelae of SARS COV-2 infection cognitive impairment: potential biomarkers and a window into pathogenesis. Front Neurol 2024; 15:1350848. [PMID: 38756214 PMCID: PMC11097901 DOI: 10.3389/fneur.2024.1350848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/19/2024] [Indexed: 05/18/2024] Open
Abstract
Objective To investigate the association between blood-brain barrier permeability, brain metabolites, microstructural integrity of the white matter, and cognitive impairment (CI) in post-acute sequelae of SARS-COV-2 infection (PASC). Methods In this multimodal longitudinal MRI study 14 PASC participants with CI and 10 healthy controls were enrolled. All completed investigations at 3 months following acute infection (3 months ± 2 weeks SD), and 10 PASC participants completed at 12 months ± 2.22 SD weeks. The assessments included a standard neurological assessment, a cognitive screen using the brief CogState battery and multi-modal MRI derived metrics from Dynamic contrast enhanced (DCE) perfusion Imaging, Diffusion Tensor Imaging (DTI), and single voxel proton Magnetic Resonance Spectroscopy. These measures were compared between patients and controls and correlated with cognitive scores. Results At baseline, and relative to controls, PASC participants had higher K-Trans and Myo-inositol, and lower levels of Glutamate/Glutamine in the frontal white matter (FWM) (p < 0.01) as well as in brain stem (p < 0.05), and higher FA and lower MD in the FWM (p < 0.05). In PASC participants, FA and MD decreased in the FWM at 12 months compared to baseline (p < 0.05). K-Trans and metabolite concentrations did not change significantly over time. Neurocognitive scores did not correlation with the increased permeability (K trans). Interpretation PASC with CI is associated with BBB impairment, loss of WM integrity, and inflammation at 3 months which significantly but not uniformly improved at 12 months. The loss of WM integrity is possibly mediated by BBB impairment and associated glutamatergic excitotoxicity.
Collapse
Affiliation(s)
- Joga Chaganti
- Thomas Jefferson University, Philadelphia, PA, United States
| | - Govinda Poudel
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, VIC, Australia
| | - Lucette Adeline Cysique
- Department of Neurology and Immunology, Peter Duncan Neuroscience Unit, St Vincent’s Hospital, University of New South Wales, Darlinghurst, NSW, Australia
| | - Gregory J. Dore
- The Kirby Institute, Faculty of Medicine, University of New South Wales, Kensington, NSW, Australia
| | - Anthony Kelleher
- The Kirby Institute, Faculty of Medicine, University of New South Wales, Kensington, NSW, Australia
- St Vincent’s Hospital, University of NSW, Darlinghurst, NSW, Australia
| | - Gael Matthews
- The Kirby Institute, Faculty of Medicine, University of New South Wales, Kensington, NSW, Australia
| | - David Darley
- Department of Neurology and Immunology, Peter Duncan Neuroscience Unit, St Vincent’s Hospital, University of New South Wales, Darlinghurst, NSW, Australia
| | - Anthony Byrne
- St Vincent’s Hospital, University of NSW, Darlinghurst, NSW, Australia
| | - David Jakabek
- St Vincent’s Hospital, University of NSW, Darlinghurst, NSW, Australia
| | - Xin Zhang
- Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Marrissa Lewis
- St Vincent’s Hospital, University of NSW, Darlinghurst, NSW, Australia
| | - Nikhil Jha
- The Canberra Hospital, Canberra, ACT, Australia
| | - Bruce James Brew
- Department of Neurology and Immunology, Peter Duncan Neuroscience Unit, St Vincent’s Hospital, University of New South Wales, Darlinghurst, NSW, Australia
- University of Notre Dame, Sydney, NSW, Australia
| |
Collapse
|
39
|
Frontera JA, Betensky RA, Pirofski LA, Wisniewski T, Yoon H, Ortigoza MB. Trajectories of Inflammatory Markers and Post-COVID-19 Cognitive Symptoms: A Secondary Analysis of the CONTAIN COVID-19 Randomized Trial. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200227. [PMID: 38626359 PMCID: PMC11087048 DOI: 10.1212/nxi.0000000000200227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/24/2024] [Indexed: 04/18/2024]
Abstract
BACKGROUND AND OBJECTIVES Chronic systemic inflammation has been hypothesized to be a mechanistic factor leading to post-acute cognitive dysfunction after COVID-19. However, little data exist evaluating longitudinal inflammatory markers. METHODS We conducted a secondary analysis of data collected from the CONTAIN randomized trial of convalescent plasma in patients hospitalized for COVID-19, including patients who completed an 18-month assessment of cognitive symptoms and PROMIS Global Health questionnaires. Patients with pre-COVID-19 dementia/cognitive abnormalities were excluded. Trajectories of serum cytokine panels, D-dimer, fibrinogen, C-reactive peptide (CRP), ferritin, lactate dehydrogenase (LDH), and absolute neutrophil counts (ANCs) were evaluated over 18 months using repeated measures and Friedman nonparametric tests. The relationships between the area under the curve (AUC) for each inflammatory marker and 18-month cognitive and global health outcomes were assessed. RESULTS A total of 279 patients (N = 140 received plasma, N = 139 received placebo) were included. At 18 months, 76/279 (27%) reported cognitive abnormalities and 78/279 (28%) reported fair or poor overall health. PROMIS Global Mental and Physical Health T-scores were 0.5 standard deviations below normal in 24% and 51% of patients, respectively. Inflammatory marker levels declined significantly from hospitalization to 18 months for all markers (IL-2, IL-2R, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, IL-13, INFγ, TNFα, D-dimer, fibrinogen, ferritin, LDH, CRP, neutrophils; all p < 0.05), with the exception of IL-1β, which remained stable over time. There were no significant associations between the AUC for any inflammatory marker and 18-month cognitive symptoms, any neurologic symptom, or PROMIS Global Physical or Mental health T-scores. Receipt of convalescent plasma was not associated with any outcome measure. DISCUSSION At 18 months posthospitalization for COVID-19, cognitive abnormalities were reported in 27% of patients, and below average PROMIS Global Mental and Physical Health scores occurred in 24% and 51%, respectively. However, there were no associations with measured inflammatory markers, which decreased over time.
Collapse
Affiliation(s)
- Jennifer A Frontera
- From the Department of Neurology (J.A.F., T.W.), New York University Grossman School of Medicine; Department of Biostatistics (R.A.B.), NYU; Division of Infectious Disease (L.P.), Department of Medicine, Montefiore Medical Center; Department of Microbiology and Immunology; Division of Infectious Disease (H.Y.), Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx; and Division of Infectious Disease (M.B.O.), Department of Medicine, NYU Grossman School of Medicine, New York
| | - Rebecca A Betensky
- From the Department of Neurology (J.A.F., T.W.), New York University Grossman School of Medicine; Department of Biostatistics (R.A.B.), NYU; Division of Infectious Disease (L.P.), Department of Medicine, Montefiore Medical Center; Department of Microbiology and Immunology; Division of Infectious Disease (H.Y.), Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx; and Division of Infectious Disease (M.B.O.), Department of Medicine, NYU Grossman School of Medicine, New York
| | - Liise-Anne Pirofski
- From the Department of Neurology (J.A.F., T.W.), New York University Grossman School of Medicine; Department of Biostatistics (R.A.B.), NYU; Division of Infectious Disease (L.P.), Department of Medicine, Montefiore Medical Center; Department of Microbiology and Immunology; Division of Infectious Disease (H.Y.), Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx; and Division of Infectious Disease (M.B.O.), Department of Medicine, NYU Grossman School of Medicine, New York
| | - Thomas Wisniewski
- From the Department of Neurology (J.A.F., T.W.), New York University Grossman School of Medicine; Department of Biostatistics (R.A.B.), NYU; Division of Infectious Disease (L.P.), Department of Medicine, Montefiore Medical Center; Department of Microbiology and Immunology; Division of Infectious Disease (H.Y.), Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx; and Division of Infectious Disease (M.B.O.), Department of Medicine, NYU Grossman School of Medicine, New York
| | - Hyunah Yoon
- From the Department of Neurology (J.A.F., T.W.), New York University Grossman School of Medicine; Department of Biostatistics (R.A.B.), NYU; Division of Infectious Disease (L.P.), Department of Medicine, Montefiore Medical Center; Department of Microbiology and Immunology; Division of Infectious Disease (H.Y.), Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx; and Division of Infectious Disease (M.B.O.), Department of Medicine, NYU Grossman School of Medicine, New York
| | - Mila B Ortigoza
- From the Department of Neurology (J.A.F., T.W.), New York University Grossman School of Medicine; Department of Biostatistics (R.A.B.), NYU; Division of Infectious Disease (L.P.), Department of Medicine, Montefiore Medical Center; Department of Microbiology and Immunology; Division of Infectious Disease (H.Y.), Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx; and Division of Infectious Disease (M.B.O.), Department of Medicine, NYU Grossman School of Medicine, New York
| |
Collapse
|
40
|
Qiao H, Deng X, Qiu L, Qu Y, Chiu Y, Chen F, Xia S, Muenzel C, Ge T, Zhang Z, Song P, Bonnin A, Zhao Z, Yuan W. SARS-CoV-2 induces blood-brain barrier and choroid plexus barrier impairments and vascular inflammation in mice. J Med Virol 2024; 96:e29671. [PMID: 38747003 PMCID: PMC11446308 DOI: 10.1002/jmv.29671] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 07/18/2024]
Abstract
The coronavirus disease of 2019 (COVID-19) pandemic has led to more than 700 million confirmed cases and nearly 7 million deaths. Although severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) virus mainly infects the respiratory system, neurological complications are widely reported in both acute infection and long-COVID cases. Despite the success of vaccines and antiviral treatments, neuroinvasiveness of SARS-CoV-2 remains an important question, which is also centered on the mystery of whether the virus is capable of breaching the barriers into the central nervous system. By studying the K18-hACE2 infection model, we observed clear evidence of microvascular damage and breakdown of the blood-brain barrier (BBB). Mechanistically, SARS-CoV-2 infection caused pericyte damage, tight junction loss, endothelial activation and vascular inflammation, which together drive microvascular injury and BBB impairment. In addition, the blood-cerebrospinal fluid barrier at the choroid plexus was also impaired after infection. Therefore, cerebrovascular and choroid plexus dysfunctions are important aspects of COVID-19 and may contribute to neurological complications both acutely and in long COVID.
Collapse
Affiliation(s)
- Haowen Qiao
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Xiangxue Deng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Lingxi Qiu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Yafei Qu
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Yuanpu Chiu
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Feixiang Chen
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Shangzhou Xia
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, 90033, USA
| | - Cheyene Muenzel
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Tenghuan Ge
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Zixin Zhang
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Pengfei Song
- Department of Electrical and Computer Engineering, the Department of Bioengineering, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana–Champaign, Urbana, IL, USA
| | - Alexandre Bonnin
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| | - Zhen Zhao
- Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, 90033, USA
| | - Weiming Yuan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, USA
| |
Collapse
|
41
|
Ayyubova G, Fazal N. Beneficial versus Detrimental Effects of Complement-Microglial Interactions in Alzheimer's Disease. Brain Sci 2024; 14:434. [PMID: 38790413 PMCID: PMC11119363 DOI: 10.3390/brainsci14050434] [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: 04/04/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Research indicates that brain-region-specific synapse loss and dysfunction are early hallmarks and stronger neurobiological correlates of cognitive decline in Alzheimer's disease (AD) than amyloid plaque and neurofibrillary tangle counts or neuronal loss. Even though the precise mechanisms underlying increased synaptic pruning in AD are still unknown, it has been confirmed that dysregulation of the balance between complement activation and inhibition is a crucial driver of its pathology. The complement includes three distinct activation mechanisms, with the activation products C3a and C5a, potent inflammatory effectors, and a membrane attack complex (MAC) leading to cell lysis. Besides pro-inflammatory cytokines, the dysregulated complement proteins released by activated microglia bind to amyloid β at the synaptic regions and cause the microglia to engulf the synapses. Additionally, research indicating that microglia-removed synapses are not always degenerating and that suppression of synaptic engulfment can repair cognitive deficits points to an essential opportunity for intervention that can prevent the loss of intact synapses. In this study, we focus on the latest research on the role and mechanisms of complement-mediated microglial synaptic pruning at different stages of AD to find the right targets that could interfere with complement dysregulation and be relevant for therapeutic intervention at the early stages of the disease.
Collapse
Affiliation(s)
- Gunel Ayyubova
- Department of Cytology, Embryology and Histology, Azerbaijan Medical University, Baku 370022, Azerbaijan;
| | - Nadeem Fazal
- College of Health Sciences and Pharmacy, Chicago State University, Chicago, IL 60628, USA
| |
Collapse
|
42
|
Sun Z, Shi C, Jin L. Mechanisms by Which SARS-CoV-2 Invades and Damages the Central Nervous System: Apart from the Immune Response and Inflammatory Storm, What Else Do We Know? Viruses 2024; 16:663. [PMID: 38793545 PMCID: PMC11125732 DOI: 10.3390/v16050663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/29/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Initially reported as pneumonia of unknown origin, COVID-19 is increasingly being recognized for its impact on the nervous system, despite nervous system invasions being extremely rare. As a result, numerous studies have been conducted to elucidate the mechanisms of nervous system damage and propose appropriate coping strategies. This review summarizes the mechanisms by which SARS-CoV-2 invades and damages the central nervous system, with a specific focus on aspects apart from the immune response and inflammatory storm. The latest research findings on these mechanisms are presented, providing new insights for further in-depth research.
Collapse
Affiliation(s)
- Zihan Sun
- Qingdao Medical College, Qingdao University, Qingdao 266071, China
| | - Chunying Shi
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Lixin Jin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| |
Collapse
|
43
|
Sharma AA, Nenert R, Goodman AM, Szaflarski JP. Brain temperature and free water increases after mild COVID-19 infection. Sci Rep 2024; 14:7450. [PMID: 38548815 PMCID: PMC10978935 DOI: 10.1038/s41598-024-57561-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 03/19/2024] [Indexed: 04/01/2024] Open
Abstract
The pathophysiology underlying the post-acute sequelae of COVID-19 remains understudied and poorly understood, particularly in healthy adults with a history of mild infection. Chronic neuroinflammation may underlie these enduring symptoms, but studying neuroinflammatory phenomena in vivo is challenging, especially without a comparable pre-COVID-19 dataset. In this study, we present a unique dataset of 10 otherwise healthy individuals scanned before and after experiencing mild COVID-19. Two emerging MR-based methods were used to map pre- to post-COVID-19 brain temperature and free water changes. Post-COVID-19 brain temperature and free water increases, which are indirect biomarkers of neuroinflammation, were found in structures functionally associated with olfactory, cognitive, and memory processing. The largest pre- to post-COVID brain temperature increase was observed in the left olfactory tubercle (p = 0.007, 95% CI [0.48, 3.01]), with a mean increase of 1.75 °C. Notably, the olfactory tubercle is also the region of the primary olfactory cortex where participants with chronic olfactory dysfunction showed the most pronounced increases as compared to those without lingering olfactory dysfunction (adjusted pFDR = 0.0189, 95% CI [1.42, 5.27]). These preliminary insights suggest a potential link between neuroinflammation and chronic cognitive and olfactory dysfunction following mild COVID-19, although further investigations are needed to improve our understanding of what underlies these phenomena.
Collapse
Affiliation(s)
- Ayushe A Sharma
- Department of Neurology, UAB Epilepsy Center, University of Alabama at Birmingham (UAB), 1719 6th Avenue South, CIRC 312, Birmingham, AL, 35294-0021, USA.
- Department of Neurobiology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.
| | - Rodolphe Nenert
- Department of Neurology, UAB Epilepsy Center, University of Alabama at Birmingham (UAB), 1719 6th Avenue South, CIRC 312, Birmingham, AL, 35294-0021, USA
| | - Adam M Goodman
- Department of Neurology, UAB Epilepsy Center, University of Alabama at Birmingham (UAB), 1719 6th Avenue South, CIRC 312, Birmingham, AL, 35294-0021, USA
| | - Jerzy P Szaflarski
- Department of Neurology, UAB Epilepsy Center, University of Alabama at Birmingham (UAB), 1719 6th Avenue South, CIRC 312, Birmingham, AL, 35294-0021, USA.
- Department of Neurobiology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.
- Department of Neurosurgery, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.
- University of Alabama at Birmingham Epilepsy Center (UABEC), Birmingham, AL, USA.
| |
Collapse
|
44
|
Grant RA, Poor TA, Sichizya L, Diaz E, Bailey JI, Soni S, Senkow KJ, Pérez-Leonor XG, Abdala-Valencia H, Lu Z, Donnelly HK, Simons LM, Ozer EA, Tighe RM, Lomasney JW, Wunderink RG, Singer BD, Misharin AV, Budinger GS, for The Northwestern University Successful Clinical Response In Pneumonia Therapy (NU SCRIPT) Investigators. Prolonged exposure to lung-derived cytokines is associated with activation of microglia in patients with COVID-19. JCI Insight 2024; 9:e178859. [PMID: 38502186 PMCID: PMC11141878 DOI: 10.1172/jci.insight.178859] [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: 12/27/2023] [Accepted: 03/13/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUNDSurvivors of pneumonia, including SARS-CoV-2 pneumonia, are at increased risk for cognitive dysfunction and dementia. In rodent models, cognitive dysfunction following pneumonia has been linked to the systemic release of lung-derived pro-inflammatory cytokines. Microglia are poised to respond to inflammatory signals from the circulation, and their dysfunction has been linked to cognitive impairment in murine models of dementia and in humans.METHODSWe measured levels of 55 cytokines and chemokines in bronchoalveolar lavage fluid and plasma from 341 patients with respiratory failure and 13 healthy controls, including 93 unvaccinated patients with COVID-19 and 203 patients with other causes of pneumonia. We used flow cytometry to sort neuroimmune cells from postmortem brain tissue from 5 patients who died from COVID-19 and 3 patients who died from other causes for single-cell RNA-sequencing.RESULTSMicroglia from patients with COVID-19 exhibited a transcriptomic signature suggestive of their activation by circulating pro-inflammatory cytokines. Peak levels of pro-inflammatory cytokines were similar in patients with pneumonia irrespective of etiology, but cumulative cytokine exposure was higher in patients with COVID-19. Treatment with corticosteroids reduced expression of COVID-19-specific cytokines.CONCLUSIONProlonged lung inflammation results in sustained elevations in circulating cytokines in patients with SARS-CoV-2 pneumonia compared with those with pneumonia secondary to other pathogens. Microglia from patients with COVID-19 exhibit transcriptional responses to inflammatory cytokines. These findings support data from rodent models causally linking systemic inflammation with cognitive dysfunction in pneumonia and support further investigation into the role of microglia in pneumonia-related cognitive dysfunction.FUNDINGSCRIPT U19AI135964, UL1TR001422, P01AG049665, P01HL154998, R01HL149883, R01LM013337, R01HL153122, R01HL147290, R01HL147575, R01HL158139, R01ES034350, R01ES027574, I01CX001777, U01TR003528, R21AG075423, T32AG020506, F31AG071225, T32HL076139.
Collapse
Affiliation(s)
- Rogan A. Grant
- Division of Pulmonary and Critical Care Medicine, Department of Medicine; and
| | - Taylor A. Poor
- Division of Pulmonary and Critical Care Medicine, Department of Medicine; and
| | - Lango Sichizya
- Division of Pulmonary and Critical Care Medicine, Department of Medicine; and
| | - Estefani Diaz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine; and
| | - Joseph I. Bailey
- Division of Pulmonary and Critical Care Medicine, Department of Medicine; and
| | - Sahil Soni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine; and
| | - Karolina J. Senkow
- Division of Pulmonary and Critical Care Medicine, Department of Medicine; and
| | | | | | - Ziyan Lu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine; and
| | - Helen K. Donnelly
- Division of Pulmonary and Critical Care Medicine, Department of Medicine; and
| | - Lacy M. Simons
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Robert J. Havey, MD Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Egon A. Ozer
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Center for Pathogen Genomics and Microbial Evolution, Robert J. Havey, MD Institute for Global Health, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Robert M. Tighe
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University School of Medicine, Duke University, Durham, North Carolina, USA
| | | | | | - Benjamin D. Singer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine; and
- Department of Biochemistry and Molecular Genetics, and Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - G.R. Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine; and
| | | |
Collapse
|
45
|
Trevino TN, Almousawi AA, Robinson KF, Fogel AB, Class J, Minshall RD, Tai LM, Richner JM, Lutz SE. Caveolin-1 mediates blood-brain barrier permeability, neuroinflammation, and cognitive impairment in SARS-CoV-2 infection. J Neuroimmunol 2024; 388:578309. [PMID: 38335781 PMCID: PMC11212674 DOI: 10.1016/j.jneuroim.2024.578309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024]
Abstract
Blood-brain barrier (BBB) permeability can cause neuroinflammation and cognitive impairment. Caveolin-1 (Cav-1) critically regulates BBB permeability, but its influence on the BBB and consequent neurological outcomes in respiratory viral infections is unknown. We used Cav-1-deficient mice with genetically encoded fluorescent endothelial tight junctions to determine how Cav-1 influences BBB permeability, neuroinflammation, and cognitive impairment following respiratory infection with mouse adapted (MA10) SARS-CoV-2 as a model for COVID-19. We found that SARS-CoV-2 infection increased brain endothelial Cav-1 and increased transcellular BBB permeability to albumin, decreased paracellular BBB Claudin-5 tight junctions, and caused T lymphocyte infiltration in the hippocampus, a region important for learning and memory. Concordantly, we observed learning and memory deficits in SARS-CoV-2 infected mice. Importantly, genetic deficiency in Cav-1 attenuated transcellular BBB permeability and paracellular BBB tight junction losses, T lymphocyte infiltration, and gliosis induced by SARS-CoV-2 infection. Moreover, Cav-1 KO mice were protected from the learning and memory deficits caused by SARS-CoV-2 infection. These results establish the contribution of Cav-1 to BBB permeability and behavioral dysfunction induced by SARS-CoV-2 neuroinflammation.
Collapse
Affiliation(s)
- Troy N Trevino
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago College of Medicine, USA
| | - Ali A Almousawi
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago College of Medicine, USA
| | - KaReisha F Robinson
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago College of Medicine, USA
| | - Avital B Fogel
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago College of Medicine, USA
| | - Jake Class
- Departments of Microbiology and Immunology, University of Illinois at Chicago College of Medicine, USA
| | - Richard D Minshall
- Departments of Anesthesiology, and Pharmacology and Regenerative Medicine, University of Illinois at Chicago College of Medicine, USA
| | - Leon M Tai
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago College of Medicine, USA
| | - Justin M Richner
- Departments of Microbiology and Immunology, University of Illinois at Chicago College of Medicine, USA
| | - Sarah E Lutz
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago College of Medicine, USA.
| |
Collapse
|
46
|
Owens CD, Bonin Pinto C, Detwiler S, Olay L, Pinaffi-Langley ACDC, Mukli P, Peterfi A, Szarvas Z, James JA, Galvan V, Tarantini S, Csiszar A, Ungvari Z, Kirkpatrick AC, Prodan CI, Yabluchanskiy A. Neurovascular coupling impairment as a mechanism for cognitive deficits in COVID-19. Brain Commun 2024; 6:fcae080. [PMID: 38495306 PMCID: PMC10943572 DOI: 10.1093/braincomms/fcae080] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/08/2024] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
Components that comprise our brain parenchymal and cerebrovascular structures provide a homeostatic environment for proper neuronal function to ensure normal cognition. Cerebral insults (e.g. ischaemia, microbleeds and infection) alter cellular structures and physiologic processes within the neurovascular unit and contribute to cognitive dysfunction. COVID-19 has posed significant complications during acute and convalescent stages in multiple organ systems, including the brain. Cognitive impairment is a prevalent complication in COVID-19 patients, irrespective of severity of acute SARS-CoV-2 infection. Moreover, overwhelming evidence from in vitro, preclinical and clinical studies has reported SARS-CoV-2-induced pathologies in components of the neurovascular unit that are associated with cognitive impairment. Neurovascular unit disruption alters the neurovascular coupling response, a critical mechanism that regulates cerebromicrovascular blood flow to meet the energetic demands of locally active neurons. Normal cognitive processing is achieved through the neurovascular coupling response and involves the coordinated action of brain parenchymal cells (i.e. neurons and glia) and cerebrovascular cell types (i.e. endothelia, smooth muscle cells and pericytes). However, current work on COVID-19-induced cognitive impairment has yet to investigate disruption of neurovascular coupling as a causal factor. Hence, in this review, we aim to describe SARS-CoV-2's effects on the neurovascular unit and how they can impact neurovascular coupling and contribute to cognitive decline in acute and convalescent stages of the disease. Additionally, we explore potential therapeutic interventions to mitigate COVID-19-induced cognitive impairment. Given the great impact of cognitive impairment associated with COVID-19 on both individuals and public health, the necessity for a coordinated effort from fundamental scientific research to clinical application becomes imperative. This integrated endeavour is crucial for mitigating the cognitive deficits induced by COVID-19 and its subsequent burden in this especially vulnerable population.
Collapse
Affiliation(s)
- Cameron D Owens
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Camila Bonin Pinto
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Sam Detwiler
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Lauren Olay
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Ana Clara da C Pinaffi-Langley
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Peter Mukli
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
| | - Anna Peterfi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
| | - Zsofia Szarvas
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
| | - Judith A James
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Veronica Galvan
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - Stefano Tarantini
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Anna Csiszar
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
| | - Zoltan Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Angelia C Kirkpatrick
- Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Andriy Yabluchanskiy
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73117, USA
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, 1089, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
47
|
van der Knaap N, Ariës MJH, van der Horst ICC, Jansen JFA. On the merits and potential of advanced neuroimaging techniques in COVID-19: A scoping review. Neuroimage Clin 2024; 42:103589. [PMID: 38461701 PMCID: PMC10938171 DOI: 10.1016/j.nicl.2024.103589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 03/12/2024]
Abstract
Many Coronavirus Disease 2019 (COVID-19) patients are suffering from long-term neuropsychological sequelae. These patients may benefit from a better understanding of the underlying neuropathophysiological mechanisms and identification of potential biomarkers and treatment targets. Structural clinical neuroimaging techniques have limited ability to visualize subtle cerebral abnormalities and to investigate brain function. This scoping review assesses the merits and potential of advanced neuroimaging techniques in COVID-19 using literature including advanced neuroimaging or postmortem analyses in adult COVID-19 patients published from the start of the pandemic until December 2023. Findings were summarized according to distinct categories of reported cerebral abnormalities revealed by different imaging techniques. Although no unified COVID-19-specific pattern could be subtracted, a broad range of cerebral abnormalities were revealed by advanced neuroimaging (likely attributable to hypoxic, vascular, and inflammatory pathology), even in absence of structural clinical imaging findings. These abnormalities are validated by postmortem examinations. This scoping review emphasizes the added value of advanced neuroimaging compared to structural clinical imaging and highlights implications for brain functioning and long-term consequences in COVID-19.
Collapse
Affiliation(s)
- Noa van der Knaap
- Department of Intensive Care Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Research Institute of Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Marcel J H Ariës
- Department of Intensive Care Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Research Institute of Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Iwan C C van der Horst
- Department of Intensive Care Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Research Institute of Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
48
|
Greene C, Connolly R, Brennan D, Laffan A, O'Keeffe E, Zaporojan L, O'Callaghan J, Thomson B, Connolly E, Argue R, Meaney JFM, Martin-Loeches I, Long A, Cheallaigh CN, Conlon N, Doherty CP, Campbell M. Blood-brain barrier disruption and sustained systemic inflammation in individuals with long COVID-associated cognitive impairment. Nat Neurosci 2024; 27:421-432. [PMID: 38388736 PMCID: PMC10917679 DOI: 10.1038/s41593-024-01576-9] [Citation(s) in RCA: 138] [Impact Index Per Article: 138.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/09/2024] [Indexed: 02/24/2024]
Abstract
Vascular disruption has been implicated in coronavirus disease 2019 (COVID-19) pathogenesis and may predispose to the neurological sequelae associated with long COVID, yet it is unclear how blood-brain barrier (BBB) function is affected in these conditions. Here we show that BBB disruption is evident during acute infection and in patients with long COVID with cognitive impairment, commonly referred to as brain fog. Using dynamic contrast-enhanced magnetic resonance imaging, we show BBB disruption in patients with long COVID-associated brain fog. Transcriptomic analysis of peripheral blood mononuclear cells revealed dysregulation of the coagulation system and a dampened adaptive immune response in individuals with brain fog. Accordingly, peripheral blood mononuclear cells showed increased adhesion to human brain endothelial cells in vitro, while exposure of brain endothelial cells to serum from patients with long COVID induced expression of inflammatory markers. Together, our data suggest that sustained systemic inflammation and persistent localized BBB dysfunction is a key feature of long COVID-associated brain fog.
Collapse
Affiliation(s)
- Chris Greene
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Ruairi Connolly
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland
| | - Declan Brennan
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland
| | - Aoife Laffan
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland
| | - Eoin O'Keeffe
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Lilia Zaporojan
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland
| | | | - Bennett Thomson
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Emma Connolly
- The Irish Longitudinal Study on Ageing, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Ruth Argue
- Clinical Research Facility, St James's Hospital, Dublin, Ireland
| | - James F M Meaney
- Thomas Mitchell Centre for Advanced Medical Imaging (CAMI), St. James's Hospital & Trinity College Dublin, Dublin, Ireland
| | - Ignacio Martin-Loeches
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization, Trinity Centre for Health Sciences, St James's University Hospital, Dublin, Ireland
| | - Aideen Long
- Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin, Ireland
| | - Cliona Ni Cheallaigh
- Trinity Translational Medicine Institute, Trinity College Dublin, St James's Hospital, Dublin, Ireland
- Department of Immunology, St James's Hospital, Dublin, Ireland
| | - Niall Conlon
- Department of Immunology, St James's Hospital, Dublin, Ireland
- St James's Hospital, Tallaght University Hospital, Trinity College Dublin Allied Researchers (STTAR) Bioresource, Trinity College Dublin, Dublin, Ireland
| | - Colin P Doherty
- Department of Neurology, Health Care Centre, St James's Hospital, Dublin, Ireland.
- Academic Unit of Neurology, Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland.
| | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland.
- FutureNeuro, Science Foundation Ireland Research Centre for Chronic and Rare Neurological Diseases, Royal College of Surgeons in Ireland, University of Medicine and Health Sciences, Dublin, Ireland.
| |
Collapse
|
49
|
Nath A. Brainstem Encephalitis as a Cause of Sudden Infant Death Syndrome. JAMA Neurol 2024; 81:231-232. [PMID: 38285466 DOI: 10.1001/jamaneurol.2023.5384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Affiliation(s)
- Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
50
|
Jung JM, Gruber A, Heseltine P, Rajamani K, Ameriso SF, Fisher MJ. New Directions in Infection-Associated Ischemic Stroke. J Clin Neurol 2024; 20:140-152. [PMID: 38330416 PMCID: PMC10921058 DOI: 10.3988/jcn.2023.0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 09/06/2023] [Accepted: 11/12/2023] [Indexed: 02/10/2024] Open
Abstract
The relationship between infections and stroke has not been fully characterized, probably delaying the development of specific treatments. This narrative review addresses mechanisms of stroke linked to infections, including hypercoagulability, endothelial dysfunction, vasculitis, and impaired thrombolysis. SARS-CoV-2, the virus that causes COVID-19, may promote the development of stroke, which may represent its most severe neurological complication. The development of specific therapies for infection-associated stroke remains a profound challenge. Perhaps the most important remaining issue is the distinction between infections that trigger a stroke versus infections that are truly incidental. This distinction likely requires the establishment of appropriate biomarkers, candidates of which are elevated levels of fibrin D-dimer and anticardiolipin/antiphospholipid antibodies. These candidate biomarkers might have potential use in identifying pathogenic infections preceding stroke, which is a precursor to establishing specific therapies for this syndrome.
Collapse
Affiliation(s)
- Jin-Man Jung
- Department of Neurology, Korea University Ansan Hospital, Korea University College of Medicine, Ansan, Korea; Korea University Zebrafish, Translational Medical Research Center, Ansan, Korea
| | | | - Peter Heseltine
- Division of Infectious Diseases, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Kumar Rajamani
- Department of Neurology, Wayne State University-Detroit Medical Center, Detroit, MI, USA
| | - Sebastián F Ameriso
- Division of Vascular Neurology, Department of Neurology, Fleni, Autonomous City of Buenos Aires, Argentina
| | - Mark J Fisher
- Department of Neurology, University of California Irvine Medical Center, Orange, CA, USA.
| |
Collapse
|