201
|
Gaire BP, Koronyo Y, Fuchs DT, Shi H, Rentsendorj A, Danziger R, Vit JP, Mirzaei N, Doustar J, Sheyn J, Hampel H, Vergallo A, Davis MR, Jallow O, Baldacci F, Verdooner SR, Barron E, Mirzaei M, Gupta VK, Graham SL, Tayebi M, Carare RO, Sadun AA, Miller CA, Dumitrascu OM, Lahiri S, Gao L, Black KL, Koronyo-Hamaoui M. Alzheimer's disease pathophysiology in the Retina. Prog Retin Eye Res 2024; 101:101273. [PMID: 38759947 PMCID: PMC11285518 DOI: 10.1016/j.preteyeres.2024.101273] [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: 02/11/2023] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
The retina is an emerging CNS target for potential noninvasive diagnosis and tracking of Alzheimer's disease (AD). Studies have identified the pathological hallmarks of AD, including amyloid β-protein (Aβ) deposits and abnormal tau protein isoforms, in the retinas of AD patients and animal models. Moreover, structural and functional vascular abnormalities such as reduced blood flow, vascular Aβ deposition, and blood-retinal barrier damage, along with inflammation and neurodegeneration, have been described in retinas of patients with mild cognitive impairment and AD dementia. Histological, biochemical, and clinical studies have demonstrated that the nature and severity of AD pathologies in the retina and brain correspond. Proteomics analysis revealed a similar pattern of dysregulated proteins and biological pathways in the retina and brain of AD patients, with enhanced inflammatory and neurodegenerative processes, impaired oxidative-phosphorylation, and mitochondrial dysfunction. Notably, investigational imaging technologies can now detect AD-specific amyloid deposits, as well as vasculopathy and neurodegeneration in the retina of living AD patients, suggesting alterations at different disease stages and links to brain pathology. Current and exploratory ophthalmic imaging modalities, such as optical coherence tomography (OCT), OCT-angiography, confocal scanning laser ophthalmoscopy, and hyperspectral imaging, may offer promise in the clinical assessment of AD. However, further research is needed to deepen our understanding of AD's impact on the retina and its progression. To advance this field, future studies require replication in larger and diverse cohorts with confirmed AD biomarkers and standardized retinal imaging techniques. This will validate potential retinal biomarkers for AD, aiding in early screening and monitoring.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ron Danziger
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jean-Philippe Vit
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jonah Doustar
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Miyah R Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ousman Jallow
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Filippo Baldacci
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | | | - Ernesto Barron
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Vivek K Gupta
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Stuart L Graham
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia; Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Roxana O Carare
- Department of Clinical Neuroanatomy, University of Southampton, Southampton, UK
| | - Alfredo A Sadun
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Carol A Miller
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Shouri Lahiri
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Liang Gao
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
202
|
Costas-Carrera A, Sánchez-Rodríguez MM, Ojeda A, Rodríguez-Rey MA, Martín-Villalba I, Primé-Tous M, Valdesoiro-Pulido F, Segú X, Borras R, Clougher D, Peri JM, Vieta E. Neuropsychological functioning and its correlates at 1 year follow-up of severe COVID-19. Psychogeriatrics 2024; 24:765-777. [PMID: 38576072 DOI: 10.1111/psyg.13113] [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: 09/12/2023] [Revised: 01/23/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Short-term cognitive impairment is associated with SARS-CoV-2 infection but the long-term impact is yet to be examined in detail. We aim to study the evolution of these symptoms in severe COVID-19 patients admitted to the intensive care unit (ICU) between April and December 2020 1 year after hospital discharge and to analyze its clinical correlates. METHOD A total of 58 patients agreed to participate in the 6 months follow-up and 30 at 1 year after hospital discharge. Demographic, clinical and laboratory data were collected and a comprehensive neuropsychological battery including validated tests for the main cognitive domains was administered. To test the magnitude of neurocognitive sequelae, two standard deviations below normative group were considered. To compare the neuropsychological performance at 6 and 12 months follow-up we used repeated measures tests. Finally, regression analyses were performed to test the main effects of medical and psychological factors on multiple cognition. RESULTS Almost half of the sample continued to have impaired performance on neuropsychological tests at 12 months follow-up. In comparison with the results obtained at 6 months, significant improvements were found in immediate recall (d = 0.49), delayed recall (d = 0.45), and inhibitory control (d = 0.53). Medical variables predicted cognitive performance at 6 months but not at 12 months follow-up, while anxiety and depression predicted cognitive deficits in the long-term. CONCLUSIONS A generalised improvement was observed in severe COVID-19 patients at follow-up. This improvement was particularly notable in verbal memory and executive functioning. However, a considerable proportion of the sample continued to present deficits at 1 year follow-up.
Collapse
Affiliation(s)
| | | | - Antonio Ojeda
- Anaesthesiology Reanimation and Pain Therapy, Hospital Clinic, Barcelona, Spain
| | | | | | | | | | - Xavier Segú
- Neuroscience Institute, Hospital Clinic, Barcelona, Spain
| | - Roger Borras
- Institute of Biomedical Research Agusti Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Derek Clougher
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | | | - Eduard Vieta
- Neuroscience Institute, Hospital Clinic, Barcelona, Spain
- Institute of Biomedical Research Agusti Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
203
|
Mandolfo O, Parker H, Aguado È, Ishikawa Learmonth Y, Liao AY, O'Leary C, Ellison S, Forte G, Taylor J, Wood S, Searle R, Holley RJ, Boutin H, Bigger BW. Systemic immune challenge exacerbates neurodegeneration in a model of neurological lysosomal disease. EMBO Mol Med 2024; 16:1579-1602. [PMID: 38890537 PMCID: PMC11251277 DOI: 10.1038/s44321-024-00092-4] [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: 07/07/2023] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024] Open
Abstract
Mucopolysaccharidosis type IIIA (MPS IIIA) is a rare paediatric lysosomal storage disorder, caused by the progressive accumulation of heparan sulphate, resulting in neurocognitive decline and behavioural abnormalities. Anecdotal reports from paediatricians indicate a more severe neurodegeneration in MPS IIIA patients, following infection, suggesting inflammation as a potential driver of neuropathology. To test this hypothesis, we performed acute studies in which WT and MPS IIIA mice were challenged with the TLR3-dependent viral mimetic poly(I:C). The challenge with an acute high poly(I:C) dose exacerbated systemic and brain cytokine expression, especially IL-1β in the hippocampus. This was accompanied by an increase in caspase-1 activity within the brain of MPS IIIA mice with concomitant loss of hippocampal GFAP and NeuN expression. Similar levels of cell damage, together with exacerbation of gliosis, were also observed in MPS IIIA mice following low chronic poly(I:C) dosing. While further investigation is warranted to fully understand the extent of IL-1β involvement in MPS IIIA exacerbated neurodegeneration, our data robustly reinforces our previous findings, indicating IL-1β as a pivotal catalyst for neuropathological processes in MPS IIIA.
Collapse
Affiliation(s)
- Oriana Mandolfo
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Helen Parker
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Èlia Aguado
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Yuko Ishikawa Learmonth
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Ai Yin Liao
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Claire O'Leary
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Stuart Ellison
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Gabriella Forte
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Jessica Taylor
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Shaun Wood
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Rachel Searle
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Rebecca J Holley
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK
| | - Hervé Boutin
- Division of Neuroscience & Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
- INSERM, UMR 1253, iBrain, Université de Tours, Tours, France
| | - Brian W Bigger
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 3.721 Stopford Building, Manchester, UK.
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
204
|
Schreiber CS, Wiesweg I, Stanelle-Bertram S, Beck S, Kouassi NM, Schaumburg B, Gabriel G, Richter F, Käufer C. Sex-specific biphasic alpha-synuclein response and alterations of interneurons in a COVID-19 hamster model. EBioMedicine 2024; 105:105191. [PMID: 38865747 PMCID: PMC11293593 DOI: 10.1016/j.ebiom.2024.105191] [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: 12/13/2023] [Revised: 05/02/2024] [Accepted: 05/25/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) frequently leads to neurological complications after recovery from acute infection, with higher prevalence in women. However, mechanisms by which SARS-CoV-2 disrupts brain function remain unclear and treatment strategies are lacking. We previously demonstrated neuroinflammation in the olfactory bulb of intranasally infected hamsters, followed by alpha-synuclein and tau accumulation in cortex, thus mirroring pathogenesis of neurodegenerative diseases such as Parkinson's or Alzheimer's disease. METHODS To uncover the sex-specific spatiotemporal profiles of neuroinflammation and neuronal dysfunction following intranasal SARS-CoV-2 infection, we quantified microglia cell density, alpha-synuclein immunoreactivity and inhibitory interneurons in cortical regions, limbic system and basal ganglia at acute and late post-recovery time points. FINDINGS Unexpectedly, microglia cell density and alpha-synuclein immunoreactivity decreased at 6 days post-infection, then rebounded to overt accumulation at 21 days post-infection. This biphasic response was most pronounced in amygdala and striatum, regions affected early in Parkinson's disease. Several brain regions showed altered densities of parvalbumin and calretinin interneurons which are involved in cognition and motor control. Of note, females appeared more affected. INTERPRETATION Our results demonstrate that SARS-CoV-2 profoundly disrupts brain homeostasis without neuroinvasion, via neuroinflammatory and protein regulation mechanisms that persist beyond viral clearance. The regional patterns and sex differences are in line with neurological deficits observed after SARS-CoV-2 infection. FUNDING Federal Ministry of Health, Germany (BMG; ZMV I 1-2520COR501 to G.G.), Federal Ministry of Education and Research, Germany (BMBF; 03COV06B to G.G.), Ministry of Science and Culture of Lower Saxony in Germany (14-76403-184, to G.G. and F.R.).
Collapse
Affiliation(s)
- Cara Sophie Schreiber
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover (ZSN), Germany
| | - Ivo Wiesweg
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine Hannover, Hannover, Germany
| | | | - Sebastian Beck
- Department for Viral Zoonoses-One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Nancy Mounogou Kouassi
- Department for Viral Zoonoses-One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Berfin Schaumburg
- Department for Viral Zoonoses-One Health, Leibniz Institute of Virology, Hamburg, Germany
| | - Gülsah Gabriel
- Department for Viral Zoonoses-One Health, Leibniz Institute of Virology, Hamburg, Germany; Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover (ZSN), Germany.
| | - Christopher Käufer
- Department of Pharmacology, Toxicology, and Pharmacy; University of Veterinary Medicine Hannover, Hannover, Germany; Center for Systems Neuroscience Hannover (ZSN), Germany.
| |
Collapse
|
205
|
Radmanesh D, Powell E, Trinh H. Too tired to think: Relationship between post-COVID-19 fatigue and cognition in a veteran sample. Neuropsychol Rehabil 2024; 34:823-844. [PMID: 37584412 DOI: 10.1080/09602011.2023.2244159] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 07/26/2023] [Indexed: 08/17/2023]
Abstract
COVID-19 survivors often endorse persistent physical and neuropsychiatric problems following disease recovery, a phenomenon described as "long COVID." Research exploring long-COVID continues to evolve in large-scale studies but remains limited among smaller populations (e.g., veterans). We explored the relationship between persistent post-COVID-19 fatigue and cognition among a sample of 246 veterans who voluntarily enrolled in a COVID-19 Convalescence Programme and completed a mental health evaluation of post-illness mood (depression, anxiety, PTSD), cognition (subjective complaints, Modified Telephone Interview for Cognitive Status [TICS-M] performance), fatigue, pain, and sleep. In concert with our hypotheses, subjective cognitive complaints are not significantly correlated with TICS-M performance, but rather are strongly correlated with long-COVID fatigue. Although cognitive changes are common post-COVID complaints, these are likely better predicted by other factors, (e.g., fatigue, mood, pain, and sleep disruption). Furthermore, comorbid mood, sleep, and pain complaints appeared to mediate the relationship between subjective cognitive complaints and fatigue. Limitations to this study included use of retrospective chart review data, limited access to pre-disease data for comparison, and lack of healthy controls. Clinicians should consider the impact of modifiable conditions associated with cognitive and functional decline, as these conditions may be targets for interdisciplinary treatment in a long-COVID veteran population.
Collapse
Affiliation(s)
| | - Eric Powell
- South Texas Veterans Health Care System, San Antonio, TX, USA
| | - Hanh Trinh
- South Texas Veterans Health Care System, San Antonio, TX, USA
| |
Collapse
|
206
|
Haverty R, McCormack J, Evans C, Purves K, O'Reilly S, Gautier V, Rochfort K, Fabre A, Fletcher NF. SARS-CoV-2 infects neurons, astrocytes, choroid plexus epithelial cells and pericytes of the human central nervous system in vitro. J Gen Virol 2024; 105:002009. [PMID: 38995681 PMCID: PMC11317966 DOI: 10.1099/jgv.0.002009] [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: 12/12/2023] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is associated with neurological sequelae including haemorrhage, thrombosis and ischaemic necrosis and encephalitis. However, the mechanism by which this occurs is unclear. Neurological disease associated with COVID-19 has been proposed to occur following direct infection of the central nervous system and/or indirectly by local or systemic immune activation. We evaluated the expression of angiotensin-converting enzyme-2 and transmembrane protease, serine 2 (TMPRSS2) in brain tissue from five healthy human donors and observed low-level expression of these proteins in cells morphologically consistent with astrocytes, neurons and choroidal ependymal cells within the frontal cortex and medulla oblongata. Primary human astrocytes, neurons, choroid plexus epithelial cells and pericytes supported productive SARS-CoV-2 infection with ancestral, Alpha, Delta and Omicron variants. Infected cells supported the full viral life cycle, releasing infectious virus particles. In contrast, primary brain microvascular endothelial cells and microglia were refractory to SARS-CoV-2 infection. These data support a model whereby SARS-CoV-2 can infect human brain cells, and the mechanism of viral entry warrants further investigation.
Collapse
Affiliation(s)
- Ruth Haverty
- Veterinary Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland
| | - Janet McCormack
- Research Pathology Core Facility, Conway Institute of Biomedical and Biomolecular Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Christopher Evans
- Veterinary Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland
| | - Kevin Purves
- Veterinary Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sophie O'Reilly
- Centre for Experimental Pathogen Host Research, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Virginie Gautier
- Centre for Experimental Pathogen Host Research, School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomedical and Biomolecular Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Keith Rochfort
- School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Aurelie Fabre
- Research Pathology Core Facility, Conway Institute of Biomedical and Biomolecular Research, University College Dublin, Belfield, Dublin 4, Ireland
- Department of Histopathology, St. Vincent’s University Hospital, Dublin 4, Ireland
| | - Nicola F. Fletcher
- Veterinary Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland
- Conway Institute of Biomedical and Biomolecular Research, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
207
|
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
|
208
|
Theofilis P, Oikonomou E, Vasileiadou M, Tousoulis D. A Narrative Review on Prolonged Neuropsychiatric Consequences of COVID-19: A Serious Concern. HEART AND MIND 2024; 8:177-183. [DOI: 10.4103/hm.hm-d-24-00019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 05/09/2024] [Indexed: 03/03/2025] Open
Abstract
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is characterized by prolonged, postacute sequelae of COVID-19 (PASC). Marked by persistent or new-onset symptoms within 3 months following COVID-19 recovery, PASC significantly affects a diverse spectrum of survivors. Beyond cardiovascular implications, neuropsychiatric PASC demonstrates prolonged symptoms with diverse phenotypic profiles affecting memory, attention, and mood. The pathophysiologic basis points to SARS-CoV-2’s neurotropism, instigating inflammatory responses in the central nervous system. A comprehensive multimodal assessment, integrating psychological evaluations, fluid examinations, neurophysiology, and imaging, emerges as a critical diagnostic approach. Managing neuropsychiatric PASC necessitates personalized interventions to enhance resilience and coping mechanisms, emphasizing the role of physical fitness, creative engagement, and social support in mitigating its impact on identity and well-being. In addition, early initiation of cognitive rehabilitation and cognitive behavioral therapy is proposed to address symptom chronicity, emotional distress, and cognitive dysfunction, enhancing the quality of life. The urgency for targeted interventions, early neuropsychological support, and ongoing research to comprehensively address the multifaceted neuropsychiatric effects of COVID-19 is underscored in this review. Collaborative efforts involving health-care professionals, support networks, and affected individuals are imperative to navigate the evolving landscape of PASC and its persistent neuropsychiatric implications.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1Department of Cardiology, General Hospital of Athens “Ippokrateio”, Athens, Greece
| | - Evangelos Oikonomou
- 3Department of Cardiology, Regional Chest Disease Hospital “Sotiria”, Athens, Greece
| | | | - Dimitris Tousoulis
- 1Department of Cardiology, General Hospital of Athens “Ippokrateio”, Athens, Greece
| |
Collapse
|
209
|
Cahan J, Finley JCA, Cotton E, Orban ZS, Jimenez M, Weintraub S, Sorets T, Koralnik IJ. Cognitive functioning in patients with neuro-PASC: the role of fatigue, mood, and hospitalization status. Front Neurol 2024; 15:1401796. [PMID: 38994492 PMCID: PMC11236596 DOI: 10.3389/fneur.2024.1401796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024] Open
Abstract
This study sought to characterize cognitive functioning in patients with neurological post-acute sequelae of SARS-CoV-2 infection (Neuro-PASC) and investigate the association of subjective and objective functioning along with other relevant factors with prior hospitalization for COVID-19. Participants were 106 adult outpatients with Neuro-PASC referred for abbreviated neuropsychological assessment after scoring worse than one standard deviation below the mean on cognitive screening. Of these patients, 23 had been hospitalized and 83 had not been hospitalized for COVID-19. Subjective cognitive impairment was evaluated with the self-report cognition subscale from the Patient-Reported Outcome Measurement Information System. Objective cognitive performance was assessed using a composite score derived from multiple standardized cognitive measures. Other relevant factors, including fatigue and depression/mood symptoms, were assessed via the Patient-Reported Outcome Measurement Information System. Subjective cognitive impairment measures exceeded the minimal difficulties noted on objective tests and were associated with depression/mood symptoms as well as fatigue. However, fatigue independently explained the most variance (17.51%) in patients' subjective cognitive ratings. When adjusting for fatigue and time since onset of COVID-19 symptoms, neither objective nor subjective impairment were associated with prior hospitalization for COVID-19. Findings suggest that abbreviated neuropsychological assessment may not reveal objective difficulties beyond initial cognitive screening in patients with Neuro-PASC. However, subjective cognitive concerns may persist irrespective of hospitalization status, and are likely influenced by fatigue and depression/mood symptoms. The impact of concomitant management of fatigue and mood in patients with Neuro-PASC who report cognitive concerns deserve further study.
Collapse
Affiliation(s)
- Joshua Cahan
- Northwestern Medicine, Davee Department of Neurology, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Chicago, IL, United States
| | | | - Erica Cotton
- Northwestern Medicine, Davee Department of Neurology, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Northwestern Medicine, Department of Psychiatry and Behavioral Sciences, Chicago, IL, United States
| | - Zachary S. Orban
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Millenia Jimenez
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Sandra Weintraub
- Northwestern Medicine, Davee Department of Neurology, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Chicago, IL, United States
- Northwestern Medicine, Department of Psychiatry and Behavioral Sciences, Chicago, IL, United States
| | - Tali Sorets
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Igor J. Koralnik
- Northwestern Medicine, Davee Department of Neurology, Chicago, IL, United States
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
210
|
Du Y, Li C, Zhao W, Li J, Zhao L, Guo H, Jiang Y, Liu WV, Zeng S, Zhang H, Guo H, Ouyang X, Liu J. Multimodal neuroimaging exploration of the mechanisms of sleep quality deterioration after SARS-CoV-2 Omicron infection. BMC Med 2024; 22:271. [PMID: 38926881 PMCID: PMC11210028 DOI: 10.1186/s12916-024-03487-9] [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: 02/28/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND To evaluate the neurological alterations induced by Omicron infection, to compare brain changes in chronic insomnia with those in exacerbated chronic insomnia in Omicron patients, and to examine individuals without insomnia alongside those with new-onset insomnia. METHODS In this study, a total of 135 participants were recruited between January 11 and May 4, 2023, including 26 patients with chronic insomnia without exacerbation, 24 patients with chronic insomnia with exacerbation, 40 patients with no sleep disorder, and 30 patients with new-onset insomnia after infection with Omicron (a total of 120 participants with different sleep statuses after infection), as well as 15 healthy controls who were never infected with Omicron. Neuropsychiatric data, clinical symptoms, and multimodal magnetic resonance imaging data were collected. The gray matter thickness and T1, T2, proton density, and perivascular space values were analyzed. Associations between changes in multimodal magnetic resonance imaging findings and neuropsychiatric data were evaluated with correlation analyses. RESULTS Compared with healthy controls, gray matter thickness changes were similar in the patients who have and do not have a history of chronic insomnia groups after infection, including an increase in cortical thickness near the parietal lobe and a reduction in cortical thickness in the frontal, occipital, and medial brain regions. Analyses showed a reduced gray matter thickness in patients with chronic insomnia compared with those with an aggravation of chronic insomnia post-Omicron infection, and a reduction was found in the right medial orbitofrontal region (mean [SD], 2.38 [0.17] vs. 2.67 [0.29] mm; P < 0.001). In the subgroups of Omicron patients experiencing sleep deterioration, patients with a history of chronic insomnia whose insomnia symptoms worsened after infection displayed heightened medial orbitofrontal cortical thickness and increased proton density values in various brain regions. Conversely, patients with good sleep quality who experienced a new onset of insomnia after infection exhibited reduced cortical thickness in pericalcarine regions and decreased proton density values. In new-onset insomnia patients post-Omicron infection, the thickness in the right pericalcarine was negatively correlated with the Self-rating Anxiety Scale (r = - 0.538, P = 0.002, PFDR = 0.004) and Self-rating Depression Scale (r = - 0.406, P = 0.026, PFDR = 0.026) scores. CONCLUSIONS These findings help us understand the pathophysiological mechanisms involved when Omicron invades the nervous system and induces various forms of insomnia after infection. In the future, we will continue to pay attention to the dynamic changes in the brain related to insomnia caused by Omicron infection.
Collapse
Affiliation(s)
- Yanyao Du
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, Hunan Province, 410011, China
| | - Cong Li
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, Hunan Province, 410011, China
| | - Wei Zhao
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, Hunan Province, 410011, China
- Clinical Research Center for Medical Imaging in Hunan Province, Changsha, Hunan, 410011, China
- Department of Radiology Quality Control Center, Changsha, Hunan, 410011, China
| | - Jinyue Li
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, 410011, China
| | - Linlin Zhao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, 410011, China
| | - Huili Guo
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, 410011, China
| | - Yingjia Jiang
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, Hunan Province, 410011, China
| | | | - Song Zeng
- MR Product, GE Healthcare, Guangzhou, 510000, China
| | - Huiting Zhang
- MR Research Collaboration, Siemens Healthineers, Wuhan, 430000, China
| | - Hu Guo
- MR Application, Siemens Healthineers, Guangzhou, 510000, China
| | - Xuan Ouyang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, 410011, China.
| | - Jun Liu
- Department of Radiology, Second Xiangya Hospital of Central South University, Changsha, Hunan Province, 410011, China.
- Clinical Research Center for Medical Imaging in Hunan Province, Changsha, Hunan, 410011, China.
- Department of Radiology Quality Control Center, Changsha, Hunan, 410011, China.
| |
Collapse
|
211
|
Sun D, Shi Z, Chen H, Du Q, Zhang Y, Wang R, Kong L, Luo W, Lang Y, Wang X, Zhou H. COVID-19 susceptibility, hospitalization and severity and the risk of brain cortical structure: a Mendelian randomization study. QJM 2024; 117:413-421. [PMID: 38195890 DOI: 10.1093/qjmed/hcad291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 12/18/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND Observational studies have reported structural changes in the brains of patients with coronavirus disease 2019 (COVID-19); it remains unclear whether these associations are causal. AIM We evaluated the causal effects of COVID-19 susceptibility, hospitalization and severity on cortical structures. DESIGN Mendelian randomization (MR) study. METHODS Data on the different COVID-19 phenotypes were obtained from the latest large-scale genome-wide association study (R7) of the COVID-19 Host Genetics Initiative. Brain structure data, including cortical thickness (TH) and surface area (SA), were obtained from the ENIGMA Consortium. Additionally, we employed the round 5 dataset released in January 2021 as the validation cohort. The inverse-variance weighted (IVW) method was used as the primary analysis in MR. Sensitivity analyses were conducted to evaluate heterogeneity and pleiotropy. We performed enrichment analysis on the MR analyses that passed the sensitivity analysis filtering. RESULTS After IVW and sensitivity analyses, we observed causal associations between COVID-19 susceptibility and rostral middle frontal SAw (P = 0.0308, β = -39.1236), cuneus THw (P = 0.0170, β = -0.0121), medial orbitofrontal THw (P = 0.0002, β = 0.0225), postcentral THw (P = 0.0217, β = -0.0106), temporal pole THw (P = 0.0077, β = 0.0359), medial orbitofrontal SAnw (P = 0.0106, β = -24.0397), medial orbitofrontal THnw (P = 0.0007, β = 0.0232), paracentral SAnw (P = 0.0483, β = -20.1442), rostral middle frontal SAnw (P = 0.0368, β = -81.9719) and temporal pole THnw (P = 0.0429, β = 0.0353). COVID-19 hospitalization had causal effects on medial orbitofrontal THw (P = 0.0053, β = 0.0063), postcentral THw (P = 0.0143, β = -0.0042), entorhinal THnw (P = 0.0142, β = 0.0142), medial orbitofrontal THnw (P = 0.0147, β = 0.0065) and paracentral SAnw (P = 0.0119, β = -7.9970). COVID-19 severity had causal effects on rostral middle frontal SAw (P = 0.0122, β = -11.8296), medial orbitofrontal THw (P = 0.0155, β = 0.0038), superior parietal THw (P = 0.0291, β = -0.0021), lingual SAnw (P = 0.0202, β = -11.5270), medial orbitofrontal THnw (P = 0.0290, β = 0.0039), paracentral SAnw (P = 0.0180, β = -5.7744) and pars triangularis SAnw (P = 0.0151, β = -5.4520). CONCLUSION Our MR results demonstrate a causal relationship between different COVID-19 phenotypes and cortical structures.
Collapse
Affiliation(s)
- D Sun
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Z Shi
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - H Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Q Du
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Y Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - R Wang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - L Kong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - W Luo
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Y Lang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - X Wang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - H Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
212
|
Santos Guedes de Sa K, Silva J, Bayarri-Olmos R, Brinda R, Alec Rath Constable R, Colom Diaz PA, Kwon DI, Rodrigues G, Wenxue L, Baker C, Bhattacharjee B, Wood J, Tabacof L, Liu Y, Putrino D, Horvath TL, Iwasaki A. A causal link between autoantibodies and neurological symptoms in long COVID. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.18.24309100. [PMID: 38947091 PMCID: PMC11213106 DOI: 10.1101/2024.06.18.24309100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Acute SARS-CoV-2 infection triggers the generation of diverse and functional autoantibodies (AABs), even after mild cases. Persistently elevated autoantibodies have been found in some individuals with long COVID (LC). Using a >21,000 human protein array, we identified diverse AAB targets in LC patients that correlated with their symptoms. Elevated AABs to proteins in the nervous system were found in LC patients with neurocognitive and neurological symptoms. Purified Immunoglobulin G (IgG) samples from these individuals reacted with human pons tissue and were cross-reactive with mouse sciatic nerves, spinal cord, and meninges. Antibody reactivity to sciatic nerves and meninges correlated with patient-reported headache and disorientation. Passive transfer of IgG from patients to mice led to increased sensitivity and pain, mirroring patient-reported symptoms. Similarly, mice injected with IgG showed loss of balance and coordination, reflecting donor-reported dizziness. Our findings suggest that targeting AABs could benefit some LC patients.
Collapse
Affiliation(s)
- Keyla Santos Guedes de Sa
- Department of Immunobiology, Yale School of Medicine, Center for Infection and Immunity, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Julio Silva
- Department of Immunobiology, Yale School of Medicine, Center for Infection and Immunity, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Rafael Bayarri-Olmos
- Department of Immunobiology, Yale School of Medicine, Center for Infection and Immunity, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Ryan Brinda
- Department of Immunobiology, Yale School of Medicine, Center for Infection and Immunity, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Robert Alec Rath Constable
- Department of Immunobiology, Yale School of Medicine, Center for Infection and Immunity, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Patricia A. Colom Diaz
- Department of Immunobiology, Yale School of Medicine, Center for Infection and Immunity, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Dong-il Kwon
- Department of Immunobiology, Yale School of Medicine, Center for Infection and Immunity, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Gisele Rodrigues
- Department of Immunobiology, Yale School of Medicine, Center for Infection and Immunity, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Li Wenxue
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - Christopher Baker
- Department of Immunobiology, Yale School of Medicine, Center for Infection and Immunity, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Bornali Bhattacharjee
- Department of Immunobiology, Yale School of Medicine, Center for Infection and Immunity, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Jamie Wood
- Cohen Center for Recovery from Complex Chronic Illness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura Tabacof
- Cohen Center for Recovery from Complex Chronic Illness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yansheng Liu
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, USA
| | - David Putrino
- Cohen Center for Recovery from Complex Chronic Illness, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tamas L. Horvath
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520
| | - Akiko Iwasaki
- Department of Immunobiology, Yale School of Medicine, Center for Infection and Immunity, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
213
|
Chau SWH, Chue TM, Chan RNY, Lai YL, Wong PWC, Li SX, Liu Y, Chan JWY, Chan PKS, Lai CKC, Leung TWH, Wing YK. Chronic post-COVID neuropsychiatric symptoms persisting beyond one year from infection: a case-control study and network analysis. Transl Psychiatry 2024; 14:261. [PMID: 38898009 PMCID: PMC11187160 DOI: 10.1038/s41398-024-02978-w] [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: 10/31/2023] [Revised: 05/18/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024] Open
Abstract
Our study aims to delineate the phenotypes of chronic neuropsychiatric symptoms among adult subjects recovering from their first COVID that occurred more than one year ago. We also aim to explore the clinical and socioeconomic risk factors of having a high loading of chronic neuropsychiatric symptoms. We recruited a post-COVID group who suffered from their first pre-Omicron COVID more than a year ago, and a control group who had never had COVID. The subjects completed app-based questionnaires on demographic, socioeconomic and health status, a COVID symptoms checklist, mental and sleep health measures, and neurocognitive tests. The post-COVID group has a statistically significantly higher level of fatigue compared to the control group (p < 0.001). Among the post-COVID group, the lack of any COVID vaccination before the first COVID and a higher level of material deprivation before the COVID pandemic predicts a higher load of chronic post-COVID neuropsychiatric symptoms. Partial correlation network analysis suggests that the chronic post-COVID neuropsychiatric symptoms can be clustered into two major (cognitive complaints -fatigue and anxiety-depression) and one minor (headache-dizziness) cluster. A higher level of material deprivation predicts a higher number of symptoms in both major clusters, but the lack of any COVID vaccination before the first COVID only predicts a higher number of symptoms in the cognitive complaints-fatigue cluster. Our result suggests heterogeneity among chronic post-COVID neuropsychiatric symptoms, which are associated with the complex interplay of biological and socioeconomic factors.
Collapse
Affiliation(s)
- Steven Wai Ho Chau
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| | - Timothy Mitchell Chue
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Rachel Ngan Yin Chan
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yee Lok Lai
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Paul W C Wong
- Department of Social Work and Social Administration, Faculty of Social Science, The University of Hong Kong, Hong Kong, China
| | - Shirley Xin Li
- Department of Psychology, Faculty of Social Science, The University of Hong Kong, Hong Kong, China
| | - Yaping Liu
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Center for Sleep and Circadian Medicine, The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Joey Wing Yan Chan
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Paul Kay-Sheung Chan
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Christopher K C Lai
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Thomas W H Leung
- Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yun Kwok Wing
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Li Chiu Kong Family Sleep Assessment Unit, Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
214
|
Navolokin N, Adushkina V, Zlatogorskaya D, Telnova V, Evsiukova A, Vodovozova E, Eroshova A, Dosadina E, Diduk S, Semyachkina-Glushkovskaya O. Promising Strategies to Reduce the SARS-CoV-2 Amyloid Deposition in the Brain and Prevent COVID-19-Exacerbated Dementia and Alzheimer's Disease. Pharmaceuticals (Basel) 2024; 17:788. [PMID: 38931455 PMCID: PMC11206883 DOI: 10.3390/ph17060788] [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: 04/29/2024] [Revised: 06/02/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
The COVID-19 pandemic, caused by infection with the SARS-CoV-2 virus, is associated with cognitive impairment and Alzheimer's disease (AD) progression. Once it enters the brain, the SARS-CoV-2 virus stimulates accumulation of amyloids in the brain that are highly toxic to neural cells. These amyloids may trigger neurological symptoms in COVID-19. The meningeal lymphatic vessels (MLVs) play an important role in removal of toxins and mediate viral drainage from the brain. MLVs are considered a promising target to prevent COVID-19-exacerbated dementia. However, there are limited methods for augmentation of MLV function. This review highlights new discoveries in the field of COVID-19-mediated amyloid accumulation in the brain associated with the neurological symptoms and the development of promising strategies to stimulate clearance of amyloids from the brain through lymphatic and other pathways. These strategies are based on innovative methods of treating brain dysfunction induced by COVID-19 infection, including the use of photobiomodulation, plasmalogens, and medicinal herbs, which offer hope for addressing the challenges posed by the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Nikita Navolokin
- Department of Pathological Anatomy, Saratov Medical State University, Bolshaya Kazachaya Str. 112, 410012 Saratov, Russia;
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (V.A.); (D.Z.); (V.T.); (A.E.)
| | - Viktoria Adushkina
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (V.A.); (D.Z.); (V.T.); (A.E.)
| | - Daria Zlatogorskaya
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (V.A.); (D.Z.); (V.T.); (A.E.)
| | - Valeria Telnova
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (V.A.); (D.Z.); (V.T.); (A.E.)
| | - Arina Evsiukova
- Department of Biology, Saratov State University, Astrakhanskaya 82, 410012 Saratov, Russia; (V.A.); (D.Z.); (V.T.); (A.E.)
| | - Elena Vodovozova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Miklukho-Maklaya 16/10, 117997 Moscow, Russia;
| | - Anna Eroshova
- Department of Biotechnology, Leeners LLC, Nagornyi Proezd 3a, 117105 Moscow, Russia; (A.E.); (E.D.); (S.D.)
| | - Elina Dosadina
- Department of Biotechnology, Leeners LLC, Nagornyi Proezd 3a, 117105 Moscow, Russia; (A.E.); (E.D.); (S.D.)
| | - Sergey Diduk
- Department of Biotechnology, Leeners LLC, Nagornyi Proezd 3a, 117105 Moscow, Russia; (A.E.); (E.D.); (S.D.)
- Research Institute of Carcinogenesis of the N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of Russia, Kashirskoe Shosse 24, 115522 Moscow, Russia
| | | |
Collapse
|
215
|
Liu X, Liu L, Liu C. Summary of the effect of an exercise intervention on elderly with mild cognitive impairment: A systematic review and meta-analysis. Medicine (Baltimore) 2024; 103:e38025. [PMID: 38875404 PMCID: PMC11175880 DOI: 10.1097/md.0000000000038025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 02/01/2024] [Accepted: 04/05/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Many randomized controlled trials have demonstrated that exercise benefits cognitive function in patients with mild cognitive impairment (MCI), but less attention has been paid to the development of exercise programs in this population. OBJECTIVE This study aimed to assess the effect of exercise intervention for elderly with MCI and provide the most effective exercise intervention plan. METHODS We searched 4 international databases (PubMed, EMBASE, Web of Science, Cochrane Library) and 4 Chinese databases (Chinese National Knowledge Infrastructure, VIP database and Wanfang database) for studies on exercises associated with MCI up to September 25, 2022. The resulting standardized mean differences (SMD) and 95% confidence intervals were statistically analyzed using Review Manager 5.3 software. RESULTS A total of 20 RCTs were comprised in this meta-analysis, including 1393 participants. The results of the meta-analysis revealed that exercise had positive effects on cognitive function in elderly with MCI and was statistically significant (SMD = 1.25, 95%CI: [0.88,1.62], P < .00001). Subgroup analysis showed that the most significant factor was the Peterson 2004 criteria, multi-exercise, 35 to 50 minutes/times, <3 times/3 to 5 times per week, >16 weeks and medium intensity. CONCLUSION Exercise intervention can significantly alleviate cognition in elderly with MCI. The best exercise program for exercise intervention for MCI is: multi-exercise, 35 to 50 minutes/times, 3 to 5 times/week, and exercise cycle for more than 16 weeks with medium intensity, has the best effect. Plus, more RCTs with larger sample sizes will be required in the future to demonstrate the link between exercise duration, intensity, and cognitive function.
Collapse
Affiliation(s)
- Xiaotang Liu
- College of Physical Education, Shanghai Normal University, Shanghai, China
| | - Lanjuan Liu
- College of Physical Education, Shanghai Normal University, Shanghai, China
| | - Cheng Liu
- Department of Sports, Donghua University, Shanghai, China
| |
Collapse
|
216
|
Austin TA, Thomas ML, Lu M, Hodges CB, Darowski ES, Bergmans R, Parr S, Pickell D, Catazaro M, Lantrip C, Twamley EW. Meta-analysis of Cognitive Function Following Non-severe SARS-CoV-2 Infection. Neuropsychol Rev 2024:10.1007/s11065-024-09642-6. [PMID: 38862725 DOI: 10.1007/s11065-024-09642-6] [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: 06/02/2022] [Accepted: 04/22/2024] [Indexed: 06/13/2024]
Abstract
To effectively diagnose and treat subjective cognitive symptoms in post-acute sequalae of COVID-19 (PASC), it is important to understand objective cognitive impairment across the range of acute COVID-19 severity. Despite the importance of this area of research, to our knowledge, there are no current meta-analyses of objective cognitive functioning following non-severe initial SARS-CoV-2 infection. The aim of this meta-analysis is to describe objective cognitive impairment in individuals with non-severe (mild or moderate) SARS-CoV-2 cases in the post-acute stage of infection. This meta-analysis was pre-registered with Prospero (CRD42021293124) and utilized the PRISMA checklist for reporting guidelines, with screening conducted by at least two independent reviewers for all aspects of the screening and data extraction process. Fifty-nine articles (total participants = 22,060) with three types of study designs met our full criteria. Individuals with non-severe (mild/moderate) initial SARS-CoV-2 infection demonstrated worse objective cognitive performance compared to healthy comparison participants. However, those with mild (nonhospitalized) initial SARS-CoV-2 infections had better objective cognitive performance than those with moderate (hospitalized but not requiring ICU care) or severe (hospitalized with ICU care) initial SARS-CoV-2 infections. For studies that used normative data comparisons instead of healthy comparison participants, there was a small and nearly significant effect when compared to normative data. There were high levels of heterogeneity (88.6 to 97.3%), likely reflecting small sample sizes and variations in primary study methodology. Individuals who have recovered from non-severe cases of SARS-CoV-2 infections may be at risk for cognitive decline or impairment and may benefit from cognitive health interventions.
Collapse
Affiliation(s)
- Tara A Austin
- The VISN 17 Center of Excellence for Research on Returning War Veterans, 4800 Memorial Drive, Waco, TX, 76711, USA.
- Center of Excellence for Stress and Mental Health, San Diego Healthcare System, San Diego, CA, USA.
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA.
| | - Michael L Thomas
- Department of Psychology, Colorado State University, Colorado Springs, Fort Collins, USA
| | - Min Lu
- University of Miami, Miami, FL, USA
| | - Cooper B Hodges
- Department of Psychology, Brigham Young University, Provo, UT, USA
| | | | - Rachel Bergmans
- Chronic Pain and Fatigue Research Center, Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Sarah Parr
- The VISN 17 Center of Excellence for Research on Returning War Veterans, 4800 Memorial Drive, Waco, TX, 76711, USA
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, USA
| | - Delaney Pickell
- Center of Excellence for Stress and Mental Health, San Diego Healthcare System, San Diego, CA, USA
| | - Mikayla Catazaro
- The VISN 17 Center of Excellence for Research on Returning War Veterans, 4800 Memorial Drive, Waco, TX, 76711, USA
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, USA
| | - Crystal Lantrip
- The VISN 17 Center of Excellence for Research on Returning War Veterans, 4800 Memorial Drive, Waco, TX, 76711, USA
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, USA
| | - Elizabeth W Twamley
- Center of Excellence for Stress and Mental Health, San Diego Healthcare System, San Diego, CA, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| |
Collapse
|
217
|
Arrigoni A, Previtali M, Bosticardo S, Pezzetti G, Poloni S, Capelli S, Napolitano A, Remuzzi A, Zangari R, Lorini FL, Sessa M, Daducci A, Caroli A, Gerevini S. Brain microstructure and connectivity in COVID-19 patients with olfactory or cognitive impairment. Neuroimage Clin 2024; 43:103631. [PMID: 38878591 PMCID: PMC11225694 DOI: 10.1016/j.nicl.2024.103631] [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: 03/05/2024] [Revised: 05/31/2024] [Accepted: 06/11/2024] [Indexed: 07/08/2024]
Abstract
INTRODUCTION The COVID-19 pandemic has affected millions worldwide, causing mortality and multi-organ morbidity. Neurological complications have been recognized. This study aimed to assess brain structural, microstructural, and connectivity alterations in patients with COVID-19-related olfactory or cognitive impairment using post-acute (time from onset: 264[208-313] days) multi-directional diffusion-weighted MRI (DW-MRI). METHODS The study included 16 COVID-19 patients with cognitive impairment (COVID-CM), 35 COVID-19 patients with olfactory disorder (COVID-OD), and 14 controls. A state-of-the-art processing pipeline was developed for DW-MRI pre-processing, mean diffusivity and fractional anisotropy computation, fiber density and cross-section analysis, and tractography of white-matter bundles. Brain parcellation required for probing network connectivity, region-specific microstructure and volume, and cortical thickness was based on T1-weighted scans and anatomical atlases. RESULTS Compared to controls, COVID-CM patients showed overall gray matter atrophy (age and sex corrected p = 0.004), and both COVID-19 patient groups showed regional atrophy and cortical thinning. Both groups presented an increase in gray matter mean diffusivity (corrected p = 0.001), decrease in white matter fiber density and cross-section (corrected p < 0.05), , and COVID-CM patients also displayed an overall increased diffusivity (p = 0.022) and decreased anisotropy (corrected p = 0.038) in white matter. Graph-based analysis revealed reduced network modularity, with an extensive pattern of connectivity increase, in conjunction with a localized reduction in a few connections, mainly located in the left hemisphere. The left cingulate, anterior cingulate, and insula were primarily involved. CONCLUSION Expanding upon previous findings, this study further investigated significant alterations in brain morphology, microstructure, and connectivity in COVID-19 patients with olfactory or cognitive disfunction. These findings suggest underlying neurodegeneration, neuroinflammation, and concomitant compensatory mechanisms. Future longitudinal studies are required to monitor the alterations over time and assess their transient or permanent nature.
Collapse
Affiliation(s)
- Alberto Arrigoni
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Ranica, Italy.
| | - Mattia Previtali
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Ranica, Italy
| | - Sara Bosticardo
- Department of Computer Science, University of Verona, Italy; Translational Imaging in Neurology (ThINK), Department of Biomedical Engineering, Faculty of Medicine, Basel, Switzerland.
| | - Giulio Pezzetti
- Department of Neuroradiology, ASST Papa Giovanni XXIII, Bergamo, Italy.
| | - Sofia Poloni
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Ranica, Italy.
| | - Serena Capelli
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Ranica, Italy.
| | - Angela Napolitano
- Department of Neuroradiology, ASST Papa Giovanni XXIII, Bergamo, Italy.
| | - Andrea Remuzzi
- Department of Management Information and Production Engineering, University of Bergamo, Dalmine, Italy.
| | - Rosalia Zangari
- FROM Research Foundation, ASST Papa Giovanni XXIII, Bergamo, Italy.
| | - Ferdinando Luca Lorini
- Department of Emergency and Critical Care Area, ASST Papa Giovanni XXIII, Bergamo, Italy.
| | - Maria Sessa
- Department of Neurology, ASST Papa Giovanni XXIII, Bergamo, Italy.
| | | | - Anna Caroli
- Department of Biomedical Engineering, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Ranica, Italy.
| | | |
Collapse
|
218
|
Petersen M, Becker B, Schell M, Mayer C, Naegele FL, Petersen E, Twerenbold R, Thomalla G, Cheng B, Betz C, Hoffmann AS. Reduced olfactory bulb volume accompanies olfactory dysfunction after mild SARS-CoV-2 infection. Sci Rep 2024; 14:13396. [PMID: 38862636 PMCID: PMC11167024 DOI: 10.1038/s41598-024-64367-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 06/06/2024] [Indexed: 06/13/2024] Open
Abstract
Despite its high prevalence, the determinants of smelling impairment in COVID-19 remain not fully understood. In this work, we aimed to examine the association between olfactory bulb volume and the clinical trajectory of COVID-19-related smelling impairment in a large-scale magnetic resonance imaging (MRI) analysis. Data of non-vaccinated COVID-19 convalescents recruited within the framework of the prospective Hamburg City Health Study COVID Program between March and December 2020 were analyzed. At baseline, 233 participants underwent MRI and neuropsychological testing as well as a structured questionnaire for olfactory function. Between March and April 2022, olfactory function was assessed at follow-up including quantitative olfactometric testing with Sniffin' Sticks. This study included 233 individuals recovered from mainly mild to moderate SARS-CoV-2 infections. Longitudinal assessment demonstrated a declining prevalence of self-reported olfactory dysfunction from 67.1% at acute infection, 21.0% at baseline examination and 17.5% at follow-up. Participants with post-acute self-reported olfactory dysfunction had a significantly lower olfactory bulb volume at baseline than normally smelling individuals. Olfactory bulb volume at baseline predicted olfactometric scores at follow-up. Performance in neuropsychological testing was not significantly associated with the olfactory bulb volume. Our work demonstrates an association of long-term self-reported smelling dysfunction and olfactory bulb integrity in a sample of individuals recovered from mainly mild to moderate COVID-19. Collectively, our results highlight olfactory bulb volume as a surrogate marker that may inform diagnosis and guide rehabilitation strategies in COVID-19.
Collapse
Affiliation(s)
- Marvin Petersen
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Benjamin Becker
- Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Schell
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Carola Mayer
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Felix L Naegele
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Elina Petersen
- Population Health Research Department, University Heart and Vascular Center, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany
| | - Raphael Twerenbold
- Population Health Research Department, University Heart and Vascular Center, Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Luebeck, Hamburg, Germany
- University Center of Cardiovascular Science, University Heart and Vascular Center, Hamburg, Germany
| | - Götz Thomalla
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Bastian Cheng
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Christian Betz
- Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna S Hoffmann
- Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
219
|
Gusev E, Sarapultsev A. Exploring the Pathophysiology of Long COVID: The Central Role of Low-Grade Inflammation and Multisystem Involvement. Int J Mol Sci 2024; 25:6389. [PMID: 38928096 PMCID: PMC11204317 DOI: 10.3390/ijms25126389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Long COVID (LC), also referred to as Post COVID-19 Condition, Post-Acute Sequelae of SARS-CoV-2 Infection (PASC), and other terms, represents a complex multisystem disease persisting after the acute phase of COVID-19. Characterized by a myriad of symptoms across different organ systems, LC presents significant diagnostic and management challenges. Central to the disorder is the role of low-grade inflammation, a non-classical inflammatory response that contributes to the chronicity and diversity of symptoms observed. This review explores the pathophysiological underpinnings of LC, emphasizing the importance of low-grade inflammation as a core component. By delineating the pathogenetic relationships and clinical manifestations of LC, this article highlights the necessity for an integrated approach that employs both personalized medicine and standardized protocols aimed at mitigating long-term consequences. The insights gained not only enhance our understanding of LC but also inform the development of therapeutic strategies that could be applicable to other chronic conditions with similar pathophysiological features.
Collapse
Affiliation(s)
| | - Alexey Sarapultsev
- Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Science, 620049 Ekaterinburg, Russia;
| |
Collapse
|
220
|
Rudroff T. Long COVID in Brain Health Research: A Call to Action. Brain Sci 2024; 14:587. [PMID: 38928587 PMCID: PMC11201626 DOI: 10.3390/brainsci14060587] [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: 05/22/2024] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
The COVID-19 pandemic has brought attention to the long-term consequences of the virus, particularly the persistent symptoms that characterize long COVID. This syndrome, which can last for months after the initial infection, includes a range of neurological and neuropsychiatric manifestations that have significant implications for brain health and dementia research. This review explores the current understanding of long COVID's cognitive, neurological, and psychiatric symptoms and their potential impact on brain stimulation and neuroimaging studies. It argues that researchers must adapt their study designs and screening processes to account for the confounding effects of long COVID and ensure the accuracy and reliability of their findings. To advance the understanding of this condition and its long-term effects on brain health, the review proposes a series of strategies, including the development of standardized screening tools, the investigation of underlying mechanisms, and the identification of risk factors and protective factors. It also emphasizes the importance of collaborative research efforts and international data sharing platforms in accelerating the pace of discovery and developing targeted interventions for individuals with long COVID. As the prevalence of this condition continues to grow, it is imperative that the neuroscience community comes together to address this challenge and support those affected by long COVID.
Collapse
Affiliation(s)
- Thorsten Rudroff
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA 52242, USA;
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| |
Collapse
|
221
|
Kettunen P, Koistinaho J, Rolova T. Contribution of CNS and extra-CNS infections to neurodegeneration: a narrative review. J Neuroinflammation 2024; 21:152. [PMID: 38845026 PMCID: PMC11157808 DOI: 10.1186/s12974-024-03139-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
Central nervous system infections have been suggested as a possible cause for neurodegenerative diseases, particularly sporadic cases. They trigger neuroinflammation which is considered integrally involved in neurodegenerative processes. In this review, we will look at data linking a variety of viral, bacterial, fungal, and protozoan infections to Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis and unspecified dementia. This narrative review aims to bring together a broad range of data currently supporting the involvement of central nervous system infections in the development of neurodegenerative diseases. The idea that no single pathogen or pathogen group is responsible for neurodegenerative diseases will be discussed. Instead, we suggest that a wide range of susceptibility factors may make individuals differentially vulnerable to different infectious pathogens and subsequent pathologies.
Collapse
Affiliation(s)
- Pinja Kettunen
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Jari Koistinaho
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.
| | - Taisia Rolova
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
222
|
Monteiro S, Dessenne C, Perquin M. Long COVID cognitive sequelae 6 months postinfection and beyond: a scoping review protocol. BMJ Open 2024; 14:e084798. [PMID: 38844389 PMCID: PMC11163643 DOI: 10.1136/bmjopen-2024-084798] [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: 01/28/2024] [Accepted: 05/15/2024] [Indexed: 06/12/2024] Open
Abstract
INTRODUCTION The novel and expanding field of long COVID research has undergone diverse methodological approaches in recent years. This protocol lays out the methodological approach, which aims at identifying nuances in current research. It underscores the necessity for a more precise understanding of prolonged cognitive sequelae and their relation to initial disease severity. The findings will add valuable insights for the development of targeted rehabilitation, healthcare interventions and thereby aid patients, clinicians, policymakers and researchers. Our upcoming research is introduced here. METHODS AND ANALYSIS To map current research in the field, a scoping review will be conducted and documented in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses for Scoping Review Extension standards. A systematic search of scientific databases (PubMed, EMBASE), presented 1409 eligible results, published up to 21 December 2023. After removal of duplicates, 925 articles were extracted for screening. Two independent reviewers will screen for titles, abstracts and full texts, to extract data, which will then be organised using charting software. Data for various variables, that is, journal info, studied population demographics, study design, long COVID related data, cognitive outcomes and neuropsychological tests will be gathered. Descriptive analyses, evidence gap maps, heat map quantifications and narrative synthesis will be conducted for reporting of results.This scoping review has been registered with the Open Science Framework (https://doi.org/10.17605/OSF.IO/JHFX6). ETHICS AND DISSEMINATION Ethical approval is not required, as the study does not involve human participants. The findings will be disseminated through a publication in a scientific journal and within the professional network.
Collapse
Affiliation(s)
- Sara Monteiro
- Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Coralie Dessenne
- Science Office, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Magali Perquin
- Department of Precision Health, Luxembourg Institute of Health, Strassen, Luxembourg
| |
Collapse
|
223
|
Guillén N, Pérez-Millan A, Falgàs N, Lledó-Ibáñez GM, Rami L, Sarto J, Botí MA, Arnaldos-Pérez C, Ruiz-García R, Naranjo L, Segura B, Balasa M, Sala-Llonch R, Lladó A, Gray SM, Johannesen JK, Pantoni MM, Rutledge GA, Sawant R, Wang Y, Watson LS, Dalmau J, Sanchez-Valle R. Cognitive profile, neuroimaging and fluid biomarkers in post-acute COVID-19 syndrome. Sci Rep 2024; 14:12927. [PMID: 38839833 PMCID: PMC11153491 DOI: 10.1038/s41598-024-63071-2] [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: 11/16/2023] [Accepted: 05/24/2024] [Indexed: 06/07/2024] Open
Abstract
We aimed to characterize the cognitive profile of post-acute COVID-19 syndrome (PACS) patients with cognitive complaints, exploring the influence of biological and psychological factors. Participants with confirmed SARS-CoV-2 infection and cognitive complaints ≥ 8 weeks post-acute phase were included. A comprehensive neuropsychological battery (NPS) and health questionnaires were administered at inclusion and at 1, 3 and 6 months. Blood samples were collected at each visit, MRI scan at baseline and at 6 months, and, optionally, cerebrospinal fluid. Cognitive features were analyzed in relation to clinical, neuroimaging, and biochemical markers at inclusion and follow-up. Forty-nine participants, with a mean time from symptom onset of 10.4 months, showed attention-executive function (69%) and verbal memory (39%) impairment. Apathy (64%), moderate-severe anxiety (57%), and severe fatigue (35%) were prevalent. Visual memory (8%) correlated with total gray matter (GM) and subcortical GM volume. Neuronal damage and inflammation markers were within normal limits. Over time, cognitive test scores, depression, apathy, anxiety scores, MRI indexes, and fluid biomarkers remained stable, although fewer participants (50% vs. 75.5%; p = 0.012) exhibited abnormal cognitive evaluations at follow-up. Altered attention/executive and verbal memory, common in PACS, persisted in most subjects without association with structural abnormalities, elevated cytokines, or neuronal damage markers.
Collapse
Affiliation(s)
- Núria Guillén
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
| | - Agnès Pérez-Millan
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
- Institut de Neurociències, Faculty of Medicine and Medical Sciences, University of Barcelona, Barcelona, Spain
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Neus Falgàs
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
| | | | - Lorena Rami
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
| | - Jordi Sarto
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
| | - Maria A Botí
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
| | - Cristina Arnaldos-Pérez
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
- Immunology Service, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Raquel Ruiz-García
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
- Immunology Service, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Laura Naranjo
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
- Immunology Service, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Bàrbara Segura
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
- Institut de Neurociències, Faculty of Medicine and Medical Sciences, University of Barcelona, Barcelona, Spain
- Medical Psychology Unit, Department of Medicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Mircea Balasa
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
| | - Roser Sala-Llonch
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
- Institut de Neurociències, Faculty of Medicine and Medical Sciences, University of Barcelona, Barcelona, Spain
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - Albert Lladó
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
- Institut de Neurociències, Faculty of Medicine and Medical Sciences, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | - Yi Wang
- Sage Therapeutics, Cambridge, USA
| | | | - Josep Dalmau
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain
- Institut de Neurociències, Faculty of Medicine and Medical Sciences, University of Barcelona, Barcelona, Spain
- Department of Neurology, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
- Enfermedades Raras, Centro de Investigación Biomédica en Red, Madrid, Spain
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Catalan Institute for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Raquel Sanchez-Valle
- Alzheimer's Disease and Other Cognitive Disorders Unit, Service of Neurology, Hospital Clínic de Barcelona, Barcelona, Spain.
- Fundació Recerca Clínic Barcelona-IDIBAPS, Barcelona, Spain.
- Institut de Neurociències, Faculty of Medicine and Medical Sciences, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
224
|
Barboza-Solis C, Fantin R, Hildesheim A, Pfeiffer R, Porras C, Butt J, Waterboer T, Raventós H, Abdelnour A, Aparicio A, Loria V, Prevots DR, Gail MH, Herrero R. COVID-19 and long-term impact on symptoms and Health-Related Quality of Life in Costa Rica: the RESPIRA cohort study. BMC Infect Dis 2024; 24:557. [PMID: 38834971 DOI: 10.1186/s12879-024-09450-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Evidence continues to accumulate regarding the potential long-term health consequences of COVID-19 in the population. To distinguish between COVID-19-related symptoms and health limitations from those caused by other conditions, it is essential to compare cases with community controls using prospective data ensuring case-control status. The RESPIRA study addresses this need by investigating the lasting impact of COVID-19 on Health-related Quality of Life (HRQoL) and symptomatology in a population-based cohort in Costa Rica, thereby providing a robust framework for controlling HRQoL and symptoms. METHODS The study comprised 641 PCR-confirmed, unvaccinated cases of COVID-19 and 947 matched population-based controls. Infection was confirmed using antibody tests on enrollment serum samples and symptoms were monitored monthly for 6 months post-enrolment. Administered at the 6-month visit (occurring between 6- and 2-months post-diagnosis for cases and 6 months after enrollment for controls), HRQoL and Self-Perceived Health Change were assessed using the SF-36, while brain fog, using three items from the Mental Health Inventory (MHI). Regression models were utilized to analyze SF-36, MHI scores, and Self-Perceived Health Change, adjusted for case/control status, severity (mild case, moderate case, hospitalized) and additional independent variables. Sensitivity analyses confirmed the robustness of the findings. RESULTS Cases showed significantly higher prevalences of joint pain, chest tightness, and skin manifestations, that stabilized at higher frequencies from the fourth month post-diagnosis onwards (2.0%, 1.2%, and 0.8% respectively) compared to controls (0.9%, 0.4%, 0.2% respectively). Cases also exhibited significantly lower HRQoL than controls across all dimensions in the fully adjusted model, with a 12.4 percentage-point difference [95%CI: 9.4-14.6], in self-reported health compared to one year prior. Cases reported 8.0% [95%CI: 4.2, 11.5] more physical limitations, 7.3% [95%CI: 3.5, 10.5] increased lack of vitality, and 6.0% [95%CI: 2.4, 9.0] more brain fog compared to controls with similar characteristics. Undiagnosed cases detected with antibody tests among controls had HRQoL comparable to antibody negative controls. Differences were more pronounced in individuals with moderate or severe disease and among women. CONCLUSIONS PCR-confirmed unvaccinated cases experienced prolonged HRQoL reductions 6 months to 2 years after diagnosis, this was particularly the case in severe cases and among women. Mildly symptomatic cases showed no significant long-term sequelae.
Collapse
Affiliation(s)
| | - Romain Fantin
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - Allan Hildesheim
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - Ruth Pfeiffer
- Biostatistics Branch, Division of Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Carolina Porras
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - Julia Butt
- Division of Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Waterboer
- Division of Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Henriette Raventós
- Centro de Investigación en Biología Celular y Molecular, Universidad de Costa Rica, San José, Costa Rica
- Escuela de Biología, Universidad de Costa Rica, San José, Costa Rica
| | | | - Amada Aparicio
- Caja Costarricense de Seguro Social, San José, Costa Rica
| | - Viviana Loria
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - D Rebecca Prevots
- Epidemiology and Population Studies Unit, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Rockville, MD, USA
| | - Mitchell H Gail
- Biostatistics Branch, Division of Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive RM 7-E138, MSC 9780, Bethesda, MD, 20892, USA
| | - Rolando Herrero
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| |
Collapse
|
225
|
Jing Q, Song J, An G, Zhu E, Ai Z, Xiong L, Li C. Effective early strategy to prevent olfactory and gustatory dysfunction in COVID-19: a randomized controlled trial. QJM 2024; 117:348-352. [PMID: 37988146 DOI: 10.1093/qjmed/hcad262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/02/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Olfactory and gustatory dysfunctions (OGDs) are key symptoms of coronavirus disease 2019 (COVID-19), which may lead to neurological complications, and lack of effective treatment. This may be because post-disease treatments may be too late to protect the olfactory and gustatory functions. AIM To evaluate the effectiveness of early use of saline nasal irrigation (SNI), corticosteroid nasal spray, and saline or chlorhexidine gluconate mouthwash for preventing OGDs in COVID-19. DESIGN This study was a double-blind randomized controlled trial. METHODS The study was conducted from 5 May to 16 June 2022. We recruited patients from three hospitals who were admitted with COVID-19 but without OGDs on the day of admission. Olfactory and gustatory functions were evaluated using the Taste and Smell Survey and the numerical visual analog scale. Participants were randomized to the saline, drug or control groups. The control group received no intervention, saline group received SNI plus saline nasal spray and mouthwash, and the trial group received SNI plus budesonide nasal spray and chlorhexidine gluconate mouthwash. Participants were assessed again on the day of discharge. RESULTS A total of 379 patients completed the trial. The prevalence of OGDs was significantly lower in the saline (11.8%, 95% CI, 6.6-19.0%; P < 0.001) and drug (8.3%, 95% CI, 4.1-14.8%; P < 0.001) groups than in the control group (40.0%, 95% CI, 31.8-48.6%). Additionally, both interventions reduced the severity of OGDs. CONCLUSIONS We demonstrated effective strategies for preventing COVID-19-related OGDs, and the findings may guide early management of severe acute respiratory disease coronavirus 2 (SARS-CoV-2) infection to reduce the incidence of COVID-19-related complications.
Collapse
Affiliation(s)
- Q Jing
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - J Song
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - G An
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - E Zhu
- Department of Medical Statistics, Tongji University School of Medicine, Shanghai 200092, China
| | - Z Ai
- Department of Medical Statistics, Tongji University School of Medicine, Shanghai 200092, China
| | - L Xiong
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| | - C Li
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China
| |
Collapse
|
226
|
Verma AK, Lowery S, Lin LC, Duraisami E, Lloréns JEA, Qiu Q, Hefti M, Yu CR, Albers MW, Perlman S. Persistent Neurological Deficits in Mouse PASC Reveal Antiviral Drug Limitations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.596989. [PMID: 38895239 PMCID: PMC11185538 DOI: 10.1101/2024.06.02.596989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Post-Acute Sequelae of COVID-19 (PASC) encompasses persistent neurological symptoms, including olfactory and autonomic dysfunction. Here, we report chronic neurological dysfunction in mice infected with a virulent mouse-adapted SARS-CoV-2 that does not infect the brain. Long after recovery from nasal infection, we observed loss of tyrosine hydroxylase (TH) expression in olfactory bulb glomeruli and neurotransmitter levels in the substantia nigra (SN) persisted. Vulnerability of dopaminergic neurons in these brain areas was accompanied by increased levels of proinflammatory cytokines and neurobehavioral changes. RNAseq analysis unveiled persistent microglia activation, as found in human neurodegenerative diseases. Early treatment with antivirals (nirmatrelvir and molnupiravir) reduced virus titers and lung inflammation but failed to prevent neurological abnormalities, as observed in patients. Together these results show that chronic deficiencies in neuronal function in SARS-CoV-2-infected mice are not directly linked to ongoing olfactory epithelium dysfunction. Rather, they bear similarity with neurodegenerative disease, the vulnerability of which is exacerbated by chronic inflammation.
Collapse
Affiliation(s)
- Abhishek Kumar Verma
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
| | - Shea Lowery
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
| | - Li-Chin Lin
- Iowa Neuroscience Institute, University of Iowa, IA, USA 52242
- Department of Neurology, University of Iowa, Iowa City, IA 52242
| | | | | | - Qiang Qiu
- Stowers Institute for Medical Research, Kansas City, MO 64110
| | - Marco Hefti
- Department of Pathology, University of Iowa, Iowa City, IA 52242
| | - C. Ron Yu
- Stowers Institute for Medical Research, Kansas City, MO 64110
| | - Mark W. Albers
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Stanley Perlman
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
227
|
Abstract
Soon after the outbreak of coronavirus disease 2019 (COVID-19), unexplained sustained fatigue, cognitive disturbance, and muscle ache/weakness were reported in patients who had recovered from acute COVID-19 infection. This abnormal condition has been recognized as "long COVID (postacute sequelae of COVID-19 [PASC])" with a prevalence estimated to be from 10 to 20% of convalescent patients. Although the pathophysiology of PASC has been studied, the exact mechanism remains obscure. Microclots in circulation can represent one of the possible causes of PASC. Although hypercoagulability and thrombosis are critical mechanisms of acute COVID-19, recent studies have reported that thromboinflammation continues in some patients, even after the virus has cleared. Viral spike proteins and RNA can be detected months after patients have recovered, findings that may be responsible for persistent thromboinflammation and the development of microclots. Despite this theory, long-term results of anticoagulation, antiplatelet therapy, and vascular endothelial protection are inconsistent, and could not always show beneficial treatment effects. In summary, PASC reflects a heterogeneous condition, and microclots cannot explain all the presenting symptoms. After clarification of the pathomechanisms of each symptom, a symptom- or biomarker-based stratified approach should be considered for future studies.
Collapse
Affiliation(s)
- Toshiaki Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Jean M Connors
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jerrold H Levy
- Department of Anesthesiology, Critical Care, and Surgery, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
228
|
Pszczołowska M, Walczak K, Misków W, Antosz K, Batko J, Karska J, Leszek J. Molecular cross-talk between long COVID-19 and Alzheimer's disease. GeroScience 2024; 46:2885-2899. [PMID: 38393535 PMCID: PMC11009207 DOI: 10.1007/s11357-024-01096-1] [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: 11/22/2023] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
The long COVID (coronavirus disease), a multisystemic condition following severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection, is one of the widespread problems. Some of its symptoms affect the nervous system and resemble symptoms of Alzheimer's disease (AD)-a neurodegenerative condition caused by the accumulation of amyloid beta and hyperphosphorylation of tau proteins. Multiple studies have found dependence between these two conditions. Patients with Alzheimer's disease have a greater risk of SARS-CoV-2 infection due to increased levels of angiotensin-converting enzyme 2 (ACE2), and the infection itself promotes amyloid beta generation which enhances the risk of AD. Also, the molecular pathways are alike-misregulations in folate-mediated one-carbon metabolism, a deficit of Cq10, and disease-associated microglia. Medical imaging in both of these diseases shows a decrease in the volume of gray matter, global brain size reduction, and hypometabolism in the parahippocampal gyrus, thalamus, and cingulate cortex. In some studies, a similar approach to applied medication can be seen, including the use of amino adamantanes and phenolic compounds of rosemary. The significance of these connections and their possible application in medical practice still needs further study but there is a possibility that they will help to better understand long COVID.
Collapse
Affiliation(s)
| | - Kamil Walczak
- Faculty of Medicine, Wrocław Medical University, Wrocław, Poland
| | - Weronika Misków
- Faculty of Medicine, Wrocław Medical University, Wrocław, Poland
| | - Katarzyna Antosz
- Faculty of Medicine, Wrocław Medical University, Wrocław, Poland
| | - Joanna Batko
- Faculty of Medicine, Wrocław Medical University, Wrocław, Poland
| | - Julia Karska
- Clinic of Psychiatry, Department of Psychiatry, Medical Department, Wrocław Medical University, Wrocław, Poland
| | - Jerzy Leszek
- Clinic of Psychiatry, Department of Psychiatry, Medical Department, Wrocław Medical University, Wrocław, Poland
| |
Collapse
|
229
|
Montalba C. Editorial for "Gray Matter Changes Following Mild COVID-19: An MR Morphometric Study in Healthy Young People". J Magn Reson Imaging 2024; 59:2162-2163. [PMID: 37670484 DOI: 10.1002/jmri.28999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023] Open
Affiliation(s)
- Cristian Montalba
- Biomedical Imaging Center, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
230
|
Shen Q, Zhou YH, Zhou YQ. A prospects tool in virus research: Analyzing the applications of organoids in virus studies. Acta Trop 2024; 254:107182. [PMID: 38479469 DOI: 10.1016/j.actatropica.2024.107182] [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: 11/28/2023] [Revised: 02/23/2024] [Accepted: 03/10/2024] [Indexed: 04/28/2024]
Abstract
Organoids have emerged as a powerful tool for understanding the biology of the respiratory, digestive, nervous as well as urinary system, investigating infections, and developing new therapies. This article reviews recent progress in the development of organoid and advancements in virus research. The potential applications of these models in studying virul infections, pathogenesis, and antiviral drug discovery are discussed.
Collapse
Affiliation(s)
- Qi Shen
- Institute of Microbiology Laboratory, Shanghai Municipal Center for Disease Control and Prevention, Shanghai 20036, China; Institute of Microbiology Laboratory, Shanghai Institute of Preventive Medicine, Shanghai 20036, China
| | - Yu-Han Zhou
- College of Public Health, Jilin University, Changchun 130021, China
| | - Yan-Qiu Zhou
- Institute of Microbiology Laboratory, Shanghai Municipal Center for Disease Control and Prevention, Shanghai 20036, China; Institute of Microbiology Laboratory, Shanghai Institute of Preventive Medicine, Shanghai 20036, China.
| |
Collapse
|
231
|
Boyes A, Levenstein JM, McLoughlin LT, Driver C, Mills L, Lagopoulos J, Hermens DF. A short-interval longitudinal study of associations between psychological distress and hippocampal grey matter in early adolescence. Brain Imaging Behav 2024; 18:519-528. [PMID: 38216837 PMCID: PMC11222233 DOI: 10.1007/s11682-023-00847-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2023] [Indexed: 01/14/2024]
Abstract
This study of Australian adolescents (N = 88, 12-13-years-old) investigated the relationship between hippocampal grey matter volume (GMV) and self-reported psychological distress (K10) at four timepoints, across 12 months. Participants were divided into two groups; those who had K10 scores between 10 and 15 for all four timepoints were categorised as "low distress" (i.e., control group; n = 38), while participants who had K10 scores of 16 or higher at least once over the year were categorised as "moderate-high distress" (n = 50). Associations were tested by GEE fitting of GMV and K10 measures at the same time point, and in the preceding and subsequent timepoints. Analyses revealed smaller preceding left GMV and larger preceding right GMV were associated with higher subsequent K10 scores in the "moderate-high distress" group. This was not observed in the control group. In contrast, the control group showed significant co-occurring associations (i.e., at the same TP) between GMV and K10 scores. The "moderate-high distress" group experienced greater variability in distress. These results suggest that GMV development in early adolescence is differently associated with psychological distress for those who experience "moderate-high distress" at some point over the year, compared to controls. These findings offer a novel way to utilise short-interval, multiple time-point longitudinal data to explore changes in volume and experience of psychological distress in early adolescents. The results suggest hippocampal volume in early adolescence may be linked to fluctuations in psychological distress.
Collapse
Affiliation(s)
- Amanda Boyes
- Thompson Institute, UniSC, 12 Innovation Parkway, Birtinya, QLD, 4575, Australia.
| | - Jacob M Levenstein
- Thompson Institute, UniSC, 12 Innovation Parkway, Birtinya, QLD, 4575, Australia
| | - Larisa T McLoughlin
- Thompson Institute, UniSC, 12 Innovation Parkway, Birtinya, QLD, 4575, Australia
| | - Christina Driver
- Thompson Institute, UniSC, 12 Innovation Parkway, Birtinya, QLD, 4575, Australia
| | - Lia Mills
- Thompson Institute, UniSC, 12 Innovation Parkway, Birtinya, QLD, 4575, Australia
| | - Jim Lagopoulos
- Thompson Institute, UniSC, 12 Innovation Parkway, Birtinya, QLD, 4575, Australia
| | - Daniel F Hermens
- Thompson Institute, UniSC, 12 Innovation Parkway, Birtinya, QLD, 4575, Australia
| |
Collapse
|
232
|
Pandey S, Bapat V, Abraham JN, Abraham NM. Long COVID: From olfactory dysfunctions to viral Parkinsonism. World J Otorhinolaryngol Head Neck Surg 2024; 10:137-147. [PMID: 38855289 PMCID: PMC11156689 DOI: 10.1002/wjo2.175] [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: 12/27/2023] [Revised: 02/20/2024] [Accepted: 03/08/2024] [Indexed: 06/11/2024] Open
Abstract
Neurological and psychiatric complications continue to be a public health concern in long coronavirus disease 2019 (COVID-19). This varies from olfactory dysfunctions such as parosmia to cognitive and emotional challenges. Historically, the surge of neurological disorders followed the viral pandemics, for example, the emergence of Encephalitis Lethargica after the outbreak of Spanish Influenza. During and after COVID-19 infection, the problems associated with the sense of smell and the reports of affected olfactory and limbic brain areas are leading to a growing concern about the similarity with the symptoms and the pattern of degeneration observed at the onset of Parkinson's disease and Alzheimer's disease. These reports reveal the essentiality of long-term studies of olfactory and cognitive functions in the post-COVID era and the experiments using animal models to dissect the neural basis of these complications. In this manuscript, we summarize the research reporting the potential correlation between neurological disorders and viral pandemic outbreaks with a historical perspective. Further, we discuss the studies providing evidence of neurodegeneration due to severe acute respiratory syndrome coronavirus 2 infection by focusing on viral Parkinsonism.
Collapse
Affiliation(s)
- Sanyukta Pandey
- Department of Biology, Laboratory of Neural Circuits and Behaviour (LNCB)Indian Institute of Science Education and Research (IISER)PuneMaharashtraIndia
| | - Vibha Bapat
- Department of Biology, Laboratory of Neural Circuits and Behaviour (LNCB)Indian Institute of Science Education and Research (IISER)PuneMaharashtraIndia
| | - Jancy Nixon Abraham
- Department of Biology, Laboratory of Neural Circuits and Behaviour (LNCB)Indian Institute of Science Education and Research (IISER)PuneMaharashtraIndia
- Department of Life Sciences, Centre of Excellence in EpigeneticsShiv Nadar Institution of EminenceGautam Buddha NagarUttar PradeshIndia
| | - Nixon M. Abraham
- Department of Biology, Laboratory of Neural Circuits and Behaviour (LNCB)Indian Institute of Science Education and Research (IISER)PuneMaharashtraIndia
| |
Collapse
|
233
|
Zhao Y, Liang Q, Jiang Z, Mei H, Zeng N, Su S, Wu S, Ge Y, Li P, Lin X, Yuan K, Shi L, Yan W, Liu X, Sun J, Liu W, van Wingen G, Gao Y, Tan Y, Hong Y, Lu Y, Wu P, Zhang X, Wang Y, Shi J, Wang Y, Lu L, Li X, Bao Y. Brain abnormalities in survivors of COVID-19 after 2-year recovery: a functional MRI study. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2024; 47:101086. [PMID: 38774424 PMCID: PMC11107230 DOI: 10.1016/j.lanwpc.2024.101086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/03/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024]
Abstract
Background A variety of symptoms, particularly cognitive, psychiatric and neurological symptoms, may persist for a long time among individuals recovering from COVID-19. However, the underlying mechanism of these brain abnormalities remains unclear. This study aimed to investigate the long-term neuroimaging effects of COVID-19 infection on brain functional activities using resting-state functional magnetic resonance imaging (rs-fMRI). Methods Fifty-two survivors 27 months after infection (mild-moderate group: 25 participants, severe-critical: 27 participants), from our previous community participants, along with 35 healthy controls, were recruited to undergo fMRI scans and comprehensive cognitive function measurements. Participants were evaluated by subjective assessment of Cognitive Failures Questionnaire-14 (CFQ-14) and Fatigue Scale-14 (FS-14), and objective assessment of Montreal Cognitive Assessment (MoCA), N-back, and Simple Reaction Time (SRT). Each had rs-fMRI at 3T. Measures such as the amplitude of low-frequency fluctuation (ALFF), fractional amplitude of low-frequency fluctuations (fALFF), and regional homogeneity (ReHo) were calculated. Findings Compared with healthy controls, survivors of mild-moderate acute symptoms group and severe-critical group had a significantly higher score of cognitive complains involving cognitive failure and mental fatigue. However, there was no difference of cognitive complaints between two groups of COVID-19 survivors. The performance of three groups was similar on the score of MoCA, N-back and SRT. The rs-fMRI results showed that COVID-19 survivors exhibited significantly increased ALFF values in the left putamen (PUT.L), right inferior temporal gyrus (ITG.R) and right pallidum (PAL.R), while decreased ALFF values were observed in the right superior parietal gyrus (SPG.R) and left superior temporal gyrus (STG.L). Additionally, decreased ReHo values in the right precentral gyrus (PreCG.R), left postcentral gyrus (PoCG.L), left calcarine fissure and surrounding cortex (CAL.L) and left superior temporal gyrus (STG.L). Furthermore, significant negative correlations between the ReHo values in the STG.L, and CFQ-14 and mental fatigue were found. Interpretation This long-term study suggests that individuals recovering from COVID-19 continue to experience cognitive complaints, psychiatric and neurological symptoms, and brain functional alteration. The rs-fMRI results indicated that the changes in brain function in regions such as the putamen, temporal lobe, and superior parietal gyrus may contribute to cognitive complaints in individuals with long COVID even after 2-year infection. Funding The National Programs for Brain Science and Brain-like Intelligence Technology of China, the National Natural Science Foundation of China, Natural Science Foundation of Beijing Municipality of China, and the National Key Research and Development Program of China.
Collapse
Affiliation(s)
- Yimiao Zhao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing 100191, China
- School of Public Health, Peking University, Beijing 100191, China
| | - Qiongdan Liang
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing 100191, China
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100191, China
| | - Zhendong Jiang
- Wuhan Wuchang Hospital, Wuhan University of Science and Technology, Wuhan, Hubei Province 430063, China
| | - Huan Mei
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing 100191, China
- School of Public Health, Peking University, Beijing 100191, China
| | - Na Zeng
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing 100191, China
- School of Public Health, Peking University, Beijing 100191, China
| | - Sizhen Su
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing 100191, China
| | - Shanshan Wu
- Wuhan Wuchang Hospital, Wuhan University of Science and Technology, Wuhan, Hubei Province 430063, China
| | - Yinghong Ge
- The Third Hospital of Wuhan City, Wuhan, Hubei Province 430000, China
| | - Peng Li
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing 100191, China
| | - Xiao Lin
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing 100191, China
| | - Kai Yuan
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing 100191, China
| | - Le Shi
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing 100191, China
| | - Wei Yan
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing 100191, China
| | - Xiaoxing Liu
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing 100191, China
| | - Jie Sun
- Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Weijian Liu
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing 100191, China
- Chinese Academy of Medical Sciences Research Unit (No. 2018RU006), Peking University, Beijing 100191, China
- Department of Psychiatry, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Guido van Wingen
- Department of Psychiatry, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Yujun Gao
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing 100191, China
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430000, China
| | - Yiqing Tan
- The Third Hospital of Wuhan City, Wuhan, Hubei Province 430000, China
| | - Yi Hong
- Wuhan Wuchang Hospital, Wuhan University of Science and Technology, Wuhan, Hubei Province 430063, China
| | - Yu Lu
- Wuhan Wuchang Hospital, Wuhan University of Science and Technology, Wuhan, Hubei Province 430063, China
| | - Ping Wu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing 100191, China
| | - Xiujun Zhang
- School of Psychology, College of Public Health, North China University of Science and Technology, 21 Bohai Road, Tang'shan, Hebei Province 063210, China
| | - Yongxiang Wang
- Shandong Institute of Brain Science and Brain-Inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province 271016, China
| | - Jie Shi
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing 100191, China
| | - Yumei Wang
- Shandong Institute of Brain Science and Brain-Inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province 271016, China
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province 250021, China
| | - Lin Lu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing 100191, China
- Institute of Mental Health, National Clinical Research Center for Mental Disorders, Key Laboratory of Mental Health and Peking University Sixth Hospital, Peking University, Beijing 100191, China
- Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100191, China
- Chinese Academy of Medical Sciences Research Unit (No. 2018RU006), Peking University, Beijing 100191, China
- Shandong Institute of Brain Science and Brain-Inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province 271016, China
| | - Xiangyou Li
- Wuhan Wuchang Hospital, Wuhan University of Science and Technology, Wuhan, Hubei Province 430063, China
| | - Yanping Bao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing 100191, China
- School of Public Health, Peking University, Beijing 100191, China
- Shandong Institute of Brain Science and Brain-Inspired Research, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong Province 271016, China
| |
Collapse
|
234
|
Muhamad NA, Masutani K, Furukawa S, Yuri S, Toriyama M, Matsumoto C, Itoh S, Shinagawa Y, Isotani A, Toriyama M, Itoh H. Astrocyte-Specific Inhibition of the Primary Cilium Suppresses C3 Expression in Reactive Astrocyte. Cell Mol Neurobiol 2024; 44:48. [PMID: 38822888 PMCID: PMC11144130 DOI: 10.1007/s10571-024-01482-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 05/21/2024] [Indexed: 06/03/2024]
Abstract
C3-positive reactive astrocytes play a neurotoxic role in various neurodegenerative diseases. However, the mechanisms controlling C3-positive reactive astrocyte induction are largely unknown. We found that the length of the primary cilium, a cellular organelle that receives extracellular signals was increased in C3-positive reactive astrocytes, and the loss or shortening of primary cilium decreased the count of C3-positive reactive astrocytes. Pharmacological experiments suggested that Ca2+ signalling may synergistically promote C3 expression in reactive astrocytes. Conditional knockout (cKO) mice that specifically inhibit primary cilium formation in astrocytes upon drug stimulation exhibited a reduction in the proportions of C3-positive reactive astrocytes and apoptotic cells in the brain even after the injection of lipopolysaccharide (LPS). Additionally, the novel object recognition (NOR) score observed in the cKO mice was higher than that observed in the neuroinflammation model mice. These results suggest that the primary cilium in astrocytes positively regulates C3 expression. We propose that regulating astrocyte-specific primary cilium signalling may be a novel strategy for the suppression of neuroinflammation.
Collapse
Affiliation(s)
- Nor Atiqah Muhamad
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Kohei Masutani
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Shota Furukawa
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Shunsuke Yuri
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Michinori Toriyama
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, 1 Gakuenuegahara, Sanda, Hyogo, 669-1330, Japan
| | - Chuya Matsumoto
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Seiya Itoh
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Yuichiro Shinagawa
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Ayako Isotani
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan
| | - Manami Toriyama
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan.
| | - Hiroshi Itoh
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5, Takayama Cho, Ikoma, Nara, 630-0192, Japan.
| |
Collapse
|
235
|
Perlaki G, Darnai G, Arató Á, Alhour HA, Szente A, Áfra E, Nagy SA, Horváth R, Kovács N, Dóczi T, Orsi G, Janszky J. Gray Matter Changes Following Mild COVID-19: An MR Morphometric Study in Healthy Young People. J Magn Reson Imaging 2024; 59:2152-2161. [PMID: 37602529 DOI: 10.1002/jmri.28970] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 08/22/2023] Open
Abstract
BACKGROUND Although COVID-19 is primarily an acute respiratory infection, 5%-40% of patients develop late and prolonged symptoms with frequent neurological complaints, known as long COVID syndrome. The presentation of the disease suggests that COVID infection may cause functional and/or morphological central nervous system alterations, but studies published in the literature report contradictory findings. PURPOSE To investigate the chronic effects of COVID-19 on cerebral grey matter in a group of young patients without comorbidities, with mild course of COVID infection and no medical complaints at the time of examination. STUDY TYPE Prospective. POPULATION Thirty-eight young (age = 26.6 ± 5.0 years; male/female = 14/24), adult participants who recovered from mild COVID infection without a history of clinical long COVID and 37 healthy control subjects (age = 25.9 ± 2.8 years; male/female = 14/23). FIELD STRENGTH/SEQUENCE Three Tesla, 3D T1-weighted magnetization-prepared rapid gradient-echo, 2D T2-weighted turbo spin-echo. ASSESSMENT MRI-based morphometry and volumetry along with neuropsychological testing and self-assessed questionnaire. STATISTICAL TESTS Fisher's exact test, Mann-Whitney U-test, and multiple linear regression analyses were used to assess differences between COVID and healthy control groups. P < 0.05 was used as cutoff for significance. RESULTS In the COVID group, significantly lower bilateral mean cortical thickness (left/right-hemisphere: 2.51 ± 0.06 mm vs. 2.56 ± 0.07 mm, η2 p = 0.102/2.50 ± 0.06 mm vs. 2.54 ± 0.07 mm, η2 p = 0.101), lower subcortical gray matter (57881 ± 3998 mm3 vs. 60470 ± 5211 mm3, η2 p = 0.100) and lower right olfactory bulb volume (52.28 ± 13.55 mm3 vs. 60.98 ± 15.8 mm3, η2 p = 0.078) were found. In patients with moderate to severe anosmia, cortical thickness was significantly lower bilaterally, as compared to patients without olfactory function loss (left/right-hemisphere: 2.50 ± 0.06 mm vs. 2.56 ± 0.05 mm, η2 = 0.173/2.49 ± 0.06 mm vs. 2.55 ± 0.05 mm, η2 = 0.189). Using further exploratory analysis, significantly reduced cortical thickness was detected locally in the right lateral orbitofrontal cortex in the COVID group (2.53 ± 0.10 mm vs. 2.60 ± 0.09 mm, η2 p = 0.112). DATA CONCLUSION Even without any subjective or objective neurological complaints at the time of the MR scan, subjects in the COVID group showed gray matter alterations in cortical thickness and subcortical gray matter volume. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Gábor Perlaki
- Department of Neurology, Medical School, University of Pécs, Pécs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, Eötvös Loránd Research Network, Pécs, Hungary
- Pécs Diagnostic Centre, NeuroCT Ltd., Pécs, Hungary
| | - Gergely Darnai
- Department of Neurology, Medical School, University of Pécs, Pécs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, Eötvös Loránd Research Network, Pécs, Hungary
- Department of Behavioural Sciences, Medical School, University of Pécs, Pécs, Hungary
| | - Ákos Arató
- Department of Neurology, Medical School, University of Pécs, Pécs, Hungary
| | | | - Anna Szente
- Department of Neurology, Medical School, University of Pécs, Pécs, Hungary
| | - Eszter Áfra
- Department of Behavioural Sciences, Medical School, University of Pécs, Pécs, Hungary
| | - Szilvia Anett Nagy
- Department of Neurology, Medical School, University of Pécs, Pécs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, Eötvös Loránd Research Network, Pécs, Hungary
- Pécs Diagnostic Centre, NeuroCT Ltd., Pécs, Hungary
- Structural Neurobiology Research Group, Szentágothai Research Centre, University of Pecs, Pécs, Hungary
| | - Réka Horváth
- Department of Neurology, Medical School, University of Pécs, Pécs, Hungary
| | - Norbert Kovács
- Department of Neurology, Medical School, University of Pécs, Pécs, Hungary
| | - Tamás Dóczi
- Pécs Diagnostic Centre, NeuroCT Ltd., Pécs, Hungary
- Department of Neurosurgery, Medical School, University of Pécs, Pécs, Hungary
| | - Gergely Orsi
- Department of Neurology, Medical School, University of Pécs, Pécs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, Eötvös Loránd Research Network, Pécs, Hungary
- Pécs Diagnostic Centre, NeuroCT Ltd., Pécs, Hungary
- Department of Neurosurgery, Medical School, University of Pécs, Pécs, Hungary
| | - József Janszky
- Department of Neurology, Medical School, University of Pécs, Pécs, Hungary
- ELKH-PTE Clinical Neuroscience MR Research Group, Eötvös Loránd Research Network, Pécs, Hungary
| |
Collapse
|
236
|
Dias M, Shaida Z, Haloob N, Hopkins C. Recovery rates and long-term olfactory dysfunction following COVID-19 infection. World J Otorhinolaryngol Head Neck Surg 2024; 10:121-128. [PMID: 38855291 PMCID: PMC11156684 DOI: 10.1002/wjo2.163] [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: 11/06/2023] [Accepted: 02/08/2024] [Indexed: 06/11/2024] Open
Abstract
Objectives Olfactory dysfunction is one of the most recognized symptoms of COVID-19, significantly impacting quality of life, particularly in cases where recovery is prolonged. This review aims to explore patterns of olfactory recovery post-COVID-19 infection, with particular focus on delayed recovery. Data Sources Published literature in the English language, including senior author's own work, online and social media platforms, and patients' anecdotal reports. Method A comprehensive review of the literature was undertaken by the authors with guidance from the senior author with expertise in the field of olfaction. Results Based on self-report, an estimated 95% of patients recover their olfactory function within 6 months post-COVID-19 infection. However, psychophysical testing detects higher rates of persistent olfactory dysfunction. Recovery has been found to continue for at least 2 years postinfection; negative prognostic indicators include severe olfactory loss in the acute phase, female sex, and older age. Variability in quantitative and qualitative disturbance in prolonged cases likely reflects both peripheral and central pathophysiological mechanisms. Limitations of many of the reviewed studies reflect lack of psychophysical testing and baseline olfactory assessment. Conclusions Post-COVID-19 olfactory dysfunction remains a significant health and psychosocial burden. Emerging evidence is improving awareness and knowledge among clinicians to better support patients through their olfactory rehabilitation, with hope of recovery after several months or years. Further research is needed to better understand the underlying pathogenesis of delayed recovery, identify at risk individuals earlier in the disease course, and develop therapeutic targets.
Collapse
|
237
|
Biddle G, Beck RT, Raslan O, Ebinu J, Jenner Z, Hamer J, Hacein-Bey L, Apperson M, Ivanovic V. Autoimmune diseases of the spine and spinal cord. Neuroradiol J 2024; 37:285-303. [PMID: 37394950 PMCID: PMC11138326 DOI: 10.1177/19714009231187340] [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: 07/04/2023] Open
Abstract
Magnetic resonance imaging (MRI) and clinicopathological tools have led to the identification of a wide spectrum of autoimmune entities that involve the spine. A clearer understanding of the unique imaging features of these disorders, along with their clinical presentations, will prove invaluable to clinicians and potentially limit the need for more invasive procedures such as tissue biopsies. Here, we review various autoimmune diseases affecting the spine and highlight salient imaging features that distinguish them radiologically from other disease entities.
Collapse
Affiliation(s)
- Garrick Biddle
- Radiology Department, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Ryan T Beck
- Neuroradiology, Radiology Department, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Osama Raslan
- Radiology Department, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Julius Ebinu
- Neurosurgery Department, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Zach Jenner
- Radiology Department, University of California Davis School of Medicine, Sacramento, CA, USA
| | - John Hamer
- Neuroradiology, Radiology Department, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lotfi Hacein-Bey
- Radiology Department, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Michelle Apperson
- Neurology Department, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Vladimir Ivanovic
- Neuroradiology, Radiology Department, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
238
|
Rahman A, Russell M, Zheng W, Eckrich D, Ahmed I, On behalf of the N3C Consortium. SARS-CoV-2 infection is associated with an increase in new diagnoses of schizophrenia spectrum and psychotic disorder: A study using the US national COVID cohort collaborative (N3C). PLoS One 2024; 19:e0295891. [PMID: 38814888 PMCID: PMC11139284 DOI: 10.1371/journal.pone.0295891] [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: 12/02/2023] [Accepted: 03/13/2024] [Indexed: 06/01/2024] Open
Abstract
Amid the ongoing global repercussions of SARS-CoV-2, it is crucial to comprehend its potential long-term psychiatric effects. Several recent studies have suggested a link between COVID-19 and subsequent mental health disorders. Our investigation joins this exploration, concentrating on Schizophrenia Spectrum and Psychotic Disorders (SSPD). Different from other studies, we took acute respiratory distress syndrome (ARDS) and COVID-19 lab-negative cohorts as control groups to accurately gauge the impact of COVID-19 on SSPD. Data from 19,344,698 patients, sourced from the N3C Data Enclave platform, were methodically filtered to create propensity matched cohorts: ARDS (n = 222,337), COVID-19 positive (n = 219,264), and COVID-19 negative (n = 213,183). We systematically analyzed the hazard rate of new-onset SSPD across three distinct time intervals: 0-21 days, 22-90 days, and beyond 90 days post-infection. COVID-19 positive patients consistently exhibited a heightened hazard ratio (HR) across all intervals [0-21 days (HR: 4.6; CI: 3.7-5.7), 22-90 days (HR: 2.9; CI: 2.3 -3.8), beyond 90 days (HR: 1.7; CI: 1.5-1.)]. These are notably higher than both ARDS and COVID-19 lab-negative patients. Validations using various tests, including the Cochran Mantel Haenszel Test, Wald Test, and Log-rank Test confirmed these associations. Intriguingly, our data indicated that younger individuals face a heightened risk of SSPD after contracting COVID-19, a trend not observed in the ARDS and COVID-19 negative groups. These results, aligned with the known neurotropism of SARS-CoV-2 and earlier studies, accentuate the need for vigilant psychiatric assessment and support in the era of Long-COVID, especially among younger populations.
Collapse
Affiliation(s)
- Asif Rahman
- Department of Industrial & Management Systems Engineering, West Virginia University, Morgantown, WV, United States of America
| | - Michael Russell
- School of Medicine, West Virginia University, Morgantown, WV, United States of America
| | - Wanhong Zheng
- School of Medicine, West Virginia University, Morgantown, WV, United States of America
| | - Daniel Eckrich
- Nemours Children’s Health, Jacksonville, FL, United States of America
| | - Imtiaz Ahmed
- Department of Industrial & Management Systems Engineering, West Virginia University, Morgantown, WV, United States of America
| | | |
Collapse
|
239
|
Han Q, Ma Y, Ye W. Association between COVID-19 and the Risk of Vascular Dementia: A Mendelian Randomisation Study of the Potential Cognitive Sequela of COVID-19. Behav Sci (Basel) 2024; 14:465. [PMID: 38920797 PMCID: PMC11200909 DOI: 10.3390/bs14060465] [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: 04/12/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
A growing body of observational studies and Mendelian Randomisation analyses suggest an increased risk of Alzheimer's disease and dementia following COVID-19 infection. However, evidence on the potential association between COVID-19 and vascular dementia, which is plausible given the vascular complications of COVID-19 infection, is still limited. In this study, we conducted a two-sample Mendelian Randomisation analysis to examine the potential causal relationship between COVID-19 phenotypes and the risk of vascular dementia, using summary data from large-scale GWASs. The two-sample Mendelian Randomisation analysis did not detect any significant associations of COVID-19 infection, COVID-19 hospitalisation, or critical COVID-19 with the risk of vascular dementia, with weighted average β values of -0.29 (95% CI: -0.84, 0.26; p = 0.301), -0.12 (95% CI: -0.36, 0.13; p = 0.345), and -0.07 (95% CI: -0.23, 0.09; p = 0.374), respectively. Our findings do not support the hypothesis that vascular dementia is one of the long-term sequelae of COVID-19.
Collapse
Affiliation(s)
- Qing Han
- Department of Social Policy and Intervention, University of Oxford, Oxford OX1 2ER, UK
| | - Yue Ma
- Faculty of Medicine, Macau University of Science and Technology, Macao, China
- School of Nursing, Southern Medical University, Guangzhou 510515, China
| | - Wenting Ye
- Faculty of Psychology, Southwest University (SWU), Chongqing 400715, China
| |
Collapse
|
240
|
Duve K, Petakh P, Kamyshnyi O. COVID-19-associated encephalopathy: connection between neuroinflammation and microbiota-gut-brain axis. Front Microbiol 2024; 15:1406874. [PMID: 38863751 PMCID: PMC11165208 DOI: 10.3389/fmicb.2024.1406874] [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: 04/03/2024] [Accepted: 05/16/2024] [Indexed: 06/13/2024] Open
Abstract
While neurological complications of COVID-19, such as encephalopathy, are relatively rare, their potential significant impact on long-term morbidity is substantial, especially given the large number of infected patients. Two proposed hypotheses for the pathogenesis of this condition are hypoxia and the uncontrolled release of proinflammatory cytokines. The gut microbiota plays an important role in regulating immune homeostasis and overall gut health, including its effects on brain health through various pathways collectively termed the gut-brain axis. Recent studies have shown that COVID-19 patients exhibit gut dysbiosis, but how this dysbiosis can affect inflammation in the central nervous system (CNS) remains unclear. In this context, we discuss how dysbiosis could contribute to neuroinflammation and provide recent data on the features of neuroinflammation in COVID-19 patients.
Collapse
Affiliation(s)
- Khrystyna Duve
- Department of Neurology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Oleksandr Kamyshnyi
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
| |
Collapse
|
241
|
Tomasa-Irriguible TM, Monfà R, Miranda-Jiménez C, Morros R, Robert N, Bordejé-Laguna L, Vidal S, Torán-Monserrat P, Barriocanal AM. Preventive Intake of a Multiple Micronutrient Supplement during Mild, Acute SARS-CoV-2 Infection to Reduce the Post-Acute COVID-19 Condition: A Double-Blind, Placebo-Controlled, Randomized Clinical Trial. Nutrients 2024; 16:1631. [PMID: 38892564 PMCID: PMC11174626 DOI: 10.3390/nu16111631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Patients hospitalized with COVID-19 have low levels of vitamins and trace elements. This could lead to a post-acute COVID-19 condition (PCC) that can worsen a patient's quality of life. We aimed to study the baseline micronutrient status of patients and assess whether a multiple micronutrient supplement (MMS) taken for 2 weeks at the first sign of COVID-19 symptoms would be able to reduce the incidence of PCC. This double-blind, placebo-controlled, randomized clinical trial was conducted in adult outpatients with acute COVID-19, recruited between 2021 and 2023 in Spain. Of the 285 patients assessed for eligibility, 267 were randomized and 246 were included in the intent-to-treat population. The mean age was 46.8 years, and 68% were female. Overall, 54.6% had micronutrient deficiencies in the acute phase of COVID-19 at baseline, and 26.2% had PCC after 180 days of follow-up (D180). The most frequently recorded PCC symptoms were neurological (14.1%), with 24% patients scoring worse in the cognitive tests compared to their baseline status. The rate of PCC at D180 was similar between the placebo (25.0%) and intervention (27.7%) groups, without significant differences (p = 0.785). Age over 50 years was the most relevant risk factor for developing PCC, followed by female sex. The most important protective factor against PCC was SARS-CoV-2 vaccination. In this population of predominantly middle-aged, white women with acute COVID-19 not requiring hospital admission, MMS intake for 14 days at symptom onset did not prevent PCC nor improve their micronutrient status at D180.
Collapse
Affiliation(s)
| | - Ramon Monfà
- Jordi Gol University Research Institute in Primary Care (IDIAP Jordi Gol), 08007 Barcelona, Spain; (R.M.); (C.M.-J.)
| | - Cristina Miranda-Jiménez
- Jordi Gol University Research Institute in Primary Care (IDIAP Jordi Gol), 08007 Barcelona, Spain; (R.M.); (C.M.-J.)
| | - Rosa Morros
- Department of Pharmacology, Therapeutics and Toxicology, Autonomous University of Barcelona (UAB), 08193 Bellaterra, Spain;
| | - Neus Robert
- Emergency Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain;
| | - Luisa Bordejé-Laguna
- Intensive Care Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain;
| | - Sandra Vidal
- Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain; (S.V.); (A.M.B.)
| | - Pere Torán-Monserrat
- North Metropolitan Research Support Unit, Jordi Gol University Research Institute in Primary Care (NM-IDIAP Jordi Gol), 08303 Mataró, Spain;
| | - Ana Maria Barriocanal
- Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain; (S.V.); (A.M.B.)
| |
Collapse
|
242
|
Kim AY, Al Jerdi S, MacDonald R, Triggle CR. Alzheimer's disease and its treatment-yesterday, today, and tomorrow. Front Pharmacol 2024; 15:1399121. [PMID: 38868666 PMCID: PMC11167451 DOI: 10.3389/fphar.2024.1399121] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/25/2024] [Indexed: 06/14/2024] Open
Abstract
Alois Alzheimer described the first patient with Alzheimer's disease (AD) in 1907 and today AD is the most frequently diagnosed of dementias. AD is a multi-factorial neurodegenerative disorder with familial, life style and comorbidity influences impacting a global population of more than 47 million with a projected escalation by 2050 to exceed 130 million. In the USA the AD demographic encompasses approximately six million individuals, expected to increase to surpass 13 million by 2050, and the antecedent phase of AD, recognized as mild cognitive impairment (MCI), involves nearly 12 million individuals. The economic outlay for the management of AD and AD-related cognitive decline is estimated at approximately 355 billion USD. In addition, the intensifying prevalence of AD cases in countries with modest to intermediate income countries further enhances the urgency for more therapeutically and cost-effective treatments and for improving the quality of life for patients and their families. This narrative review evaluates the pathophysiological basis of AD with an initial focus on the therapeutic efficacy and limitations of the existing drugs that provide symptomatic relief: acetylcholinesterase inhibitors (AChEI) donepezil, galantamine, rivastigmine, and the N-methyl-D-aspartate receptor (NMDA) receptor allosteric modulator, memantine. The hypothesis that amyloid-β (Aβ) and tau are appropriate targets for drugs and have the potential to halt the progress of AD is critically analyzed with a particular focus on clinical trial data with anti-Aβ monoclonal antibodies (MABs), namely, aducanumab, lecanemab and donanemab. This review challenges the dogma that targeting Aβ will benefit the majority of subjects with AD that the anti-Aβ MABs are unlikely to be the "magic bullet". A comparison of the benefits and disadvantages of the different classes of drugs forms the basis for determining new directions for research and alternative drug targets that are undergoing pre-clinical and clinical assessments. In addition, we discuss and stress the importance of the treatment of the co-morbidities, including hypertension, diabetes, obesity and depression that are known to increase the risk of developing AD.
Collapse
Affiliation(s)
- A. Y. Kim
- Medical Education, Weill Cornell Medicine—Qatar, Doha, Qatar
| | | | - R. MacDonald
- Health Sciences Library, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - C. R. Triggle
- Department of Pharmacology and Medical Education, Weill Cornell Medicine—Qatar, Doha, Qatar
| |
Collapse
|
243
|
Hu WT, Kaluzova M, Dawson A, Sotelo V, Papas J, Lemenze A, Shu C, Jomartin M, Nayyar A, Hussain S. Clinical and CSF single-cell profiling of post-COVID-19 cognitive impairment. Cell Rep Med 2024; 5:101561. [PMID: 38744274 PMCID: PMC11148803 DOI: 10.1016/j.xcrm.2024.101561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/15/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Natural history and mechanisms for persistent cognitive symptoms ("brain fog") following acute and often mild COVID-19 are unknown. In a large prospective cohort of people who underwent testing a median of 9 months after acute COVID-19 in the New York City/New Jersey area, we found that cognitive dysfunction is common; is not influenced by mood, fatigue, or sleepiness; and is correlated with MRI changes in very few people. In a subgroup that underwent cerebrospinal fluid analysis, there are no changes related to Alzheimer's disease or neurodegeneration. Single-cell gene expression analysis in the cerebrospinal fluid shows findings consistent with monocyte recruitment, chemokine signaling, cellular stress, and suppressed interferon response-especially in myeloid cells. Longitudinal analysis shows slow recovery accompanied by key alterations in inflammatory genes and increased protein levels of CXCL8, CCL3L1, and sTREM2. These findings suggest that the prognosis for brain fog following COVID-19 correlates with myeloid-related chemokine and interferon-responsive genes.
Collapse
Affiliation(s)
- William T Hu
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA.
| | - Milota Kaluzova
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Alice Dawson
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA
| | - Victor Sotelo
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA
| | - Julia Papas
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA; Center for Innovation in Health and Aging Research, Institute for Health, Health Care Policy, and Aging Research, New Brunswick, NJ, USA
| | - Alexander Lemenze
- Department of Pathology and Laboratory Medicine, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Carol Shu
- Department of Medicine-Pulmonary and Critical Care, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Mini Jomartin
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Ashima Nayyar
- Department of Neurology, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Sabiha Hussain
- Department of Medicine-Pulmonary and Critical Care, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
244
|
Hosp JA, Reisert M, Dressing A, Götz V, Kellner E, Mast H, Arndt S, Waller CF, Wagner D, Rieg S, Urbach H, Weiller C, Schröter N, Rau A. Cerebral microstructural alterations in Post-COVID-condition are related to cognitive impairment, olfactory dysfunction and fatigue. Nat Commun 2024; 15:4256. [PMID: 38762609 PMCID: PMC11102465 DOI: 10.1038/s41467-024-48651-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/08/2024] [Indexed: 05/20/2024] Open
Abstract
After contracting COVID-19, a substantial number of individuals develop a Post-COVID-Condition, marked by neurologic symptoms such as cognitive deficits, olfactory dysfunction, and fatigue. Despite this, biomarkers and pathophysiological understandings of this condition remain limited. Employing magnetic resonance imaging, we conduct a comparative analysis of cerebral microstructure among patients with Post-COVID-Condition, healthy controls, and individuals that contracted COVID-19 without long-term symptoms. We reveal widespread alterations in cerebral microstructure, attributed to a shift in volume from neuronal compartments to free fluid, associated with the severity of the initial infection. Correlating these alterations with cognition, olfaction, and fatigue unveils distinct affected networks, which are in close anatomical-functional relationship with the respective symptoms.
Collapse
Affiliation(s)
- Jonas A Hosp
- Department of Neurology and Clinical Neuroscience, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Marco Reisert
- Department of Diagnostic and Interventional Radiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Stereotactic and Functional Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andrea Dressing
- Department of Neurology and Clinical Neuroscience, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Freiburg Brain Imaging Center, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Veronika Götz
- Department of Internal Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Elias Kellner
- Department of Diagnostic and Interventional Radiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hansjörg Mast
- Department of Neuroradiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Susan Arndt
- Department of Otorhinolaryngology - Head and Neck Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cornelius F Waller
- Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dirk Wagner
- Department of Internal Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Siegbert Rieg
- Department of Internal Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Horst Urbach
- Department of Neuroradiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Cornelius Weiller
- Department of Neurology and Clinical Neuroscience, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nils Schröter
- Department of Neurology and Clinical Neuroscience, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexander Rau
- Department of Neuroradiology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
245
|
Vavougios GD, Mavridis T, Doskas T, Papaggeli O, Foka P, Hadjigeorgiou G. SARS-CoV-2-Induced Type I Interferon Signaling Dysregulation in Olfactory Networks Implications for Alzheimer's Disease. Curr Issues Mol Biol 2024; 46:4565-4579. [PMID: 38785545 PMCID: PMC11119810 DOI: 10.3390/cimb46050277] [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: 01/10/2024] [Revised: 04/15/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Type I interferon signaling (IFN-I) perturbations are major drivers of COVID-19. Dysregulated IFN-I in the brain, however, has been linked to both reduced cognitive resilience and neurodegenerative diseases such as Alzheimer's. Previous works from our group have proposed a model where peripheral induction of IFN-I may be relayed to the CNS, even in the absence of fulminant infection. The aim of our study was to identify significantly enriched IFN-I signatures and genes along the transolfactory route, utilizing published datasets of the nasal mucosa and olfactory bulb amygdala transcriptomes of COVID-19 patients. We furthermore sought to identify these IFN-I signature gene networks associated with Alzheimer's disease pathology and risk. Gene expression data involving the nasal epithelium, olfactory bulb, and amygdala of COVID-19 patients and transcriptomic data from Alzheimer's disease patients were scrutinized for enriched Type I interferon pathways. Gene set enrichment analyses and gene-Venn approaches were used to determine genes in IFN-I enriched signatures. The Agora web resource was used to identify genes in IFN-I signatures associated with Alzheimer's disease risk based on its aggregated multi-omic data. For all analyses, false discovery rates (FDR) <0.05 were considered statistically significant. Pathways associated with type I interferon signaling were found in all samples tested. Each type I interferon signature was enriched by IFITM and OAS family genes. A 14-gene signature was associated with COVID-19 CNS and the response to Alzheimer's disease pathology, whereas nine genes were associated with increased risk for Alzheimer's disease based on Agora. Our study provides further support to a type I interferon signaling dysregulation along the extended olfactory network as reconstructed herein, ranging from the nasal epithelium and extending to the amygdala. We furthermore identify the 14 genes implicated in this dysregulated pathway with Alzheimer's disease pathology, among which HLA-C, HLA-B, HLA-A, PSMB8, IFITM3, HLA-E, IFITM1, OAS2, and MX1 as genes with associated conferring increased risk for the latter. Further research into its druggability by IFNb therapeutics may be warranted.
Collapse
Affiliation(s)
- George D. Vavougios
- Department of Neurology, Medical School, University of Cyprus, Nicosia 1678, Cyprus
| | - Theodoros Mavridis
- Department of Neurology, Tallaght University Hospital (TUH)/The Adelaide and Meath Hospital, Dublin, Incorporating the National Children’s Hospital (AMNCH), D24 NR0A Dublin, Ireland;
| | | | - Olga Papaggeli
- Molecular Virology Laboratory, Hellenic Pasteur Institute, 115 21 Athens, Greece; (O.P.); (P.F.)
| | - Pelagia Foka
- Molecular Virology Laboratory, Hellenic Pasteur Institute, 115 21 Athens, Greece; (O.P.); (P.F.)
| | | |
Collapse
|
246
|
Dacosta-Aguayo R, Puig J, Lamonja-Vicente N, Carmona-Cervelló M, León-Gómez BB, Monté-Rubio G, López-Linfante VM, Zamora-Putin V, Montero-Alia P, Chacon C, Bielsa J, Moreno-Gabriel E, Garcia-Sierra R, Pachón A, Costa A, Mataró M, Prado JG, Martinez-Cáceres E, Mateu L, Massanella M, Violán C, Torán-Monserrat P. Reduced Cortical Thickness Correlates of Cognitive Dysfunction in Post-COVID-19 Condition: Insights from a Long-Term Follow-up. AJNR Am J Neuroradiol 2024; 45:647-654. [PMID: 38575319 PMCID: PMC11288549 DOI: 10.3174/ajnr.a8167] [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: 09/19/2023] [Accepted: 01/03/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND AND PURPOSE There is a paucity of data on long-term neuroimaging findings from individuals who have developed the post-coronavirus 2019 (COVID-19) condition. Only 2 studies have investigated the correlations between cognitive assessment results and structural MR imaging in this population. This study aimed to elucidate the long-term cognitive outcomes of participants with the post-COVID-19 condition and to correlate these cognitive findings with structural MR imaging data in the post-COVID-19 condition. MATERIALS AND METHODS A cohort of 53 participants with the post-COVID-19 condition underwent 3T brain MR imaging with T1 and FLAIR sequences obtained a median of 1.8 years after Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) infection. A comprehensive neuropsychological battery was used to assess several cognitive domains in the same individuals. Correlations between cognitive domains and whole-brain voxel-based morphometry were performed. Different ROIs from FreeSurfer were used to perform the same correlations with other neuroimaging features. RESULTS According to the Frascati criteria, more than one-half of the participants had deficits in the attentional (55%, n = 29) and executive (59%, n = 31) domains, while 40% (n = 21) had impairment in the memory domain. Only 1 participant (1.89%) showed problems in the visuospatial and visuoconstructive domains. We observed that reduced cortical thickness in the left parahippocampal region (t(48) = 2.28, P = .03) and the right caudal-middle-frontal region (t(48) = 2.20, P = .03) was positively correlated with the memory domain. CONCLUSIONS Our findings suggest that cognitive impairment in individuals with the post-COVID-19 condition is associated with long-term alterations in the structure of the brain. These macrostructural changes may provide insight into the nature of cognitive symptoms.
Collapse
Affiliation(s)
- Rosalia Dacosta-Aguayo
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
- Department of Clinical Psychology and Psychobiology (R.D.-A., M.M.), University of Barcelona, Barcelona, Spain. Institut de Neurociències, University of Barcelona, Barcelona, Spain. Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Josep Puig
- Comparative Medicine and Bioimaging Center (J.P., G.M.-R.), Germans Trias i Pujol Research Institute, Badalona, Spain
- Department of Radiology (IDI) (J.P.), IDIBGI Hospital Universitari de Girona Doctor Josep Trueta, Girona, Spain
| | - Noemi Lamonja-Vicente
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
- Multidisciplinary Research Group in Health and Society (N.L.-V., V.M.L.-L., P.M.-A., E.M.-G., R.G.-S., P.T.-M.), Institut Universitari d'Investigació en Atenció Primària Jordi Gol, Barcelona, Spain
| | - Meritxell Carmona-Cervelló
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
| | - Brenda Biaani León-Gómez
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
| | - Gemma Monté-Rubio
- Comparative Medicine and Bioimaging Center (J.P., G.M.-R.), Germans Trias i Pujol Research Institute, Badalona, Spain
| | - Victor M López-Linfante
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
- Multidisciplinary Research Group in Health and Society (N.L.-V., V.M.L.-L., P.M.-A., E.M.-G., R.G.-S., P.T.-M.), Institut Universitari d'Investigació en Atenció Primària Jordi Gol, Barcelona, Spain
- Palau-Solità Healthcare Centre (V.M.L.-L., V.Z.-P.), Palau-Solità Plegamans Institut Català de la Salut, Barcelona, Spain
- Department of Medicine (V.M.L.-L., E.M.-C.), Universitat Autònoma de Barcelona, Cerdanyola de Vallès, Bellaterra, Spain
| | - Valeria Zamora-Putin
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
- Palau-Solità Healthcare Centre (V.M.L.-L., V.Z.-P.), Palau-Solità Plegamans Institut Català de la Salut, Barcelona, Spain
| | - Pilar Montero-Alia
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
- Multidisciplinary Research Group in Health and Society (N.L.-V., V.M.L.-L., P.M.-A., E.M.-G., R.G.-S., P.T.-M.), Institut Universitari d'Investigació en Atenció Primària Jordi Gol, Barcelona, Spain
| | - Carla Chacon
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
- Grup de REcerca en Impacte de les Malalties Cròniques i les seves Trajectòries (C.C., C.V.), Institut Universitari d'Investigació en Atenció Primària Jordi Gol; Barcelona, Spain. Germans Trias i Pujol Research Institute, Badalona, Spain
| | - Jofre Bielsa
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
| | - Eduard Moreno-Gabriel
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
- Multidisciplinary Research Group in Health and Society (N.L.-V., V.M.L.-L., P.M.-A., E.M.-G., R.G.-S., P.T.-M.), Institut Universitari d'Investigació en Atenció Primària Jordi Gol, Barcelona, Spain
- Department of Social Psychology (E.M.-G.), Universitat Autònoma de Barcelona, Cerdanyola de Vallès, Bellaterra, Spain
| | - Rosa Garcia-Sierra
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
- Multidisciplinary Research Group in Health and Society (N.L.-V., V.M.L.-L., P.M.-A., E.M.-G., R.G.-S., P.T.-M.), Institut Universitari d'Investigació en Atenció Primària Jordi Gol, Barcelona, Spain
- Nursing Department, Faculty of Medicine (R.G.-S.), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alba Pachón
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
| | - Anna Costa
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
| | - Maria Mataró
- Department of Clinical Psychology and Psychobiology (R.D.-A., M.M.), University of Barcelona, Barcelona, Spain. Institut de Neurociències, University of Barcelona, Barcelona, Spain. Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Julia G Prado
- IrsiCaixa-AIDS Research Institute and Germans Trias i Pujol Health Research Institute (J.G.P., M.M.), Can Ruti Campus, Badalona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (J.G.-., L.M., M.M.), Instituto de Salud Carlos III, Madrid, Spain
- Germans Trias i Pujol Research Institute (J.G.P., E.M.-C., L.M., M.M., C.V., P.T.-M.), Can Ruti Campus, Badalona, Spain
| | - Eva Martinez-Cáceres
- Department of Medicine (V.M.L.-L., E.M.-C.), Universitat Autònoma de Barcelona, Cerdanyola de Vallès, Bellaterra, Spain
- Germans Trias i Pujol Research Institute (J.G.P., E.M.-C., L.M., M.M., C.V., P.T.-M.), Can Ruti Campus, Badalona, Spain
- Immunology Department (E.M.-C.), FOCIS Center of Excellence, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Immunology Division, Laboratori Clinic Metropolitana Nord (E.M.-C.), Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Lourdes Mateu
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (J.G.-., L.M., M.M.), Instituto de Salud Carlos III, Madrid, Spain
- Germans Trias i Pujol Research Institute (J.G.P., E.M.-C., L.M., M.M., C.V., P.T.-M.), Can Ruti Campus, Badalona, Spain
- Infectious Diseases Department (L.M.), Fight against AIDS Foundation (FLS), Germans Trias I Pujol Hospital, Can Ruti Campus, Badalona, Spain
- Red Española de Investigación en Covid Persisitente (L.M., M.M.), European Innovation and Collaboration Programme, Badalona, Spain
| | - Marta Massanella
- IrsiCaixa-AIDS Research Institute and Germans Trias i Pujol Health Research Institute (J.G.P., M.M.), Can Ruti Campus, Badalona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (J.G.-., L.M., M.M.), Instituto de Salud Carlos III, Madrid, Spain
- Germans Trias i Pujol Research Institute (J.G.P., E.M.-C., L.M., M.M., C.V., P.T.-M.), Can Ruti Campus, Badalona, Spain
- Red Española de Investigación en Covid Persisitente (L.M., M.M.), European Innovation and Collaboration Programme, Badalona, Spain
| | - Concepción Violán
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
- Grup de REcerca en Impacte de les Malalties Cròniques i les seves Trajectòries (C.C., C.V.), Institut Universitari d'Investigació en Atenció Primària Jordi Gol; Barcelona, Spain. Germans Trias i Pujol Research Institute, Badalona, Spain
- Germans Trias i Pujol Research Institute (J.G.P., E.M.-C., L.M., M.M., C.V., P.T.-M.), Can Ruti Campus, Badalona, Spain
- Red de Investigación en Cronicidad (C.V.), Atención Primaria y Prevención y Promoción de la Salud (RICAPPS), Instituto de Salud Carlos III, Madrid, Spain
- Universitat Autónoma de Barcelona (C.V.), Bellaterra, Spain
| | - Pere Torán-Monserrat
- From the Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol) (R.D.-A., N.L.-V., M.C.-C., B.B.L.-G., V.M.L.-L., V.Z.-P., P.M.-A., C.C., J.B., E.M.-G., R.G.-S., A.P., A.C., C.V., P.T.-M.), Unitat de Suport a la Recerca Metropolitana Nord, Mataró, Spain
- Multidisciplinary Research Group in Health and Society (N.L.-V., V.M.L.-L., P.M.-A., E.M.-G., R.G.-S., P.T.-M.), Institut Universitari d'Investigació en Atenció Primària Jordi Gol, Barcelona, Spain
- Germans Trias i Pujol Research Institute (J.G.P., E.M.-C., L.M., M.M., C.V., P.T.-M.), Can Ruti Campus, Badalona, Spain
- Department of Medicine (P.T.-M.), Faculty of Medicine, Universitat de Girona, Girona, Spain
| |
Collapse
|
247
|
Lengacher NA, Tomlinson JJ, Jochum AK, Franz J, Hasan Ali O, Flatz L, Jochum W, Penninger J, Stadelmann C, Woulfe JM, Schlossmacher MG. Neuropathological assessment of the olfactory bulb and tract in individuals with COVID-19. Acta Neuropathol Commun 2024; 12:70. [PMID: 38698465 PMCID: PMC11067107 DOI: 10.1186/s40478-024-01761-8] [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: 12/19/2023] [Accepted: 03/17/2024] [Indexed: 05/05/2024] Open
Abstract
The majority of patients with Parkinson disease (PD) experience a loss in their sense of smell and accumulate insoluble α-synuclein aggregates in their olfactory bulbs (OB). Subjects affected by a SARS-CoV-2-linked illness (COVID-19) also frequently experience hyposmia. We previously postulated that microglial activation as well as α-synuclein and tau misprocessing can occur during host responses following microbial encounters. Using semiquantitative measurements of immunohistochemical signals, we examined OB and olfactory tract specimens collected serially at autopsies between 2020 and 2023. Deceased subjects comprised 50 adults, which included COVID19 + patients (n = 22), individuals with Lewy body disease (e.g., PD; dementia with Lewy bodies (n = 6)), Alzheimer disease (AD; n = 3), and other neurodegenerative disorders (e.g., progressive supranuclear palsy (n = 2); multisystem atrophy (n = 1)). Further, we included neurologically healthy controls (n = 9), and added subjects with an inflammation-rich brain disorder as neurological controls (NCO; n = 7). When probing for microglial and histiocytic reactivity in the anterior olfactory nuclei (AON) by anti-CD68 immunostaining, scores were consistently elevated in NCO and AD cases. In contrast, microglial signals on average were not significantly altered in COVID19 + patients relative to healthy controls, although anti-CD68 reactivity in their OB and tracts declined with progression in age. Mild-to-moderate increases in phospho-α-synuclein and phospho-tau signals were detected in the AON of tauopathy- and synucleinopathy-afflicted brains, respectively, consistent with mixed pathology, as described by others. Lastly, when both sides were available for comparison in our case series, we saw no asymmetry in the degree of pathology of the left versus right OB and tracts. We concluded from our autopsy series that after a fatal course of COVID-19, microscopic changes in the rostral, intracranial portion of the olfactory circuitry -when present- reflected neurodegenerative processes seen elsewhere in the brain. In general, microglial reactivity correlated best with the degree of Alzheimer's-linked tauopathy and declined with progression of age in COVID19 + patients.
Collapse
Affiliation(s)
- Nathalie A Lengacher
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Julianna J Tomlinson
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Ann-Kristin Jochum
- Institute of Pathology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Jonas Franz
- Neuropathology Institute, University of Goettingen Medical Centre, Goettingen, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Omar Hasan Ali
- Department of Life Sciences, University of British Columbia, Vancouver, BC, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Wolfram Jochum
- Institute of Pathology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Josef Penninger
- Department of Life Sciences, University of British Columbia, Vancouver, BC, Canada
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Christine Stadelmann
- Neuropathology Institute, University of Goettingen Medical Centre, Goettingen, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - John M Woulfe
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Ottawa, ON, Canada.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| | - Michael G Schlossmacher
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
| |
Collapse
|
248
|
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
|
249
|
Zheng ZS, Simonian N, Wang J, Rosario ER. Transcutaneous vagus nerve stimulation improves Long COVID symptoms in a female cohort: a pilot study. Front Neurol 2024; 15:1393371. [PMID: 38756213 PMCID: PMC11097097 DOI: 10.3389/fneur.2024.1393371] [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: 02/29/2024] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
Background Long COVID, also known as Post-COVID-19 syndrome, is characterized by multisystemic symptoms that persists for weeks to years beyond acute infection. It disproportionately affects women and those with pre-existing anxiety/depression, conditions more prevalent in females. The vagus nerve, with its extensive innervation and regulation of critical bodily functions, has become a focal point for therapeutic interventions. Transcutaneous vagus nerve stimulation (t-VNS) has emerged as a promising non-invasive treatment for COVID-19 conditions. Methods This pilot study assessed the efficacy of t-VNS in 24 female Long COVID patients (45.8 ± 11.7 years old; 20.2 ± 7.1 months since infection), who underwent a 10-day t-VNS intervention at home (30 min/session, twice a day). Cognition was considered the primary outcome, with anxiety, depression, sleep, fatigue, and smell as secondary outcomes. Outcomes were measured at baseline, post-intervention, and 1-month follow-up. Results Significant improvements were observed in various cognitive functions, anxiety, depression, and sleep at post-intervention, with benefits remaining or progressing at 1-month follow-up. Improvements in fatigue were delayed, reaching statistical significance at 1-month follow-up compared to baseline. No significant changes were noted in olfactory performance. Conclusion This pilot study provides preliminary evidence supporting the potential of t-VNS as a therapeutic intervention for female Long COVID patients. The encouraging results justify further rigorous investigation through larger, randomized controlled trials to confirm the efficacy of t-VNS, assess its generalizability to male cohorts, and explore biological markers to inform personalized treatment approaches. Our findings support the allocation of resources to conduct such trials and advance the understanding of t-VNS as a potential treatment for Long COVID.
Collapse
Affiliation(s)
- Zhong Sheng Zheng
- Research Institute, Casa Colina Hospital and Centers for Healthcare, Pomona, CA, United States
| | - Ninette Simonian
- Institute of Advanced Consciousness Studies, Santa Monica, CA, United States
| | - Jing Wang
- Research Institute, Casa Colina Hospital and Centers for Healthcare, Pomona, CA, United States
| | - Emily R. Rosario
- Research Institute, Casa Colina Hospital and Centers for Healthcare, Pomona, CA, United States
| |
Collapse
|
250
|
Annesley SJ, Missailidis D, Heng B, Josev EK, Armstrong CW. Unravelling shared mechanisms: insights from recent ME/CFS research to illuminate long COVID pathologies. Trends Mol Med 2024; 30:443-458. [PMID: 38443223 DOI: 10.1016/j.molmed.2024.02.003] [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: 11/20/2023] [Revised: 01/29/2024] [Accepted: 02/13/2024] [Indexed: 03/07/2024]
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating chronic illness often triggered by an initiating acute event, mainly viral infections. The transition from acute to chronic disease remains unknown, but interest in this phenomenon has escalated since the COVID-19 pandemic and the post-COVID-19 illness, termed 'long COVID' (LC). Both ME/CFS and LC share many clinical similarities. Here, we present recent findings in ME/CFS research focussing on proposed disease pathologies shared with LC. Understanding these disease pathologies and how they influence each other is key to developing effective therapeutics and diagnostic tests. Given that ME/CFS typically has a longer disease duration compared with LC, with symptoms and pathologies evolving over time, ME/CFS may provide insights into the future progression of LC.
Collapse
Affiliation(s)
- Sarah J Annesley
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, VIC, Australia.
| | - Daniel Missailidis
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, VIC, Australia
| | - Benjamin Heng
- Macquarie Medical School, Faculty of Medicine, Human and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Elisha K Josev
- Neurodisability & Rehabilitation, Clinical Sciences, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, VIC, Australia; Mercy Hospital for Women, Heidelberg, VIC, Australia
| | - Christopher W Armstrong
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|