1
|
Nguyen THV, Ferron F, Murakami K. Neurotoxic Implications of Human Coronaviruses in Neurodegenerative Diseases: A Perspective from Amyloid Aggregation. ACS Chem Biol 2025; 20:983-992. [PMID: 40272376 DOI: 10.1021/acschembio.5c00153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Human coronaviruses (HCoVs) include seven species: HCoV-229E, HCoV-NL63, HCoV-OC43, HCoV-HKU1, MERS-CoV, SARS-CoV-1, and SARS-CoV-2. The last three, classified as Betacoronaviruses, are highly transmissible and have caused severe pandemics. HCoV infections primarily affect the respiratory system, leading to symptoms such as dry cough, fever, and breath shortness, which can progress to acute respiratory failure and death. Beyond respiratory effects, increasing evidence links HCoVs to neurological dysfunction. However, distinguishing direct neural complications from preexisting disorders, particularly in the elderly, remains challenging. This study examines the association between HCoVs and neurodegenerative diseases like Alzheimer disease, Parkinson disease, Lewy body dementia, amyotrophic lateral sclerosis, and Creutzfeldt-Jakob disease. It also presents the long-term neurological effects of HCoV infections and their differential impact across age groups and sexes. A key aspect of this study is the investigation of the sequence and structural similarities between amyloidogenic and HCoV spike proteins, which can provide insights into potential neuropathomechanisms.
Collapse
Affiliation(s)
- Thi Hong Van Nguyen
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | - Francois Ferron
- Aix Marseille Univ, CNRS-Architecture et Fonction des Macromolécules Biologiques (AFMB) UMR7257, Marseille 13288, France
- European Virus Bioinformatics Center, Jena 07743, Germany
| | - Kazuma Murakami
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| |
Collapse
|
2
|
Posa A. Spike protein-related proteinopathies: A focus on the neurological side of spikeopathies. Ann Anat 2025; 260:152662. [PMID: 40254264 DOI: 10.1016/j.aanat.2025.152662] [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: 02/24/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND The spike protein (SP) is an outward-projecting transmembrane glycoprotein on viral surfaces. SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2), responsible for COVID-19 (Coronavirus Disease 2019), uses SP to infect cells that express angiotensin converting enzyme 2 (ACE2) on their membrane. Remarkably, SP has the ability to cross the blood-brain barrier (BBB) into the brain and cause cerebral damage through various pathomechanisms. To combat the COVID-19 pandemic, novel gene-based products have been used worldwide to induce human body cells to produce SP to stimulate the immune system. This artificial SP also has a harmful effect on the human nervous system. STUDY DESIGN Narrative review. OBJECTIVE This narrative review presents the crucial role of SP in neurological complaints after SARS-CoV-2 infection, but also of SP derived from novel gene-based anti-SARS-CoV-2 products (ASP). METHODS Literature searches using broad terms such as "SARS-CoV-2", "spike protein", "COVID-19", "COVID-19 pandemic", "vaccines", "COVID-19 vaccines", "post-vaccination syndrome", "post-COVID-19 vaccination syndrome" and "proteinopathy" were performed using PubMed. Google Scholar was used to search for topic-specific full-text keywords. CONCLUSIONS The toxic properties of SP presented in this review provide a good explanation for many of the neurological symptoms following SARS-CoV-2 infection and after injection of SP-producing ASP. Both SP entities (from infection and injection) interfere, among others, with ACE2 and act on different cells, tissues and organs. Both SPs are able to cross the BBB and can trigger acute and chronic neurological complaints. Such SP-associated pathologies (spikeopathies) are further neurological proteinopathies with thrombogenic, neurotoxic, neuroinflammatory and neurodegenerative potential for the human nervous system, particularly the central nervous system. The potential neurotoxicity of SP from ASP needs to be critically examined, as ASPs have been administered to millions of people worldwide.
Collapse
Affiliation(s)
- Andreas Posa
- University Clinics and Outpatient Clinics for Radiology, Neuroradiology and Neurology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Straße 40, Halle 06120, Germany.
| |
Collapse
|
3
|
Kruger A, Joffe D, Lloyd-Jones G, Khan MA, Šalamon Š, Laubscher GJ, Putrino D, Kell DB, Pretorius E. Vascular Pathogenesis in Acute and Long COVID: Current Insights and Therapeutic Outlook. Semin Thromb Hemost 2025; 51:256-271. [PMID: 39348850 PMCID: PMC11906225 DOI: 10.1055/s-0044-1790603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
Long coronavirus disease 2019 (COVID-19)-a postacute consequence of severe acute respiratory syndrome coronavirus 2 infection-manifests with a broad spectrum of relapsing and remitting or persistent symptoms as well as varied levels of organ damage, which may be asymptomatic or present as acute events such as heart attacks or strokes and recurrent infections, hinting at complex underlying pathogenic mechanisms. Central to these symptoms is vascular dysfunction rooted in thrombotic endothelialitis. We review the scientific evidence that widespread endothelial dysfunction (ED) leads to chronic symptomatology. We briefly examine the molecular pathways contributing to endothelial pathology and provide a detailed analysis of how these cellular processes underpin the clinical picture. Noninvasive diagnostic techniques, such as flow-mediated dilation and peripheral arterial tonometry, are evaluated for their utility in identifying ED. We then explore mechanistic, cellular-targeted therapeutic interventions for their potential in treating ED. Overall, we emphasize the critical role of cellular health in managing Long COVID and highlight the need for early intervention to prevent long-term vascular and cellular dysfunction.
Collapse
Affiliation(s)
- Arneaux Kruger
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - David Joffe
- Respiratory and Sleep Medicine, Royal North Shore Hospital, Sydney, Australia
- World Health Network, Cambridge, Massachusetts
| | - Graham Lloyd-Jones
- Department of Radiology, Salisbury District Hospital, Salisbury NHS Foundation Trust, United Kingdom
| | - Muhammed Asad Khan
- World Health Network, Cambridge, Massachusetts
- Directorate of Respiratory Medicine, Manchester University Hospitals, Wythenshawe Hospital, Manchester, United Kingdom
| | | | | | - David Putrino
- Respiratory and Sleep Medicine, Royal North Shore Hospital, Sydney, Australia
- Department of Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- World Health Network, Cambridge, Massachusetts
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
4
|
Satyanarayanan SK, Yip TF, Han Z, Zhu H, Qin D, Lee SMY. Role of toll-like receptors in post-COVID-19 associated neurodegenerative disorders? Front Med (Lausanne) 2025; 12:1458281. [PMID: 40206484 PMCID: PMC11979212 DOI: 10.3389/fmed.2025.1458281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 03/12/2025] [Indexed: 04/11/2025] Open
Abstract
In the intricate realm of interactions between hosts and pathogens, Toll-like receptors (TLRs), which play a crucial role in the innate immune response, possess the ability to identify specific molecular signatures. This includes components originating from pathogens such as SARS-CoV-2, as well as the resulting damage-associated molecular patterns (DAMPs), the endogenous molecules released after cellular damage. A developing perspective suggests that TLRs play a central role in neuroinflammation, a fundamental factor in neurodegenerative conditions like Alzheimer's and Parkinson's disease (PD). This comprehensive review consolidates current research investigating the potential interplay between TLRs, their signaling mechanisms, and the processes of neurodegeneration following SARS-CoV-2 infection with an aim to elucidate the involvement of TLRs in the long-term neurological complications of COVID-19 and explore the potential of targeting TLRs as a means of implementing intervention strategies for the prevention or treatment of COVID-19-associated long-term brain outcomes.
Collapse
Affiliation(s)
- Senthil Kumaran Satyanarayanan
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, Hong Kong SAR, China
| | - Tsz Fung Yip
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zixu Han
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, Hong Kong SAR, China
| | - Huachen Zhu
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Dajiang Qin
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, Hong Kong SAR, China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Suki Man Yan Lee
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong Science Park, Hong Kong, Hong Kong SAR, China
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
5
|
Li X, Yang X, Liu B, Luo J, Chen H, Chen J, Wang B, Hu Y, Su Z, Qin X. One-pot synthesis of multifunctional sliver nanoparticles with controlled size for sensitive colorimetric and electrochemiluminescent immunoassay of SARS-CoV-2. Anal Chim Acta 2025; 1342:343684. [PMID: 39919859 DOI: 10.1016/j.aca.2025.343684] [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: 11/18/2024] [Revised: 01/06/2025] [Accepted: 01/16/2025] [Indexed: 02/09/2025]
Abstract
Silver nanoparticles (AgNPs) have great potential in a broad range of applications because of their biochemical functionality, unique physical and optical properties. However, it is still a great challenge to synthesize small-size AgNPs with good stability and high performance for biosensors. In this work, triethanolamine and polyacrylic acid modified AgNPs (TEOA@AgNPs-PAA) with controllable size and high catalytic activity for sensitive detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are reported. Monodisperse TEOA@AgNPs-PAA are prepared by the one-pot synthesis using the TEOA and PAA as reducing agent and surfactant, respectively. The size of TEOA@AgNPs-PAA (the average size of 8.65 nm) is 6 times smaller than that of the TEOA@AgNPs (52 ± 1.5 nm) without PAA. The as-prepared TEOA@AgNPs-PAA possess catalytic activity and present mimicking property of horseradish peroxidase, which are employed to fabricate colorimetric biosensors by catalyzing the reaction between H2O2 and 3,3',5,5'-tetramethylbenzidine (TMB) to produce blue oxTMB for sensitive detection of SARS-CoV-2 spiking proteins. Significantly, the small-size TEOA@AgNPs-PAA can catalyze electroreduction process of K2S2O8 to enhance the cathodic ECL signal, and their surface cap abundant TEOA molecules, which can also act as a coreactant to enhance the anodic ECL of Ru(bpy)32+. Under optimal conditions, the fabricate immunosensors for anodic and cathodic ECL determination of SARS-CoV-2 present the detection limits of 9.2 fg/mL and 14.3 fg/mL (S/N = 3), respectively. This work exhibits a promising novel strategy for the development of multifunctional AgNPs as an efficient sensing platform for the clinical diagnosis and biosensing application.
Collapse
Affiliation(s)
- Xiangyu Li
- School of Chemistry and Material Science, Hunan Agricultural University, Changsha, 410128, China
| | - Xiaolan Yang
- School of Chemistry and Material Science, Hunan Agricultural University, Changsha, 410128, China
| | - Bo Liu
- School of Chemistry and Material Science, Hunan Agricultural University, Changsha, 410128, China
| | - Jiali Luo
- School of Chemistry and Material Science, Hunan Agricultural University, Changsha, 410128, China
| | - Huquan Chen
- School of Chemistry and Material Science, Hunan Agricultural University, Changsha, 410128, China
| | - Jin Chen
- School of Chemistry and Material Science, Hunan Agricultural University, Changsha, 410128, China
| | - Birui Wang
- School of Chemistry and Material Science, Hunan Agricultural University, Changsha, 410128, China
| | - Yue Hu
- Bairuopu Town Center Health Center, Changsha, 410206, China
| | - Zhaohong Su
- School of Chemistry and Material Science, Hunan Agricultural University, Changsha, 410128, China
| | - Xiaoli Qin
- School of Chemistry and Material Science, Hunan Agricultural University, Changsha, 410128, China.
| |
Collapse
|
6
|
Frigori RB. Evolution and pathogenicity of SARS-CoVs: A microcanonical analysis of receptor-binding motifs. Phys Rev E 2025; 111:034401. [PMID: 40247563 DOI: 10.1103/physreve.111.034401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 02/07/2025] [Indexed: 04/19/2025]
Abstract
The rapid evolution and global impact of coronaviruses, notably SARS-CoV-1 and SARS-CoV-2, underscore the importance of understanding their molecular mechanisms in detail. This study focuses on the receptor-binding motif (RBM) within the spike protein of these viruses, a critical element for viral entry through interaction with the ACE2 receptor. We investigate the sequence variations in the RBM across SARS-CoV-1, SARS-CoV-2, and its early variants of concern (VOCs). Utilizing multicanonical simulations and microcanonical analysis, we examine how these variations influence the folding dynamics, thermostability, and solubility of the RBMs. Our methodology includes calculating the density of states (DoS) to identify structural phase transitions and assess thermodynamic properties. Furthermore, we solve the Poisson-Boltzmann equation to model the solubility of the RBMs in aqueous environments. This methodology is expected to elucidate structural and functional differences in viral evolution and pathogenicity, likely improving targeted treatments and vaccines.
Collapse
Affiliation(s)
- Rafael B Frigori
- Universidade Tecnológica Federal do Paraná, Rua Cristo Rei 19, CEP 85902-490, Toledo (PR), Brazil
| |
Collapse
|
7
|
Otzen DE, Peña-Díaz S, Widmann J, Daugberg AOH, Zhang Z, Jiang Y, Mittal C, Dueholm MKD, Louros N, Wang H, Javed I. Interactions between pathological and functional amyloid: A match made in Heaven or Hell? Mol Aspects Med 2025; 103:101351. [PMID: 40024004 DOI: 10.1016/j.mam.2025.101351] [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: 01/14/2025] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
The amyloid state of proteins occurs in many different contexts in Nature and in modern society, ranging from the pathological kind (neurodegenerative diseases and amyloidosis) via man-made forms (food processing and - to a much smaller extent - protein biologics) to functional versions (bacterial biofilm, peptide hormones and signal transmission). These classes all come together in the human body which endogenously produces amyloidogenic protein able to form pathological human amyloid (PaHA), hosts a microbiome which continuously makes functional bacterial amyloid (FuBA) and ingests food which can contain amyloid. This can have grave consequences, given that PaHA can spread throughout the body in a "hand-me-down" fashion from cell to cell through small amyloid fragments, which can kick-start growth of new amyloid wherever they encounter monomeric amyloid precursors. Amyloid proteins can also self- and cross-seed across dissimilar peptide sequences. While it is very unlikely that ingested amyloid plays a role in this crosstalk, FuBA-PaHA interactions are increasingly implicated in vivo amyloid propagation. We are now in a position to understand the structural and bioinformatic basis for this cross-talk, thanks to the very recently obtained atomic-level structures of the two major FuBAs CsgA (E. coli) and FapC (Pseudomonas). While there are many reports of homology-driven heterotypic interactions between different PaHA, the human proteome does not harbor significant homology to CsgA and FapC. Yet we and others have uncovered significant cross-stimulation (and in some cases inhibition) of FuBA and PaHA both in vitro and in vivo, which we here rationalize based on structure and sequence. These interactions have important consequences for the transmission and development of neurodegenerative diseases, not least because FuBA and PaHA can come into contact via the gut-brain interface, recurrent infections with microbes and potentially even through invasive biofilm in the brain. Whether FuBA and PaHA first interact in the gut or the brain, they can both stimulate and block each other's aggregation as well as trigger inflammatory responses. The microbiome may also affect amyloidogenesis in other ways, e.g. through their own chaperones which recognize and block growth of both PaHA and FuBA as we show both experimentally and computationally. Heterotypic interactions between and within PaHA and FuBA both in vitro and in vivo are a vital part of the amyloid phenomenon and constitute a vibrant and exciting frontier for future research.
Collapse
Affiliation(s)
- Daniel E Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark.
| | - Samuel Peña-Díaz
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark.
| | - Jeremias Widmann
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark
| | - Anders Ogechi Hostrup Daugberg
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, 9220, Aalborg OE, Denmark
| | - Zhefei Zhang
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark; Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Clinical Laboratory Center, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Yanting Jiang
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Clinical Laboratory Center, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Chandrika Mittal
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark; Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Clinical Laboratory Center, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Guangxi Zhuang Autonomous Region, Nanning, 530021, China
| | - Morten K D Dueholm
- Center for Microbial Communities, Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, 9220, Aalborg OE, Denmark
| | - Nikolaos Louros
- Center for Alzheimer's and Neurodegenerative Diseases, Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA; Department of Biophysics, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Huabing Wang
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Clinical Laboratory Center, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Guangxi Zhuang Autonomous Region, Nanning, 530021, China; Jiangsu Fuyuda Food Products Co., Ltd, Qinyou Road 88, Gaoyou City, Jiangsu Province, 225600, China.
| | - Ibrahim Javed
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Qld, 4072, Australia.
| |
Collapse
|
8
|
Adamiec-Mroczek J, Kluz J, Chwałek S, Rabczyński M, Gostomska-Pampuch K, Lewandowski Ł, Misiuk-Hojło M, Ponikowska B, Chourasia G, Dumas I, Gamian A, Fiodorenko-Dumas Ż, Konopska B, Gola A, Konikowska K, Strub D, Bronowicka-Szydełko A, Madziarska K. Development of an enzyme-linked immunosorbent assay (ELISA) for determining neutrophil elastase (NE) - a potential useful marker of multi-organ damage observed in COVID-19 and post-Covid-19 (PCS). Front Mol Biosci 2025; 12:1542898. [PMID: 40070691 PMCID: PMC11893405 DOI: 10.3389/fmolb.2025.1542898] [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: 12/10/2024] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
Background The ongoing post-COVID-19 syndrome (PCS) epidemic, causing complications of diverse etiology, necessitates the search for new diagnostic markers and the development of widely accessible methods for their detection. This would enable the prognosis of PCS progression and faster implementation of targeted treatments. One potential marker is neutrophil elastase (NE), whose elevated levels in the blood during PCS may result from organ damage caused by increased secretion of severe inflammatory mediators or amyloidosis resulting from the interaction of NE with SARS-CoV-2. The aim of this publication is to present a step-by-step method for designing an enzymatic ELISA test, enabling the quantitative assessment of NE in the blood serum of patients. Methods NE was measured using the designed ELISA test. Results The study outlines all the steps necessary for designing and optimizing the ELISA test, including the selection of standards, primary and secondary antibodies, and their dilutions. Using the test, elevated NE levels were demonstrated in patients with advanced-stage diabetic nephropathy after symptomatic COVID-19, compared to a relative group of patients sampled before COVID-19. Conclusion The undertaken efforts enabled the development of a test with high performance parameters (initially set sensitivity: ≥40 pg/μL; intra-assay precision: 7%; inter-assay precision <20%). No significant cross-reactivity with other tested proteins was observed. Serial dilution of plasma samples resulted in a proportional decrease in signal intensity.
Collapse
Affiliation(s)
| | - Joanna Kluz
- Clinical Department of Diabetology, Hypertension and Internal Disease, Wroclaw Medical University, Wroclaw, Poland
| | - Sandra Chwałek
- Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Maciej Rabczyński
- Clinical Department of Diabetology, Hypertension and Internal Disease, Wroclaw Medical University, Wroclaw, Poland
| | | | - Łukasz Lewandowski
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, Wroclaw, Poland
| | - Marta Misiuk-Hojło
- Clinical Department of Ophthalmology, Wroclaw Medical University, Wroclaw, Poland
| | - Beata Ponikowska
- Department of Physiology and Pathophysiology, Wroclaw Medical University, Wroclaw, Poland
| | - Goutam Chourasia
- Department and Clinic of Emergency Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Ilias Dumas
- Department of Clinical Physiotherapy and Rehabilitation, Wroclaw Medical University, Wroclaw, Poland
| | - Andrzej Gamian
- Laboratory of Medical Microbiology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Żanna Fiodorenko-Dumas
- Department of Clinical Physiotherapy and Rehabilitation, Wroclaw Medical University, Wroclaw, Poland
| | - Bogusława Konopska
- Diagnostic Scientific and Teaching Laboratory, Department of Laboratory Diagnostics, Wroclaw Medical University, Wroclaw, Poland
| | - Agnieszka Gola
- Department of Physical Chemistry and Biophysics, Wroclaw Medical University, Wroclaw, Poland
| | - Klaudia Konikowska
- Department of Dietetics and Bromatology, Wroclaw Medical University, Wroclaw, Poland
| | - Daniel Strub
- Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | | | - Katarzyna Madziarska
- Clinical Department of Diabetology, Hypertension and Internal Disease, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
9
|
Baig AM, Rosko S, Jaeger B, Gerlach J. Strategic Inhibition of CHRM Autoantibodies: Molecular Insights and Therapeutic Potentials in Long COVID. J Med Chem 2025; 68:2089-2094. [PMID: 39836023 DOI: 10.1021/acs.jmedchem.4c00691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
In addition to the conventional symptoms reported for COVID-19, it is becoming increasingly clear that patients with long COVID are exhibiting new symptoms due to the emergence of autoantibodies against G-protein-coupled receptors, among which human muscarinic cholinergic receptors (CHRMs) have been prominently reported. With a chronic condition such as long COVID, additional symptoms caused by anti-CHRM autoantibodies (AAbs) have proven to be an added burden on these patients. The origins of these AAbs, their interactions with, and effects on the function of neural and non-neural cells within the nervous system have remained unknown. Furthermore, the specific symptom complex to which they contribute has not been clearly understood. In this context, we address these issues here and suggest methods to combat the autoantibodies that contribute to neurological symptoms in long COVID.
Collapse
Affiliation(s)
- Abdul Mannan Baig
- Department of Long-COVID and Neuro-COVID Research, Klinik St. Georg, 83043 Bad Aibling, Germany
| | - Sandy Rosko
- Department of Long-COVID and Chronic Illness, West Lakes, Adelaide 5021, Australia
| | - Beate Jaeger
- Department of Long-COVID and Internal Medicine, Klinik St. Georg, 83043 Bad Aibling, Germany
| | - Joachim Gerlach
- Research and Development, Health-Shield, Vedicinals-9, 40764 Langenfeld, Germany
| |
Collapse
|
10
|
Narayanan SN, Padiyath S, Chandrababu K, Raj L, P S BC, Ninan GA, Sivadasan A, Jacobs AR, Li YW, Bhaskar A. Neurological, psychological, psychosocial complications of long-COVID and their management. Neurol Sci 2025; 46:1-23. [PMID: 39516425 PMCID: PMC11698801 DOI: 10.1007/s10072-024-07854-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024]
Abstract
Since it first appeared, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has had a significant and lasting negative impact on the health and economies of millions of individuals all over the globe. At the level of individual health too, many patients are not recovering fully and experiencing a long-term condition now commonly termed 'long-COVID'. Long-COVID is a collection of symptoms which must last more than 12 weeks following initial COVID infection, and which cannot be adequately explained by alternate diagnoses. The neurological and psychosocial impact of long-COVID is itself now a global health crisis and therefore preventing, diagnosing, and managing these patients is of paramount importance. This review focuses primarily on: neurological functioning deficits; mental health impacts; long-term mood problems; and associated psychosocial issues, among patients suffering from long-COVID with an eye towards the neurological basis of these symptoms. A concise account of the clinical relevance of the neurological and psychosocial impacts of long-COVID, the effects on long-term morbidity, and varied approaches in managing patients with significant chronic neurological symptoms and conditions was extracted from the literature, analysed and reported. A comprehensive account of plausible pathophysiological mechanisms involved in the development of long-COVID, its management, and future research needs have been discussed.
Collapse
Affiliation(s)
- Sareesh Naduvil Narayanan
- Department of Physiology, School of Medicine and Dentistry, AUC-UK Track, University of Central Lancashire, Preston, UK.
| | - Sreeshma Padiyath
- Department of Microbiology, School of Medicine and Dentistry, AUC-UK Track, University of Central Lancashire, Preston, UK
| | - Krishnapriya Chandrababu
- Centre for Neuroscience, Department of Biotechnology, Cochin University of Science and Technology (CUSAT), Kochi, India
| | - Lima Raj
- Department of Psychology, Sree Sankaracharya University of Sanskrit, Kalady, India
| | - Baby Chakrapani P S
- Centre for Neuroscience, Department of Biotechnology, Cochin University of Science and Technology (CUSAT), Kochi, India
- Centre for Excellence in Neurodegeneration and Brain Health (CENABH), Cochin University of Science and Technology (CUSAT), Kochi, India
| | | | - Ajith Sivadasan
- Department of Neurology, Christian Medical College (CMC), Vellore, India
| | - Alexander Ryan Jacobs
- School of Medicine and Dentistry, AUC-UK Track, University of Central Lancashire, Preston, UK
| | - Yan Wa Li
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Anand Bhaskar
- Department of Physiology, Christian Medical College (CMC), Vellore, India
| |
Collapse
|
11
|
St Dollente Mesias V, Zhang J, Zhu H, Dai X, Li J, Huang J. Distinct Effects of SARS-CoV-2 Protein Segments on Structural Stability, Amyloidogenic Potential, and α-Synuclein Aggregation. Chembiochem 2024; 25:e202400598. [PMID: 39480569 DOI: 10.1002/cbic.202400598] [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: 07/15/2024] [Revised: 10/14/2024] [Accepted: 10/27/2024] [Indexed: 11/02/2024]
Abstract
Amyloidosis is characterized by the abnormal accumulation of misfolded proteins, called amyloid fibrils, leading to diverse clinical manifestations. Recent studies on the amyloidogenesis of SARS-CoV-2 protein segments have raised concerns on their potential link to post-infection neurodegeneration, however, the mechanisms remain unclear. Herein, we investigated the structure, stability, and amyloidogenic propensity of a nine-residue segment (SK9) of the SARS-CoV-2 envelope protein and their impact on neuronal protein α-synuclein (αSyn) aggregation. Specifically, the amino acid sequence of the SK9 wildtype has been modified from a basic and positively charged peptide (SFYVYSRVK), to a nearly neutral and more hydrophobic peptide (SAAVASAVK, labelled as SK9 var1), and to an acidic and negatively charged peptide (SDAVANAVK, labelled as SK9 var2). Our findings reveal that the SK9 wildtype exhibited a pronounced amyloidogenic propensity due to its disordered and unstable nature, while the SK9 variants possessed more ordered and stable structures preventing the amyloid formation. Significantly, the SK9 wildtype demonstrated distinct effect on αSyn aggregation kinetics and aggregate morphology to facilitate the formation of αSyn aggregates with enhanced resistance against enzymatic degradation. This study highlights the potential of modifying short peptide sequences to fine-tune their properties, providing insights into understanding and regulating viral-induced amyloid aggregations.
Collapse
Affiliation(s)
- Vince St Dollente Mesias
- Department of Chemistry, The, Hong Kong University of Science and Technology, Clearwater Bay Road, Kowloon, Hong Kong SAR, China
| | - Jianing Zhang
- Department of Chemistry, The, Hong Kong University of Science and Technology, Clearwater Bay Road, Kowloon, Hong Kong SAR, China
| | - Hongni Zhu
- Department of Chemistry, The, Hong Kong University of Science and Technology, Clearwater Bay Road, Kowloon, Hong Kong SAR, China
- Research Center for Biomedical Optics and Molecular Imaging, Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xin Dai
- Department of Chemistry, The, Hong Kong University of Science and Technology, Clearwater Bay Road, Kowloon, Hong Kong SAR, China
| | - Jixi Li
- School of Life Sciences, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200438, China
| | - Jinqing Huang
- Department of Chemistry, The, Hong Kong University of Science and Technology, Clearwater Bay Road, Kowloon, Hong Kong SAR, China
| |
Collapse
|
12
|
Puławski W, Koliński A, Koliński M. Multiscale modeling of protofilament structures: A case study on insulin amyloid aggregates. Int J Biol Macromol 2024; 285:138382. [PMID: 39638203 DOI: 10.1016/j.ijbiomac.2024.138382] [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: 11/04/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Under certain conditions, proteins may undergo misfolding and form long insoluble aggregates called amyloid fibrils. The presence of these aggregates is often associated with various diseases. The molecular mechanisms governing the aggregation process are yet to be fully understood. The self-assembly of amyloid protofilaments occurs over extended time frames, making the simulation of such events problematic. In this work, we describe a pipeline for multiscale modeling protofilament structures. In the first stage, the self-assembly of short fibrillar oligomers occurs during coarse-grained docking simulations of multiple copies of aggregating peptides. Subsequently, symmetry criteria are used to select the highest-ranked oligomer structures. Selected models are then reconstructed to an all-atom representation and used for the assembly of longer protofilaments. Models are optimized using molecular dynamics. Final structures are selected using various scoring protocols. We evaluated this modeling procedure through the test prediction of insulin amyloid protofilaments whose experimental structures have been published recently. The resulting insulin protofilament models closely resemble the experimental structures. This work provides a proof of concept for the proposed modeling procedure aiming to predict amyloid protofilament structures that exhibit in-register and parallel arrangement of β-sheets based solely on the amino acid sequence of aggregating peptides.
Collapse
Affiliation(s)
- Wojciech Puławski
- Bioinformatics Laboratory, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland.
| | - Andrzej Koliński
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Michał Koliński
- Bioinformatics Laboratory, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5, 02-106 Warsaw, Poland.
| |
Collapse
|
13
|
Zhang J, Mesias VSD, Chesney AD, Anand VK, Feng X, Hsing IM, Hansmann UHE, Huang J. Differential effects of SARS-CoV-2 amyloidogenic segments on the aggregation and toxicity of human islet amyloid polypeptide within membrane environments. Int J Biol Macromol 2024; 283:137930. [PMID: 39579816 DOI: 10.1016/j.ijbiomac.2024.137930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
Human islet amyloid polypeptide (hIAPP), an intrinsically disordered protein (IDP), plays a significant role in the pathogenesis of type 2 diabetes through its aggregation. Recent studies have suggested that certain viral protein segments exhibit amyloidogenic potential and may influence its amyloid aggregations associated with pathogenesis. However, the potential link between recurrent SARS-CoV-2 infections and the exacerbation of type 2 diabetes remains poorly understood. In this study, we explore how the amyloidogenic segments of SARS-CoV-2, specifically SK9 and FI10, influence the aggregation of hIAPP and the toxicity of the resulting conformers in a membrane environment. To investigate this, we utilized a range of biophysical techniques, including circular dichroism, nuclear magnetic resonance, atomic force microscopy, dynamic light scattering, fluorescence assays, and cell cytotoxicity assays, complemented by molecular dynamics simulations. Our results indicate that SK9 and FI10 promote hIAPP aggregation in a membrane-mimicking environment, forming distinct aggregate structures. Specifically, SK9 accelerates rapid fibril formation due to inter-chain interactions, while FI10 stabilizes oligomeric aggregates primarily through intra-chain contacts. These results reveal the differential effects of viral protein segments on amyloid formation pathways and aggregate characteristics, providing new insights into the mechanisms of amyloid aggregation for developing better therapeutic strategies against amyloid-associated diseases, particularly diabetes.
Collapse
Affiliation(s)
- Jianing Zhang
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Vince St Dollente Mesias
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Andrew D Chesney
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, OK 73019, United States
| | - Vignesh K Anand
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, OK 73019, United States
| | - Xianzhen Feng
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - I-Ming Hsing
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Ulrich H E Hansmann
- Department of Chemistry & Biochemistry, University of Oklahoma, Norman, OK 73019, United States.
| | - Jinqing Huang
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China.
| |
Collapse
|
14
|
Zargan S, Jalili H, Dabirmanesh B, Mesdaghinia S, Khajeh K. Amyloidogenesis of SARS-CoV-2 delta plus and omicron variants receptor-binding domain (RBD): impact of SUMO fusion tag. Biotechnol Lett 2024; 46:1037-1048. [PMID: 39182215 DOI: 10.1007/s10529-024-03525-9] [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: 04/13/2024] [Revised: 07/16/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
PURPOSE The RBD of SARS-CoV-2 mediates viral entry into host cells by binding to the host receptor ACE2. SARS-CoV-2 infection is linked to various health issues resembling amyloid-related problems, persuading us to investigate the amyloidogenicity of the SARS-CoV-2 spike RBD. METHODS The FoldAmyloid program was used to assess the amyloidogenic propensities in the RBD of Delta Plus and RBD of the Omicron variant, with and without the SUMO tag. After the expression of RBDs, purification, and dialysis steps were performed, subsequently the ThT assay, FTIR, and TEM were employed to check the RBD ability to form fibrils. RESULTS The ThT assay, TEM, and FTIR revealed the ability of RBD to self-assemble into β-sheet-rich aggregates (48.4% β-sheet content). Additionally, the presence of the SUMO tag reduced the formation of RBD amyloid-like fibrils. The amyloidogenic potential of Omicron RBD was higher than Delta Plus, according to both in silico and experimental analyses. CONCLUSIONS The SARS-CoV-2 RBD can assemble itself by forming aggregates containing amyloid-like fibrils and the presence of a SUMO tag can significantly decrease the formation of RBD amyloid-like fibrils. In silico analysis suggested that variation in the ThT fluorescence intensity of amyloid accumulations in the two SARS-CoV-2 strains arises from specific mutations in their RBD regions.
Collapse
Affiliation(s)
- Sadegh Zargan
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Hasan Jalili
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran.
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saba Mesdaghinia
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Khosro Khajeh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
15
|
Christ W, Kapell S, Sobkowiak MJ, Mermelekas G, Evertsson B, Sork H, Saher O, Bazaz S, Gustafsson O, Cardenas EI, Villa V, Ricciarelli R, Sandberg JK, Bergquist J, Sturchio A, Svenningsson P, Malm T, Espay AJ, Pernemalm M, Lindén A, Klingström J, El Andaloussi S, Ezzat K. SARS-CoV-2 and HSV-1 Induce Amyloid Aggregation in Human CSF Resulting in Drastic Soluble Protein Depletion. ACS Chem Neurosci 2024; 15:4095-4104. [PMID: 39510798 DOI: 10.1021/acschemneuro.4c00636] [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: 11/15/2024] Open
Abstract
The corona virus (SARS-CoV-2) pandemic and the resulting long-term neurological complications in patients, known as long COVID, have renewed interest in the correlation between viral infections and neurodegenerative brain disorders. While many viruses can reach the central nervous system (CNS) causing acute or chronic infections (such as herpes simplex virus 1, HSV-1), the lack of a clear mechanistic link between viruses and protein aggregation into amyloids, a characteristic of several neurodegenerative diseases, has rendered such a connection elusive. Recently, we showed that viruses can induce aggregation of purified amyloidogenic proteins via the direct physicochemical mechanism of heterogeneous nucleation (HEN). In the current study, we show that the incubation of HSV-1 and SARS-CoV-2 with human cerebrospinal fluid (CSF) leads to the amyloid aggregation of several proteins known to be involved in neurodegenerative diseases, such as APLP1 (amyloid β precursor like protein 1), ApoE, clusterin, α2-macroglobulin, PGK-1 (phosphoglycerate kinase 1), ceruloplasmin, nucleolin, 14-3-3, transthyretin, and vitronectin. Importantly, UV-inactivation of SARS-CoV-2 does not affect its ability to induce amyloid aggregation, as amyloid formation is dependent on viral surface catalysis via HEN and not its ability to replicate. Additionally, viral amyloid induction led to a dramatic drop in the soluble protein concentration in the CSF. Our results show that viruses can physically induce amyloid aggregation of proteins in human CSF and result in soluble protein depletion, thus providing a potential mechanism that may account for the association between persistent and latent/reactivating brain infections and neurodegenerative diseases.
Collapse
Affiliation(s)
- Wanda Christ
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 14152 Stockholm, Sweden
| | - Sebastian Kapell
- Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1 Chome-12-4 Sakamoto, Nagasaki 852-8102, Japan
| | - Michal J Sobkowiak
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 14152 Stockholm, Sweden
| | - Georgios Mermelekas
- Cancer Proteomics Mass Spectrometry, SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Björn Evertsson
- Department of Clinical Neuroscience and Centrum for Molecular Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Helena Sork
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia
| | - Osama Saher
- Biomolecular and Cellular Medicine (BCM), Department of Laboratory Medicine, Karolinska Institutet, 14152 Stockholm, Sweden
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, 11562 Cairo, Egypt
| | - Safa Bazaz
- Biomolecular and Cellular Medicine (BCM), Department of Laboratory Medicine, Karolinska Institutet, 14152 Stockholm, Sweden
| | - Oskar Gustafsson
- Biomolecular and Cellular Medicine (BCM), Department of Laboratory Medicine, Karolinska Institutet, 14152 Stockholm, Sweden
| | - Eduardo I Cardenas
- Division of Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Viviana Villa
- Department of Experimental Medicine, Section of General Pathology, School of Medical and Pharmaceutical Sciences, University of Genoa, 16132 Genoa, Italy
| | - Roberta Ricciarelli
- Department of Experimental Medicine, Section of General Pathology, School of Medical and Pharmaceutical Sciences, University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Johan K Sandberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 14152 Stockholm, Sweden
| | - Jonas Bergquist
- Department of Chemistry-Biomedical Center, Analytical Chemistry and Neuro Chemistry, Uppsala University, 75105 Uppsala, Sweden
| | - Andrea Sturchio
- Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, 17177 Stockholm, Sweden
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, Ohio 45267-0525, United States
| | - Per Svenningsson
- Department of Clinical Neuroscience, Neuro Svenningsson, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Tarja Malm
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Alberto J Espay
- James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology, University of Cincinnati, Cincinnati, Ohio 45267-0525, United States
| | - Maria Pernemalm
- Cancer Proteomics Mass Spectrometry, SciLifeLab, Department of Oncology and Pathology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Anders Lindén
- Division of Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
- Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Jonas Klingström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 14152 Stockholm, Sweden
| | - Samir El Andaloussi
- Biomolecular and Cellular Medicine (BCM), Department of Laboratory Medicine, Karolinska Institutet, 14152 Stockholm, Sweden
| | - Kariem Ezzat
- Regain Therapeutics, Novum, 14157 Stockholm, Sweden
| |
Collapse
|
16
|
Zhang T, Li Y, Pan L, Sha J, Bailey M, Faure-Kumar E, Williams CK, Wohlschlegel J, Magaki S, Niu C, Lee Y, Su YC, Li X, Vinters HV, Geschwind DH. Brain-wide alterations revealed by spatial transcriptomics and proteomics in COVID-19 infection. NATURE AGING 2024; 4:1598-1618. [PMID: 39543407 PMCID: PMC11867587 DOI: 10.1038/s43587-024-00730-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 09/25/2024] [Indexed: 11/17/2024]
Abstract
Understanding the pathophysiology of neurological symptoms observed after severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infection is essential to optimizing outcomes and therapeutics. To date, small sample sizes and narrow molecular profiling have limited the generalizability of findings. In this study, we profiled multiple cortical and subcortical regions in postmortem brains of patients with coronavirus disease 2019 (COVID-19) and controls with matched pulmonary pathology (total n = 42) using spatial transcriptomics, bulk gene expression and proteomics. We observed a multi-regional antiviral response without direct active SARS-CoV2 infection. We identified dysregulation of mitochondrial and synaptic pathways in deep-layer excitatory neurons and upregulation of neuroinflammation in glia, consistent across both mRNA and protein. Remarkably, these alterations overlapped substantially with changes in age-related neurodegenerative diseases, including Parkinson's disease and Alzheimer's disease. Our work, combining multiple experimental and analytical methods, demonstrates the brain-wide impact of severe acute/subacute COVID-19, involving both cortical and subcortical regions, shedding light on potential therapeutic targets within pathways typically associated with pathological aging and neurodegeneration.
Collapse
Affiliation(s)
- Ting Zhang
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yunfeng Li
- Translational Pathology Core Laboratory, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Liuliu Pan
- Technology Access Program, Bruker Spatial Technology, Seattle, WA, USA
- Duality Biologics, Shanghai, China
| | - Jihui Sha
- Proteome Research Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Michael Bailey
- Proof of Principle Team, Translational Science, Bruker Spatial Technology, Seattle, WA, USA
| | - Emmanuelle Faure-Kumar
- Center for Systems Biomedicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Christopher Kazu Williams
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - James Wohlschlegel
- Proteome Research Center, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Shino Magaki
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chao Niu
- Technology Center for Genomics & Bioinformatics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yoojin Lee
- Technology Center for Genomics & Bioinformatics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yu-Chyuan Su
- Technology Center for Genomics & Bioinformatics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xinmin Li
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Technology Center for Genomics & Bioinformatics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Harry V Vinters
- Section of Neuropathology, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Daniel H Geschwind
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Sanislav O, Tetaj R, Metali, Ratcliffe J, Phillips W, Klein AR, Sethi A, Zhou J, Mezzenga R, Saxer SS, Charnley M, Annesley SJ, Reynolds NP. Cell invasive amyloid assemblies from SARS-CoV-2 peptides can form multiple polymorphs with varying neurotoxicity. NANOSCALE 2024; 16:19814-19827. [PMID: 39363846 DOI: 10.1039/d4nr03030c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
The neurological symptoms of COVID-19, often referred to as neuro-COVID include neurological pain, memory loss, cognitive and sensory disruption. These neurological symptoms can persist for months and are known as Post-Acute Sequalae of COVID-19 (PASC). The molecular origins of neuro-COVID, and how it contributes to PASC are unknown, however a growing body of research highlights that the self-assembly of protein fragments from SARS-CoV-2 into amyloid nanofibrils may play a causative role. Previously, we identified two fragments from the SARS-CoV-2 proteins, Open Reading Frame (ORF) 6 and ORF10, that self-assemble into neurotoxic amyloid assemblies. Here we further our understanding of the self-assembly mechanisms and nano-architectures formed by these fragments and their biological responses. By solubilising the peptides in a fluorinated solvent, we eliminate insoluble aggregates in the starting materials (seeds) that change the polymorphic landscape of the assemblies. The resultant assemblies are dominated by structures with higher free energies (e.g. ribbons and amorphous aggregates) that are less toxic to cultured neurons but do affect their mitochondrial respiration. We also show the first direct evidence of cellular uptake of viral amyloids. This work highlights the importance of understanding the polymorphic behaviour of amyloids and the correlation to neurotoxicity, particularly in the context of neuro-COVID and PASC.
Collapse
Affiliation(s)
- Oana Sanislav
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Rina Tetaj
- Institute for Chemistry and Bioanalytics, School of Life Sciences, FHNW, Muttenz, 4132, Switzerland
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, Victoria 3086, Australia.
| | - Metali
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, Victoria 3086, Australia.
| | - Julian Ratcliffe
- Bio Imaging Platform, La Trobe University, Melbourne, Victoria 3086, Australia
| | - William Phillips
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, Victoria 3086, Australia.
| | - Annaleise R Klein
- Australian Nuclear Science and Technology Organisation (ANSTO), Australian Synchrotron, Clayton, Victoria 3168, Australia
| | - Ashish Sethi
- Australian Nuclear Science and Technology Organisation (ANSTO), Australian Synchrotron, Clayton, Victoria 3168, Australia
| | - Jiangtao Zhou
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, LFO, E23, 8092, Zurich, Switzerland
| | - Raffaele Mezzenga
- Department of Health Sciences and Technology, ETH Zurich, Schmelzbergstrasse 9, LFO, E23, 8092, Zurich, Switzerland
- Department of Materials, ETH Zurich, Zurich, 8093, Switzerland
| | - Sina S Saxer
- Institute for Chemistry and Bioanalytics, School of Life Sciences, FHNW, Muttenz, 4132, Switzerland
| | - Mirren Charnley
- Optical Sciences Centre, Swinburne University of Technology, Hawthorn, Victoria 3122, Australia
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Parkville, Victoria 3000, Australia
| | - Sarah J Annesley
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Nicholas P Reynolds
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, Victoria 3086, Australia.
- The Biomedical and Environmental Sensor Technology (BEST) Research Centre, Biosensors Program, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Victoria 3086, Australia
| |
Collapse
|
18
|
Kell DB, Pretorius E. Proteomic Evidence for Amyloidogenic Cross-Seeding in Fibrinaloid Microclots. Int J Mol Sci 2024; 25:10809. [PMID: 39409138 PMCID: PMC11476703 DOI: 10.3390/ijms251910809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
In classical amyloidoses, amyloid fibres form through the nucleation and accretion of protein monomers, with protofibrils and fibrils exhibiting a cross-β motif of parallel or antiparallel β-sheets oriented perpendicular to the fibre direction. These protofibrils and fibrils can intertwine to form mature amyloid fibres. Similar phenomena can occur in blood from individuals with circulating inflammatory molecules (and also some originating from viruses and bacteria). Such pathological clotting can result in an anomalous amyloid form termed fibrinaloid microclots. Previous proteomic analyses of these microclots have shown the presence of non-fibrin(ogen) proteins, suggesting a more complex mechanism than simple entrapment. We thus provide evidence against such a simple entrapment model, noting that clot pores are too large and centrifugation would have removed weakly bound proteins. Instead, we explore whether co-aggregation into amyloid fibres may involve axial (multiple proteins within the same fibril), lateral (single-protein fibrils contributing to a fibre), or both types of integration. Our analysis of proteomic data from fibrinaloid microclots in different diseases shows no significant quantitative overlap with the normal plasma proteome and no correlation between plasma protein abundance and their presence in fibrinaloid microclots. Notably, abundant plasma proteins like α-2-macroglobulin, fibronectin, and transthyretin are absent from microclots, while less abundant proteins such as adiponectin, periostin, and von Willebrand factor are well represented. Using bioinformatic tools, including AmyloGram and AnuPP, we found that proteins entrapped in fibrinaloid microclots exhibit high amyloidogenic tendencies, suggesting their integration as cross-β elements into amyloid structures. This integration likely contributes to the microclots' resistance to proteolysis. Our findings underscore the role of cross-seeding in fibrinaloid microclot formation and highlight the need for further investigation into their structural properties and implications in thrombotic and amyloid diseases. These insights provide a foundation for developing novel diagnostic and therapeutic strategies targeting amyloidogenic cross-seeding in blood clotting disorders.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- The Novo Nordisk Foundation Centre for Biosustainability, Building 220, Søltofts Plads 200, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St., Liverpool L69 7ZB, UK
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| |
Collapse
|
19
|
Lee JH, Sergi C, Kast RE, Kanwar BA, Bourbeau J, Oh S, Sohn MG, Lee CJ, Coleman MD. Aggravating mechanisms from COVID-19. Virol J 2024; 21:228. [PMID: 39334442 PMCID: PMC11430051 DOI: 10.1186/s12985-024-02506-8] [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: 08/08/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces immune-mediated diseases. The pathophysiology of COVID-19 uses the following three mechanisms: (1) inflammasome activation mechanism; (2) cGAS-STING signaling mechanism; and (3) SAMHD1 tetramerization mechanism, which leads to IFN-I production. Interactions between the host and virus govern induction, resulting in multiorgan impacts. The NLRP3 with cGAS-STING constitutes the primary immune response. The expression of SARS-CoV-2 ORF3a, NSP6, NSP7, and NSP8 blocks innate immune activation and facilitates virus replication by targeting the RIG-I/MDA5, TRIF, and cGAS-STING signaling. SAMHD1 has a target motif for CDK1 to protect virion assembly, threonine 592 to modulate a catalytically active tetramer, and antiviral IFN responses to block retroviral infection. Plastic and allosteric nucleic acid binding of SAMHD1 modulates the antiretroviral activity of SAMHD1. Therefore, inflammasome activation, cGAS-STING signaling, and SAMHD1 tetramerization explain acute kidney injury, hepatic, cardiac, neurological, and gastrointestinal injury of COVID-19. It might be necessary to effectively block the pathological courses of diverse diseases.
Collapse
Affiliation(s)
- Jong Hoon Lee
- Science and Research Center, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Geriatrics, Gyeonggi Medical Center Pocheon Hospital, 1648 Pocheon-ro Sin-eup-dong, Pocheon-si, Gyeonggi-do, 11142, Republic of Korea.
| | - Consolato Sergi
- Division of Anatomical Pathology, Children's Hospital of Eastern Ontario (CHEO), University of Ottawa, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
| | - Richard E Kast
- IIAIGC Study Center, 11 Arlington Ct, Burlington, 05408 VT, USA
| | - Badar A Kanwar
- Haider Associates, 1999 Forest Ridge Dr, Bedford, TX, 76021, USA
| | - Jean Bourbeau
- Respiratory Epidemiology and Clinical Research Unit, McGill University Health Centre, Montréal, QC, Canada
| | - Sangsuk Oh
- Department of Food Engineering, Food Safety Laboratory, Memory Unit, Ewha Womans University, Seoul, 03670, Korea
| | - Mun-Gi Sohn
- Department of Food Science, KyungHee University College of Life Science, Seoul, 17104, Republic of Korea
| | - Chul Joong Lee
- Department of Anesthesiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Michael D Coleman
- College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|
20
|
Greenhalgh T, Sivan M, Perlowski A, Nikolich JŽ. Long COVID: a clinical update. Lancet 2024; 404:707-724. [PMID: 39096925 DOI: 10.1016/s0140-6736(24)01136-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/07/2024] [Accepted: 05/30/2024] [Indexed: 08/05/2024]
Abstract
Post-COVID-19 condition (also known as long COVID) is generally defined as symptoms persisting for 3 months or more after acute COVID-19. Long COVID can affect multiple organ systems and lead to severe and protracted impairment of function as a result of organ damage. The burden of this disease, both on the individual and on health systems and national economies, is high. In this interdisciplinary Review, with a coauthor with lived experience of severe long COVID, we sought to bring together multiple streams of literature on the epidemiology, pathophysiology (including the hypothesised mechanisms of organ damage), lived experience and clinical manifestations, and clinical investigation and management of long COVID. Although current approaches to long COVID care are largely symptomatic and supportive, recent advances in clinical phenotyping, deep molecular profiling, and biomarker identification might herald a more mechanism-informed and personally tailored approach to clinical care. We also cover the organisation of services for long COVID, approaches to preventing long COVID, and suggestions for future research.
Collapse
Affiliation(s)
- Trisha Greenhalgh
- Nuffield Department of Primary Care Health Sciences, Radcliffe Observatory Quarter, Oxford, UK.
| | - Manoj Sivan
- Academic Department of Rehabilitation Medicine, Leeds Institute of Rheumatic and Musculoskeletal Medicine University of Leeds, Leeds General Infirmary, Leeds, UK
| | | | - Janko Ž Nikolich
- Department of Immunobiology and University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The Aegis Consortium for Pandemic-Free Future, University of Arizona Health Sciences, Tucson, AZ, USA
| |
Collapse
|
21
|
Baig AM, Rosko S, Jaeger B, Gerlach J, Rausch H. Unraveling the enigma of long COVID: novel aspects in pathogenesis, diagnosis, and treatment protocols. Inflammopharmacology 2024; 32:2075-2090. [PMID: 38771409 DOI: 10.1007/s10787-024-01483-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/17/2024] [Indexed: 05/22/2024]
Abstract
Long COVID, now unmistakably identified as a syndromic entity encompassing a complex spectrum of symptoms, demands immediate resolution of its elusive pathogenic underpinnings. The intricate interplay of diverse factors presents a complex puzzle, difficult to resolve, and thus poses a substantial challenge. As instances of long COVID manifest by repeated infections of SARS-CoV-2 and genetic predisposition, a detailed understanding in this regard is needed. This endeavor is a comprehensive exploration and analysis of the cascading pathogenetic events driven by viral persistence and replication. Beyond its morbidity, long COVID, more disabling than fatal, exacts one of the most substantial tolls on public health in contemporary times, with the potential to cripple national economies. The paper introduces a unified theory of long COVID, detailing a novel pathophysiological framework that interlinks persistent SARS-CoV-2 infection, autoimmunity, and systemic vascular pathology. We posit a model where viral reservoirs, immune dysregulation, and genetic predispositions converge to perpetuate disease. It challenges prevailing hypotheses with new evidence, suggesting innovative diagnostic and therapeutic approaches. The paper aims to shift the paradigm in long COVID research by providing an integrative perspective that encapsulates the multifaceted nature of the condition. We explain the immunological mechanisms, hypercoagulability states, and viral reservoirs in the skull that feed NeuroCOVID in patients with long COVID. Also, this study hints toward a patient approach and how to prioritize treatment sequences in long COVID patients in hospitals and clinics.
Collapse
Affiliation(s)
| | - Sandy Rosko
- Clinicum St. George, Rosenheimer Str. 6-8, Bad Aibling, Germany
| | - Beate Jaeger
- Clinicum St. George, Rosenheimer Str. 6-8, Bad Aibling, Germany
| | - Joachim Gerlach
- Clinicum St. George, Rosenheimer Str. 6-8, Bad Aibling, Germany
| | - Hans Rausch
- Clinicum St. George, Rosenheimer Str. 6-8, Bad Aibling, Germany
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Koroleva ON, Kuzmina NV, Dubrovin EV, Drutsa VL. Atomic force microscopy of spherical intermediates on the pathway to fibril formation of influenza A virus nuclear export protein. Microsc Res Tech 2024; 87:1131-1145. [PMID: 38270267 DOI: 10.1002/jemt.24499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 01/02/2024] [Accepted: 01/07/2024] [Indexed: 01/26/2024]
Abstract
The nuclear export protein of the influenza A virus (NEP) is involved in many important processes of the virus life cycle. This makes it an attractive target for the treatment of a disease caused by a virus. Previously it has been shown, that recombinant variants of NEP are highly prone to aggregation in solution under various conditions with the formation of amyloid-like aggregates. In the present work, the amyloid nature of NEP aggregates was evidenced by Congo red binding assays. Atomic force microscopy has shown that NEP can form two types of spherical nanoparticles, which provide an alternative pathway for the formation of amyloid-like fibrils. Type I of these "fibrillogenic" spheres, formed under physiological conditions, represents the micelle-like particles with height 10-60 nm, which can generate worm-like flexible fibrils with the diameter 2.5-4.0 nm, length 20-500 nm and the Young's modulus ~73 MPa. Type II spherical aggregates with size of about 400-1000 nm, formed at elevated temperatures, includes fractions of drop-like and vesicle-like particles, generating more rigid amyloid-like fibrils with height of ~8 nm, and length of up to 2 μm. The hypothetical mechanism of fibril formation via nanospherical structures was suggested. RESEARCH HIGHLIGHTS: AFM has revealed two types of the influenza A virus nuclear export protein spherical aggregates. They provide an alternative pathway for the formation of amyloid-like fibrils. The mechanism of fibril formation via spherical structures is suggested.
Collapse
Affiliation(s)
- Olga N Koroleva
- Faculty of Chemistry, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Natalia V Kuzmina
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, Moscow, Russian Federation
| | - Evgeniy V Dubrovin
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russian Federation
- National University of Science and Technology, MISIS, Moscow, Russian Federation
| | - Valeriy L Drutsa
- A.N.Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation
| |
Collapse
|
25
|
Pretorius E, Kell DB. A Perspective on How Fibrinaloid Microclots and Platelet Pathology May be Applied in Clinical Investigations. Semin Thromb Hemost 2024; 50:537-551. [PMID: 37748515 PMCID: PMC11105946 DOI: 10.1055/s-0043-1774796] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Microscopy imaging has enabled us to establish the presence of fibrin(ogen) amyloid (fibrinaloid) microclots in a range of chronic, inflammatory diseases. Microclots may also be induced by a variety of purified substances, often at very low concentrations. These molecules include bacterial inflammagens, serum amyloid A, and the S1 spike protein of severe acute respiratory syndrome coronavirus 2. Here, we explore which of the properties of these microclots might be used to contribute to differential clinical diagnoses and prognoses of the various diseases with which they may be associated. Such properties include distributions in their size and number before and after the addition of exogenous thrombin, their spectral properties, the diameter of the fibers of which they are made, their resistance to proteolysis by various proteases, their cross-seeding ability, and the concentration dependence of their ability to bind small molecules including fluorogenic amyloid stains. Measuring these microclot parameters, together with microscopy imaging itself, along with methodologies like proteomics and imaging flow cytometry, as well as more conventional assays such as those for cytokines, might open up the possibility of a much finer use of these microclot properties in generative methods for a future where personalized medicine will be standard procedures in all clotting pathology disease diagnoses.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Matieland, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Matieland, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
26
|
Li M, Castro Lingl S, Yang J. Reduction of hemagglutination induced by a SARS-CoV-2 spike protein fragment using an amyloid-binding benzothiazole amphiphile. Sci Rep 2024; 14:12317. [PMID: 38811619 PMCID: PMC11137076 DOI: 10.1038/s41598-024-59585-4] [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: 12/16/2023] [Accepted: 04/12/2024] [Indexed: 05/31/2024] Open
Abstract
COVID-19 infection is associated with a variety of vascular occlusive morbidities. However, a comprehensive understanding of how this virus can induce vascular complications remains lacking. Here, we show that a peptide fragment of SARS-CoV-2 spike protein, S192 (sequence 192-211), is capable of forming amyloid-like aggregates that can induce agglutination of red blood cells, which was not observed with low- and non-aggregated S192 peptide. We subsequently screened eight amyloid-binding molecules and identified BAM1-EG6, a benzothiazole amphiphile, as a promising candidate capable of binding to aggregated S192 and partially inhibiting its agglutination activity. These results provide new insight into a potential molecular mechanism for the capability of spike protein metabolites to contribute to COVID-19-related blood complications and suggest a new therapeutic approach for combating microvascular morbidities in COVID-19 patients.
Collapse
Affiliation(s)
- Meihan Li
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093-0358, USA
| | - Sascha Castro Lingl
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093-0358, USA
| | - Jerry Yang
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, 92093-0358, USA.
| |
Collapse
|
27
|
Tan S, Li W, Yang C, Zhan Q, Lu K, Liu J, Jin YM, Bai JS, Wang L, Li J, Li Z, Yu F, Li YY, Duan YX, Lu L, Zhang T, Wei J, Li L, Zheng YT, Jiang S, Liu S. gp120-derived amyloidogenic peptides form amyloid fibrils that increase HIV-1 infectivity. Cell Mol Immunol 2024; 21:479-494. [PMID: 38443447 PMCID: PMC11061181 DOI: 10.1038/s41423-024-01144-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: 07/13/2023] [Accepted: 02/02/2024] [Indexed: 03/07/2024] Open
Abstract
Apart from mediating viral entry, the function of the free HIV-1 envelope protein (gp120) has yet to be elucidated. Our group previously showed that EP2 derived from one β-strand in gp120 can form amyloid fibrils that increase HIV-1 infectivity. Importantly, gp120 contains ~30 β-strands. We examined whether gp120 might serve as a precursor protein for the proteolytic release of amyloidogenic fragments that form amyloid fibrils, thereby promoting viral infection. Peptide array scanning, enzyme degradation assays, and viral infection experiments in vitro confirmed that many β-stranded peptides derived from gp120 can indeed form amyloid fibrils that increase HIV-1 infectivity. These gp120-derived amyloidogenic peptides, or GAPs, which were confirmed to form amyloid fibrils, were termed gp120-derived enhancers of viral infection (GEVIs). GEVIs specifically capture HIV-1 virions and promote their attachment to target cells, thereby increasing HIV-1 infectivity. Different GAPs can cross-interact to form heterogeneous fibrils that retain the ability to increase HIV-1 infectivity. GEVIs even suppressed the antiviral activity of a panel of antiretroviral agents. Notably, endogenous GAPs and GEVIs were found in the lymphatic fluid, lymph nodes, and cerebrospinal fluid (CSF) of AIDS patients in vivo. Overall, gp120-derived amyloid fibrils might play a crucial role in the process of HIV-1 infectivity and thus represent novel targets for anti-HIV therapeutics.
Collapse
Affiliation(s)
- Suiyi Tan
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Wenjuan Li
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Chan Yang
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qingping Zhan
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Kunyu Lu
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jun Liu
- Department of Infectious Disease, The Third People's Hospital of Kunming, Kunming, 650041, China
| | - Yong-Mei Jin
- Department of Infectious Disease, The Third People's Hospital of Kunming, Kunming, 650041, China
| | - Jin-Song Bai
- Department of Infectious Disease, The Third People's Hospital of Kunming, Kunming, 650041, China
| | - Lin Wang
- Department of Pathology, The Third People's Hospital of Kunming, Kunming, 650041, China
| | - Jinqing Li
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zhaofeng Li
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Fei Yu
- Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, College of Life Sciences, Hebei Agricultural University, Baoding, 071001, China
| | - Yu-Ye Li
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Yue-Xun Duan
- Yunnan Provincial Infectious Disease Hospital, Kunming, 650301, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Jiaqi Wei
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Lin Li
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yong-Tang Zheng
- State Key Laboratory of Genetic Evolution & Animal Models, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Center for Biosafety Mega-Science, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Shuwen Liu
- Guangdong-Hong Kong-Macao Joint Laboratory for New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
28
|
Kumar A, Tripathi P, Kumar P, Shekhar R, Pathak R. From Detection to Protection: Antibodies and Their Crucial Role in Diagnosing and Combatting SARS-CoV-2. Vaccines (Basel) 2024; 12:459. [PMID: 38793710 PMCID: PMC11125746 DOI: 10.3390/vaccines12050459] [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: 03/13/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Understanding the antibody response to SARS-CoV-2, the virus responsible for COVID-19, is crucial to comprehending disease progression and the significance of vaccine and therapeutic development. The emergence of highly contagious variants poses a significant challenge to humoral immunity, underscoring the necessity of grasping the intricacies of specific antibodies. This review emphasizes the pivotal role of antibodies in shaping immune responses and their implications for diagnosing, preventing, and treating SARS-CoV-2 infection. It delves into the kinetics and characteristics of the antibody response to SARS-CoV-2 and explores current antibody-based diagnostics, discussing their strengths, clinical utility, and limitations. Furthermore, we underscore the therapeutic potential of SARS-CoV-2-specific antibodies, discussing various antibody-based therapies such as monoclonal antibodies, polyclonal antibodies, anti-cytokines, convalescent plasma, and hyperimmunoglobulin-based therapies. Moreover, we offer insights into antibody responses to SARS-CoV-2 vaccines, emphasizing the significance of neutralizing antibodies in order to confer immunity to SARS-CoV-2, along with emerging variants of concern (VOCs) and circulating Omicron subvariants. We also highlight challenges in the field, such as the risks of antibody-dependent enhancement (ADE) for SARS-CoV-2 antibodies, and shed light on the challenges associated with the original antigenic sin (OAS) effect and long COVID. Overall, this review intends to provide valuable insights, which are crucial to advancing sensitive diagnostic tools, identifying efficient antibody-based therapeutics, and developing effective vaccines to combat the evolving threat of SARS-CoV-2 variants on a global scale.
Collapse
Affiliation(s)
- Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida 201309, India
| | - Prajna Tripathi
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Prashant Kumar
- R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Ritu Shekhar
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
29
|
Wang J, Dai L, Deng M, Xiao T, Zhang Z, Zhang Z. SARS-CoV-2 Spike Protein S1 Domain Accelerates α-Synuclein Phosphorylation and Aggregation in Cellular Models of Synucleinopathy. Mol Neurobiol 2024; 61:2446-2458. [PMID: 37897633 DOI: 10.1007/s12035-023-03726-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/19/2023] [Indexed: 10/30/2023]
Abstract
The 2019 novel coronavirus disease (COVID-19) is an infectious disease that began to spread globally since 2019. Some COVID-19 patients have neurological complications, such as olfactory disorders and movement disorders, which coincide with the symptoms of Parkinson's disease (PD). Increasing imaging and autopsy evidence supports that the density of dopaminergic neurons in the nigrostriatal pathway is damaged in some COVID-19 patients. However, the underlying mechanism that causes PD-like symptoms remains unclear. PD is an age-related neurodegenerative disease with Lewy bodies (LBs) as its histopathologic feature. The main component of LBs is abnormally aggregated α-synuclein (α-syn). The prion-like propagation of α-syn aggregates plays a key role in the onset and progression of PD. The spike protein (S protein) of SARS-CoV-2 is a heparin-binding protein that mediates the entry of the virus into host cells. Here we found that the S1 domain interacts with α-syn and promotes α-syn aggregation. The S1 domain induces mitochondrial dysfunction, oxidative stress, and cytotoxicity. The S1-seeded α-syn fibrils show enhanced seeding activity and induce synaptic damage and cytotoxicity. Thus, the S1 domain of SARS-CoV-2 promotes the aggregation of α-syn in the cellular model of synucleinopathy and may contribute to the pathogenesis of PD.
Collapse
Affiliation(s)
- Jiannan Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Min Deng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Tingting Xiao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430000, China.
| |
Collapse
|
30
|
Gibo M, Kojima S, Fujisawa A, Kikuchi T, Fukushima M. Increased Age-Adjusted Cancer Mortality After the Third mRNA-Lipid Nanoparticle Vaccine Dose During the COVID-19 Pandemic in Japan. Cureus 2024; 16:e57860. [PMID: 38721172 PMCID: PMC11077472 DOI: 10.7759/cureus.57860] [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] [Accepted: 04/06/2024] [Indexed: 06/14/2024] Open
Abstract
During the COVID-19 pandemic, excess deaths including cancer have become a concern in Japan, which has a rapidly aging population. Thus, this study aimed to evaluate how age-adjusted mortality rates (AMRs) for different types of cancer in Japan changed during the COVID-19 pandemic (2020-2022). Official statistics from Japan were used to compare observed annual and monthly AMRs with predicted rates based on pre-pandemic (2010-2019) figures using logistic regression analysis. No significant excess mortality was observed during the first year of the pandemic (2020). However, some excess cancer mortalities were observed in 2021 after mass vaccination with the first and second vaccine doses, and significant excess mortalities were observed for all cancers and some specific types of cancer (including ovarian cancer, leukemia, prostate cancer, lip/oral/pharyngeal cancer, pancreatic cancer, and breast cancer) after mass vaccination with the third dose in 2022. AMRs for the four cancers with the most deaths (lung, colorectal, stomach, and liver) showed a decreasing trend until the first year of the pandemic in 2020, but the rate of decrease slowed in 2021 and 2022. This study discusses possible explanations for these increases in age-adjusted cancer mortality rates.
Collapse
Affiliation(s)
- Miki Gibo
- Primary Health Care, Matsubara Clinic, Kochi, JPN
| | - Seiji Kojima
- Pediatrics, Nagoya Pediatric Cancer Fund, Nagoya, JPN
| | - Akinori Fujisawa
- Cardiovascular Medicine, Honbetsu Cardiovascular Medicine Clinic, Honbetsu, JPN
| | - Takayuki Kikuchi
- Translational Research & Health Data Science, Learning Health Society Institute, Nagoya, JPN
| | - Masanori Fukushima
- Translational Research & Health Data Science, Learning Health Society Institute, Nagoya, JPN
| |
Collapse
|
31
|
Nawaz AD, Haider MZ, Akhtar S. COVID-19 and Alzheimer's disease: Impact of lockdown and other restrictive measures during the COVID-19 pandemic. BIOMOLECULES & BIOMEDICINE 2024; 24:219-229. [PMID: 38078809 PMCID: PMC10950341 DOI: 10.17305/bb.2023.9680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/12/2023] [Accepted: 11/28/2023] [Indexed: 03/14/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection initially results in respiratory distress symptoms but can also lead to central nervous system (CNS) and neurological manifestations, significantly impacting coronavirus disease 2019 (COVID-19) patients with neurodegenerative diseases. Additionally, strict lockdown measures introduced to curtail the spread of COVID-19 have raised concerns over the wellbeing of patients with dementia and/or Alzheimer's disease. The aim of this review was to discuss the overlapping molecular pathologies and the potential bidirectional relationship between COVID-19 and Alzheimer's dementia, as well as the impact of lockdown/restriction measures on the neuropsychiatric symptoms (NPS) of patients with Alzheimer's dementia. Furthermore, we aimed to assess the impact of lockdown measures on the NPS of caregivers, exploring its potential effects on the quality and extent of care they provide to dementia patients.We utilized the PubMed and Google Scholar databases to search for articles on COVID-19, dementia, Alzheimer's disease, lockdown, and caregivers. Our review highlights that patients with Alzheimer's disease face an increased risk of COVID-19 infection and complications. Additionally, these patients are likely to experience greater cognitive decline. It appears that these issues are primarily caused by the SARS-CoV-2 infection and appear to be further exacerbated by restrictive/lockdown measures. Moreover, lockdown measures introduced during the pandemic have negatively impacted both the NPSs of caregivers and their perception of the wellbeing of their Alzheimer's patients. Thus, additional safeguard measures, along with pharmacological and non-pharmacological approaches, are needed to protect the wellbeing of dementia patients and their caregivers in light of this and possible future pandemics.
Collapse
Affiliation(s)
| | | | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
32
|
Wang J, Dai L, Chen S, Zhang Z, Fang X, Zhang Z. Protein-protein interactions regulating α-synuclein pathology. Trends Neurosci 2024; 47:209-226. [PMID: 38355325 DOI: 10.1016/j.tins.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/15/2023] [Accepted: 01/21/2024] [Indexed: 02/16/2024]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease characterized by the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and the formation of Lewy bodies (LBs). The main proteinaceous component of LBs is aggregated α-synuclein (α-syn). However, the mechanisms underlying α-syn aggregation are not yet fully understood. Converging lines of evidence indicate that, under certain pathological conditions, various proteins can interact with α-syn and regulate its aggregation. Understanding these protein-protein interactions is crucial for unraveling the molecular mechanisms contributing to PD pathogenesis. In this review we provide an overview of the current knowledge on protein-protein interactions that regulate α-syn aggregation. Additionally, we briefly summarize the methods used to investigate the influence of protein-protein interactions on α-syn aggregation and propagation.
Collapse
Affiliation(s)
- Jiannan Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Sichun Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xin Fang
- Department of Neurology, the First Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430000, China.
| |
Collapse
|
33
|
Zhang Y, Bharathi V, Dokoshi T, de Anda J, Ursery LT, Kulkarni NN, Nakamura Y, Chen J, Luo EWC, Wang L, Xu H, Coady A, Zurich R, Lee MW, Matsui T, Lee H, Chan LC, Schepmoes AA, Lipton MS, Zhao R, Adkins JN, Clair GC, Thurlow LR, Schisler JC, Wolfgang MC, Hagan RS, Yeaman MR, Weiss TM, Chen X, Li MMH, Nizet V, Antoniak S, Mackman N, Gallo RL, Wong GCL. Viral afterlife: SARS-CoV-2 as a reservoir of immunomimetic peptides that reassemble into proinflammatory supramolecular complexes. Proc Natl Acad Sci U S A 2024; 121:e2300644120. [PMID: 38306481 PMCID: PMC10861912 DOI: 10.1073/pnas.2300644120] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 10/28/2023] [Indexed: 02/04/2024] Open
Abstract
It is unclear how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leads to the strong but ineffective inflammatory response that characterizes severe Coronavirus disease 2019 (COVID-19), with amplified immune activation in diverse cell types, including cells without angiotensin-converting enzyme 2 receptors necessary for infection. Proteolytic degradation of SARS-CoV-2 virions is a milestone in host viral clearance, but the impact of remnant viral peptide fragments from high viral loads is not known. Here, we examine the inflammatory capacity of fragmented viral components from the perspective of supramolecular self-organization in the infected host environment. Interestingly, a machine learning analysis to SARS-CoV-2 proteome reveals sequence motifs that mimic host antimicrobial peptides (xenoAMPs), especially highly cationic human cathelicidin LL-37 capable of augmenting inflammation. Such xenoAMPs are strongly enriched in SARS-CoV-2 relative to low-pathogenicity coronaviruses. Moreover, xenoAMPs from SARS-CoV-2 but not low-pathogenicity homologs assemble double-stranded RNA (dsRNA) into nanocrystalline complexes with lattice constants commensurate with the steric size of Toll-like receptor (TLR)-3 and therefore capable of multivalent binding. Such complexes amplify cytokine secretion in diverse uninfected cell types in culture (epithelial cells, endothelial cells, keratinocytes, monocytes, and macrophages), similar to cathelicidin's role in rheumatoid arthritis and lupus. The induced transcriptome matches well with the global gene expression pattern in COVID-19, despite using <0.3% of the viral proteome. Delivery of these complexes to uninfected mice boosts plasma interleukin-6 and CXCL1 levels as observed in COVID-19 patients.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA9009
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
- Biomedical Engineering, School of Engineering, Westlake University, Hangzhou, Zhejiang310012, China
| | - Vanthana Bharathi
- University of North Carolina Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Tatsuya Dokoshi
- Department of Dermatology, University of California San Diego, La Jolla, CA92093
| | - Jaime de Anda
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA9009
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
| | - Lauryn Tumey Ursery
- University of North Carolina Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Nikhil N. Kulkarni
- Department of Dermatology, University of California San Diego, La Jolla, CA92093
| | - Yoshiyuki Nakamura
- Department of Dermatology, University of California San Diego, La Jolla, CA92093
| | - Jonathan Chen
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA9009
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
| | - Elizabeth W. C. Luo
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA9009
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
| | - Lamei Wang
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Hua Xu
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Alison Coady
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA92093
| | - Raymond Zurich
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA92093
| | - Michelle W. Lee
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA9009
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
| | - Tsutomu Matsui
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA94025
| | - HongKyu Lee
- Division of Molecular Medicine, Harbor-University of California Los Angeles Medical Center, Los Angeles County, Torrance, CA90502
| | - Liana C. Chan
- Division of Molecular Medicine, Harbor-University of California Los Angeles Medical Center, Los Angeles County, Torrance, CA90502
- Division of Infectious Diseases, Harbor-University of California Los Angeles Medical Center, Los Angeles County, Torrance, CA90502
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Institute for Infection & Immunity, Lundquist Institute for Biomedical Innovation, Harbor-University of California Los Angeles Medical Center, Torrance, CA90502
| | - Athena A. Schepmoes
- Environmental Molecular Science Division, Pacific Northwest National Laboratory, Richland, WA99354
| | - Mary S. Lipton
- Environmental Molecular Science Division, Pacific Northwest National Laboratory, Richland, WA99354
| | - Rui Zhao
- Environmental Molecular Science Division, Pacific Northwest National Laboratory, Richland, WA99354
| | - Joshua N. Adkins
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA99354
| | - Geremy C. Clair
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA99354
| | - Lance R. Thurlow
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Jonathan C. Schisler
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Computational Medicine Program, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Matthew C. Wolfgang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Robert S. Hagan
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Michael R. Yeaman
- Division of Molecular Medicine, Harbor-University of California Los Angeles Medical Center, Los Angeles County, Torrance, CA90502
- Division of Infectious Diseases, Harbor-University of California Los Angeles Medical Center, Los Angeles County, Torrance, CA90502
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA90095
- Institute for Infection & Immunity, Lundquist Institute for Biomedical Innovation, Harbor-University of California Los Angeles Medical Center, Torrance, CA90502
| | - Thomas M. Weiss
- Stanford Synchrotron Radiation Lightsource, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA94025
| | - Xinhua Chen
- Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Melody M. H. Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
| | - Victor Nizet
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA92093
| | - Silvio Antoniak
- Department of Pathology and Laboratory Medicine, University of North Carolina Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Nigel Mackman
- University of North Carolina Blood Research Center, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Richard L. Gallo
- Department of Dermatology, University of California San Diego, La Jolla, CA92093
| | - Gerard C. L. Wong
- Department of Bioengineering, University of California, Los Angeles, CA90095
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA9009
- California NanoSystems Institute, University of California, Los Angeles, CA90095
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA90095
| |
Collapse
|
34
|
Sánchez-Moguel I, Coffeen CF, Bustos-Jaimes I. On-column refolding and off-column assembly of parvovirus B19 virus-like particles from bacteria-expressed protein. Appl Microbiol Biotechnol 2024; 108:160. [PMID: 38252281 PMCID: PMC10803429 DOI: 10.1007/s00253-024-13004-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] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/26/2023] [Accepted: 01/04/2024] [Indexed: 01/23/2024]
Abstract
Virus-like particles (VLPs) are nanometric structures composed of structural components of virions, keeping most of the cellular recognition and internalization properties, but are non-infective as they are deprived of their genetic material. VLPs have been a versatile platform for developing vaccines by carrying their own or heterologous antigenic epitopes. Moreover, VLPs can also be used as nanovessels for encapsulating molecules with therapeutic applications, like enzymes, nucleic acids, and drugs. Parvovirus B19 (B19V) VLPs can be self-assembled in vitro from the denatured major viral particle protein VP2 by equilibrium dialysis. Despite its fair productivity, this process is currently a time-consuming task. Affinity chromatography is used as an efficient step for concentration and purification, but it is only sometimes seen as a method that facilitates the oligomerization of proteins. In this research, we report a novel approach for the in vitro assembly of B19V VLPs through the immobilization of the denatured VP2 into an immobilized metal affinity chromatography (IMAC) column, followed by the on-column folding and the final VLP assembly upon protein elution. This method is suitable for the fast production of B19V VLPs. KEY POINTS: • Biotechnological applications for inclusion bodies • Efficient single-step purification and immobilization strategies • Rapid VLP assembly strategy.
Collapse
Affiliation(s)
- Ignacio Sánchez-Moguel
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Carlos Francisco Coffeen
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Ismael Bustos-Jaimes
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
35
|
Zilio G, Masato A, Sandre M, Caregnato A, Moret F, Maciola AK, Antonini A, Brucale M, Cendron L, Plotegher N, Bubacco L. SARS-CoV-2-Mimicking Pseudoviral Particles Accelerate α-Synuclein Aggregation In Vitro. ACS Chem Neurosci 2024; 15:215-221. [PMID: 38131609 DOI: 10.1021/acschemneuro.3c00468] [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: 12/23/2023] Open
Abstract
Since the SARS-CoV-2 virus started spreading worldwide, evidence pointed toward an impact of the infection on the nervous system. COVID-19 patients present neurological manifestations and have an increased risk of developing brain-related symptoms in the long term. In fact, evidence in support of the neuroinvasive potential of SARS-CoV-2 has emerged. Considering that viral parkisonism was observed as a consequence of encephalopathies caused by viral infections, it has been already suggested that COVID-19 could affect the dopaminergic neurons and contribute to neurodegeneration in Parkinson's disease (PD), by promoting the formation of amyloid fibrils constituted by the PD-related protein α-synuclein. Here, we observe not only that SARS-CoV-2 viral spike protein and nucleocapsid protein can alone promote α-synuclein aggregation but also that the spike protein organization in a corona shape on the viral envelope may be crucial in triggering fast amyloid fibrils formation, thus possibly contributing to PD pathogenesis.
Collapse
Affiliation(s)
- Gianluca Zilio
- Department of Biology, University of Padova, Padova 35131, Italy
| | - Anna Masato
- Department of Biology, University of Padova, Padova 35131, Italy
| | - Michele Sandre
- Department of Biology, University of Padova, Padova 35131, Italy
- Department of Neuroscience, University of Padova, Padova 35121, Italy
| | - Alberto Caregnato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35131, Italy
| | - Francesca Moret
- Department of Biology, University of Padova, Padova 35131, Italy
| | | | - Angelo Antonini
- Department of Neuroscience, University of Padova, Padova 35121, Italy
| | - Marco Brucale
- Consiglio Nazionale delle Ricerche, Istituto per lo Studio dei Materiali Nanostrutturati, Bologna 40129, Italy
| | - Laura Cendron
- Department of Biology, University of Padova, Padova 35131, Italy
| | | | - Luigi Bubacco
- Department of Biology, University of Padova, Padova 35131, Italy
| |
Collapse
|
36
|
Mead MN, Seneff S, Wolfinger R, Rose J, Denhaerynck K, Kirsch S, McCullough PA. COVID-19 mRNA Vaccines: Lessons Learned from the Registrational Trials and Global Vaccination Campaign. Cureus 2024; 16:e52876. [PMID: 38274635 PMCID: PMC10810638 DOI: 10.7759/cureus.52876] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2024] [Indexed: 01/27/2024] Open
Abstract
Our understanding of COVID-19 vaccinations and their impact on health and mortality has evolved substantially since the first vaccine rollouts. Published reports from the original randomized phase 3 trials concluded that the COVID-19 mRNA vaccines could greatly reduce COVID-19 symptoms. In the interim, problems with the methods, execution, and reporting of these pivotal trials have emerged. Re-analysis of the Pfizer trial data identified statistically significant increases in serious adverse events (SAEs) in the vaccine group. Numerous SAEs were identified following the Emergency Use Authorization (EUA), including death, cancer, cardiac events, and various autoimmune, hematological, reproductive, and neurological disorders. Furthermore, these products never underwent adequate safety and toxicological testing in accordance with previously established scientific standards. Among the other major topics addressed in this narrative review are the published analyses of serious harms to humans, quality control issues and process-related impurities, mechanisms underlying adverse events (AEs), the immunologic basis for vaccine inefficacy, and concerning mortality trends based on the registrational trial data. The risk-benefit imbalance substantiated by the evidence to date contraindicates further booster injections and suggests that, at a minimum, the mRNA injections should be removed from the childhood immunization program until proper safety and toxicological studies are conducted. Federal agency approval of the COVID-19 mRNA vaccines on a blanket-coverage population-wide basis had no support from an honest assessment of all relevant registrational data and commensurate consideration of risks versus benefits. Given the extensive, well-documented SAEs and unacceptably high harm-to-reward ratio, we urge governments to endorse a global moratorium on the modified mRNA products until all relevant questions pertaining to causality, residual DNA, and aberrant protein production are answered.
Collapse
Affiliation(s)
- M Nathaniel Mead
- Biology and Nutritional Epidemiology, Independent Research, Copper Hill, USA
| | - Stephanie Seneff
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, USA
| | - Russ Wolfinger
- Biostatistics and Epidemiology, Independent Research, Research Triangle Park, USA
| | - Jessica Rose
- Immunology and Public Health Research, Independent Research, Ottawa, CAN
| | - Kris Denhaerynck
- Epidemiology and Biostatistics, Independent Research, Basel, CHE
| | - Steve Kirsch
- Data Science, Independent Research, Los Angeles, USA
| | - Peter A McCullough
- Cardiology, Epidemiology, and Public Health, McCullough Foundation, Dallas, USA
- Cardiology, Epidemiology, and Public Health, Truth for Health Foundation, Tucson, USA
| |
Collapse
|
37
|
Weissert R. Nervous system-related tropism of SARS-CoV-2 and autoimmunity in COVID-19 infection. Eur J Immunol 2024; 54:e2250230. [PMID: 37733584 DOI: 10.1002/eji.202250230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 08/05/2023] [Accepted: 09/20/2023] [Indexed: 09/23/2023]
Abstract
The effects of SARS-CoV-2 in COVID-19 on the nervous system are incompletely understood. SARS-CoV-2 can infect endothelial cells, neurons, astrocytes, and oligodendrocytes with consequences for the host. There are indications that infection of these CNS-resident cells may result in long-term effects, including emergence of neurodegenerative diseases. Indirect effects of infection with SARS-CoV-2 relate to the induction of autoimmune disease involving molecular mimicry or/and bystander activation of T- and B cells and emergence of autoantibodies against various self-antigens. Data obtained in preclinical models of coronavirus-induced disease gives important clues for the understanding of nervous system-related assault of SARS-CoV-2. The pathophysiology of long-COVID syndrome and post-COVID syndrome in which autoimmunity and immune dysregulation might be the driving forces are still incompletely understood. A better understanding of nervous-system-related immunity in COVID-19 might support the development of therapeutic approaches. In this review, the current understanding of SARS-CoV-2 tropism for the nervous system, the associated immune responses, and diseases are summarized. The data indicates that there is viral tropism of SARS-CoV-2 in the nervous system resulting in various disease conditions. Prevention of SARS-CoV-2 infection by means of vaccination is currently the best strategy for the prevention of subsequent tissue damage involving the nervous system.
Collapse
Affiliation(s)
- Robert Weissert
- Department of Neurology, University of Regensburg Hospital, Regensburg, Germany
| |
Collapse
|
38
|
Mercado G, Kaeufer C, Richter F, Peelaerts W. Infections in the Etiology of Parkinson's Disease and Synucleinopathies: A Renewed Perspective, Mechanistic Insights, and Therapeutic Implications. JOURNAL OF PARKINSON'S DISEASE 2024; 14:1301-1329. [PMID: 39331109 PMCID: PMC11492057 DOI: 10.3233/jpd-240195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/06/2024] [Indexed: 09/28/2024]
Abstract
Increasing evidence suggests a potential role for infectious pathogens in the etiology of synucleinopathies, a group of age-related neurodegenerative disorders including Parkinson's disease (PD), multiple system atrophy and dementia with Lewy bodies. In this review, we discuss the link between infections and synucleinopathies from a historical perspective, present emerging evidence that supports this link, and address current research challenges with a focus on neuroinflammation. Infectious pathogens can elicit a neuroinflammatory response and modulate genetic risk in PD and related synucleinopathies. The mechanisms of how infections might be linked with synucleinopathies as well as the overlap between the immune cellular pathways affected by virulent pathogens and disease-related genetic risk factors are discussed. Here, an important role for α-synuclein in the immune response against infections is emerging. Critical methodological and knowledge gaps are addressed, and we provide new future perspectives on how to address these gaps. Understanding how infections and neuroinflammation influence synucleinopathies will be essential for the development of early diagnostic tools and novel therapies.
Collapse
Affiliation(s)
- Gabriela Mercado
- Division of Neurobiology, Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher Kaeufer
- Center for Systems Neuroscience, Hannover, Germany
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Franziska Richter
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Wouter Peelaerts
- Laboratory for Virology and Gene Therapy, Department of Pharmacy and Pharmaceutical Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
39
|
Rashid MH, Singha S, Arshad F, Sen P. Exploring the Potential Long-term Impact of SARS-CoV-2 on Protein Misfolding and Amyloid-related Conditions. Protein Pept Lett 2024; 31:602-610. [PMID: 39253910 DOI: 10.2174/0109298665333817240821111641] [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: 05/28/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 09/11/2024]
Abstract
The long-term impact of the COVID-19 pandemic concerns risk to human health, particularly its potential association with protein misfolding and amyloidosis. This review article explores the causality relationship between SARS-CoV-2 infection, and protein misfolding, leading to amyloid-related conditions. It delves into the mechanisms by which viral proteins may accelerate amyloid formation, exacerbating post-infection complications, including neurological sequelae. Drawing from interdisciplinary research and clinical observations, the potential links between COVID-19, vaccination, and amyloidosis, emphasize the importance of understanding the longterm effect of post-COVID symptoms. This review examines the potential role of COVID-19-related proteins in the formation of amyloid in other related proteins of amyloidosis.
Collapse
Affiliation(s)
- Md Harun Rashid
- Vellore Institute of Technology, Centre for Bio Separation Technology (CBST), Technology Tower, VIT University, Vellore, Tamil Nadu 632014, India
| | - Srinjana Singha
- Vellore Institute of Technology, Centre for Bio Separation Technology (CBST), Technology Tower, VIT University, Vellore, Tamil Nadu 632014, India
| | - Faheem Arshad
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| | - Priyankar Sen
- Vellore Institute of Technology, Centre for Bio Separation Technology (CBST), Technology Tower, VIT University, Vellore, Tamil Nadu 632014, India
| |
Collapse
|
40
|
Louros N, Schymkowitz J, Rousseau F. Mechanisms and pathology of protein misfolding and aggregation. Nat Rev Mol Cell Biol 2023; 24:912-933. [PMID: 37684425 DOI: 10.1038/s41580-023-00647-2] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2023] [Indexed: 09/10/2023]
Abstract
Despite advances in machine learning-based protein structure prediction, we are still far from fully understanding how proteins fold into their native conformation. The conventional notion that polypeptides fold spontaneously to their biologically active states has gradually been replaced by our understanding that cellular protein folding often requires context-dependent guidance from molecular chaperones in order to avoid misfolding. Misfolded proteins can aggregate into larger structures, such as amyloid fibrils, which perpetuate the misfolding process, creating a self-reinforcing cascade. A surge in amyloid fibril structures has deepened our comprehension of how a single polypeptide sequence can exhibit multiple amyloid conformations, known as polymorphism. The assembly of these polymorphs is not a random process but is influenced by the specific conditions and tissues in which they originate. This observation suggests that, similar to the folding of native proteins, the kinetics of pathological amyloid assembly are modulated by interactions specific to cells and tissues. Here, we review the current understanding of how intrinsic protein conformational propensities are modulated by physiological and pathological interactions in the cell to shape protein misfolding and aggregation pathology.
Collapse
Affiliation(s)
- Nikolaos Louros
- Switch Laboratory, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| | - Frederic Rousseau
- Switch Laboratory, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.
- Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
41
|
Nolan TM, Deliyannis G, Griffith M, Braat S, Allen LF, Audsley J, Chung AW, Ciula M, Gherardin NA, Giles ML, Gordon TP, Grimley SL, Horng L, Jackson DC, Juno JA, Kedzierska K, Kent SJ, Lewin SR, Littlejohn M, McQuilten HA, Mordant FL, Nguyen THO, Soo VP, Price B, Purcell DFJ, Ramanathan P, Redmond SJ, Rockman S, Ruan Z, Sasadeusz J, Simpson JA, Subbarao K, Fabb SA, Payne TJ, Takanashi A, Tan CW, Torresi J, Wang JJ, Wang LF, Al-Wassiti H, Wong CY, Zaloumis S, Pouton CW, Godfrey DI. Interim results from a phase I randomized, placebo-controlled trial of novel SARS-CoV-2 beta variant receptor-binding domain recombinant protein and mRNA vaccines as a 4th dose booster. EBioMedicine 2023; 98:104878. [PMID: 38016322 PMCID: PMC10696466 DOI: 10.1016/j.ebiom.2023.104878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/18/2023] [Accepted: 11/02/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND SARS-CoV-2 booster vaccination should ideally enhance protection against variants and minimise immune imprinting. This Phase I trial evaluated two vaccines targeting SARS-CoV-2 beta-variant receptor-binding domain (RBD): a recombinant dimeric RBD-human IgG1 Fc-fusion protein, and an mRNA encoding a membrane-anchored RBD. METHODS 76 healthy adults aged 18-64 y, previously triple vaccinated with licensed SARS-CoV-2 vaccines, were randomised to receive a 4th dose of either an adjuvanted (MF59®, CSL Seqirus) protein vaccine (5, 15 or 45 μg, N = 32), mRNA vaccine (10, 20, or 50 μg, N = 32), or placebo (saline, N = 12) at least 90 days after a 3rd boost vaccination or SARS-CoV-2 infection. Bleeds occurred on days 1 (prior to vaccination), 8, and 29. CLINICALTRIALS govNCT05272605. FINDINGS No vaccine-related serious or medically-attended adverse events occurred. The protein vaccine reactogenicity was mild, whereas the mRNA vaccine was moderately reactogenic at higher dose levels. Best anti-RBD antibody responses resulted from the higher doses of each vaccine. A similar pattern was seen with live virus neutralisation and surrogate, and pseudovirus neutralisation assays. Breadth of immune response was demonstrated against BA.5 and more recent omicron subvariants (XBB, XBB.1.5 and BQ.1.1). Binding antibody titres for both vaccines were comparable to those of a licensed bivalent mRNA vaccine. Both vaccines enhanced CD4+ and CD8+ T cell activation. INTERPRETATION There were no safety concerns and the reactogenicity profile was mild and similar to licensed SARS-CoV-2 vaccines. Both vaccines showed strong immune boosting against beta, ancestral and omicron strains. FUNDING Australian Government Medical Research Future Fund, and philanthropies Jack Ma Foundation and IFM investors.
Collapse
Affiliation(s)
- Terry M Nolan
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Australia; Murdoch Children's Research Institute, Melbourne, Australia.
| | - Georgia Deliyannis
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Maryanne Griffith
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Australia
| | - Sabine Braat
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Lilith F Allen
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jennifer Audsley
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Amy W Chung
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Marcin Ciula
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Nicholas A Gherardin
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Michelle L Giles
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Australia
| | - Tom P Gordon
- Department of Immunology, Flinders University and SA Pathology, Flinders Medical Centre, Bedford Park, Adelaide, Australia
| | - Samantha L Grimley
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Lana Horng
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Australia; Murdoch Children's Research Institute, Melbourne, Australia
| | - David C Jackson
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jennifer A Juno
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Katherine Kedzierska
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia; Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Stephen J Kent
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Sharon R Lewin
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Australia; Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia; Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | - Mason Littlejohn
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Australia
| | - Hayley A McQuilten
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Francesca L Mordant
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Thi H O Nguyen
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Vanessa Pac Soo
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Briony Price
- Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection & Immunity, Melbourne, Australia; Murdoch Children's Research Institute, Melbourne, Australia
| | - Damian F J Purcell
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Pradhipa Ramanathan
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Samuel J Redmond
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Steven Rockman
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia; CSL Seqirus, Vaccine Innovation Unit, Parkville, Melbourne, Australia
| | - Zheng Ruan
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Joseph Sasadeusz
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia; Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | - Julie A Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Kanta Subbarao
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia; WHO Collaborating Centre for Reference and Research on Influenza at the Peter Doherty Institute for Infection and Immunity, Australia
| | - Stewart A Fabb
- Monash Institute of Pharmaceutical Sciences, Parkville, Australia
| | - Thomas J Payne
- Monash Institute of Pharmaceutical Sciences, Parkville, Australia
| | - Asuka Takanashi
- Monash Institute of Pharmaceutical Sciences, Parkville, Australia
| | - Chee Wah Tan
- Duke NUS Medical School, Programme for Emerging Infectious Diseases, Singapore
| | - Joseph Torresi
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jing Jing Wang
- Department of Immunology, Flinders University and SA Pathology, Flinders Medical Centre, Bedford Park, Adelaide, Australia
| | - Lin-Fa Wang
- Duke NUS Medical School, Programme for Emerging Infectious Diseases, Singapore
| | | | - Chinn Yi Wong
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Sophie Zaloumis
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Parkville, Australia
| | - Dale I Godfrey
- Department of Microbiology & Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| |
Collapse
|
42
|
Mir TH, Zargar PA, Sharma A, Jabeen B, Sharma S, Parvaiz MO, Bashir S, Javeed R. Post COVID-19 AA amyloidosis of the kidneys with rapidly progressive renal failure. Prion 2023; 17:111-115. [PMID: 37055928 PMCID: PMC10114959 DOI: 10.1080/19336896.2023.2201151] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 12/15/2022] [Accepted: 01/22/2023] [Indexed: 04/15/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) pandemic has taken the world by a storm, posing a gruelling challenge to the medical fraternity globally. Besides its very high infectivityinfectivity, significant organ dysfunction occurs in critically ill COVID-19 patients, leading to severe morbidity and mortality. Pulmonary involvement is the leading cause of death in these patients to be followed by the cardiovascular involvement. Kidney involvement due to COVID-19 is becoming more discernible with AKI adversely affecting the outcome. Besides AKI, a few cases of collapsing FSGS in genetically vulnerable patients and thrombotic microangiopathies have been reported as well. We report a case of AA amyloidosis of the kidney with a rapidly progressive renal failure and congestive heart failure with preserved left ventricular functions, which complicated a moderate COVID-19 pneumonia providing some clues to a possible association of this novel virus disease with this complication, which needs to be confirmed in future studies.
Collapse
Affiliation(s)
- Tajamul H. Mir
- Department of Nephrology, Government Medical College, Srinagar, Jammu and Kashmir, India
- Department of Nephrology, Khyber Medical Institute Nowpora, Srinagar, Jammu and Kashmir, India
| | - Parvaiz A Zargar
- Department of Cardiology, Government Medical College, Srinagar, Jammu and Kashmir, India
| | - Alok Sharma
- Department of Renal Pathology, Dr. Lal Path lab/National Reference lab, New Delhi, India
| | - Bushra Jabeen
- Department of Radiology, SMHS Hospital, Srinagar, Jammu and Kashmir, India
| | - Shephali Sharma
- Department of Renal Pathology, Dr. Lal Path lab/National Reference lab, New Delhi, India
| | - M. Omar Parvaiz
- Department of Medicine, Holy Family Red Crescent Medical College, Dhaka, Bangladesh
| | - Sabah Bashir
- Department of Nephrology, Khyber Medical Institute Nowpora, Srinagar, Jammu and Kashmir, India
| | - Reem Javeed
- Department of Nephrology, Government Medical College, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
43
|
Turner S, Laubscher GJ, Khan MA, Kell DB, Pretorius E. Accelerating discovery: A novel flow cytometric method for detecting fibrin(ogen) amyloid microclots using long COVID as a model. Heliyon 2023; 9:e19605. [PMID: 37809592 PMCID: PMC10558872 DOI: 10.1016/j.heliyon.2023.e19605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 10/10/2023] Open
Abstract
Long COVID has become a significant global health and economic burden, yet there are currently no established methods or diagnostic tools to identify which patients might benefit from specific treatments. One of the major pathophysiological factors contributing to Long COVID is the presence of hypercoagulability; this results in insoluble amyloid microclots that are resistant to fibrinolysis. Our previous research using fluorescence microscopy has demonstrated a significant amyloid microclot load in Long COVID patients. However, this approach lacked the elements of statistical robustness, objectivity, and rapid throughput. In the current study, we have used imaging flow cytometry for the first time to show a significantly increased concentration and size of these microclots. We identified notable variations in size and fluorescence between microclots in Long COVID and those of controls even using a 20× objective. By combining cell imaging and the high-event-rate and full-sample analysis nature of a conventional flow cytometer, imaging flow cytometry can eliminate erroneous results and increase accuracy in gating and analysis beyond what pure quantitative measurements from conventional flow cytometry can provide. Although imaging flow cytometry was used in our study, our results suggest that the signals indicating the presence of microclots should be easily detectable using a conventional flow cytometer. Flow cytometry is a more widely available technique than fluorescence microscopy and has been used in pathology laboratories for decades, rendering it a potentially more suitable and accessible method for detecting microclots in individuals suffering from Long COVID or conditions with similar pathology, such as myalgic encephalomyelitis.
Collapse
Affiliation(s)
- Simone Turner
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland, 7602, South Africa
| | | | - M Asad Khan
- Respiratory Medicine, Manchester University Hospitals, Wythenshawe Hospital, Manchester, M23 9LT, United Kingdom
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland, 7602, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, L69 7ZB, UK
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kemitorvet 200, 2800, Kgs Lyngby, Denmark
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland, 7602, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, L69 7ZB, UK
| |
Collapse
|
44
|
Cao S, Song Z, Rong J, Andrikopoulos N, Liang X, Wang Y, Peng G, Ding F, Ke PC. Spike Protein Fragments Promote Alzheimer's Amyloidogenesis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:40317-40329. [PMID: 37585091 PMCID: PMC10480042 DOI: 10.1021/acsami.3c09815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
Abstract
Alzheimer's disease (AD) is a major cause of dementia inducing memory loss, cognitive decline, and mortality among the aging population. While the amyloid aggregation of peptide Aβ has long been implicated in neurodegeneration in AD, primarily through the production of toxic polymorphic aggregates and reactive oxygen species, viral infection has a less explicit role in the etiology of the brain disease. On the other hand, while the COVID-19 pandemic is known to harm human organs and function, its adverse effects on AD pathobiology and other human conditions remain unclear. Here we first identified the amyloidogenic potential of 1058HGVVFLHVTYV1068, a short fragment of the spike protein of SARS-CoV-2 coronavirus. The peptide fragment was found to be toxic and displayed a high binding propensity for the amyloidogenic segments of Aβ, thereby promoting the aggregation and toxicity of the peptide in vitro and in silico, while retarding the hatching and survival of zebrafish embryos upon exposure. Our study implicated SARS-CoV-2 viral infection as a potential contributor to AD pathogenesis, a little explored area in our quest for understanding and overcoming Long Covid.
Collapse
Affiliation(s)
- Sujian Cao
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
| | - Zhiyuan Song
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | - Jinyu Rong
- College of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Nicholas Andrikopoulos
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Xiufang Liang
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
| | - Yue Wang
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006, China
| | - Guotao Peng
- College of Environmental Science and Engineering, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, USA
| | - Pu Chun Ke
- Nanomedicine Center, The Great Bay Area National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| |
Collapse
|
45
|
Parry PI, Lefringhausen A, Turni C, Neil CJ, Cosford R, Hudson NJ, Gillespie J. 'Spikeopathy': COVID-19 Spike Protein Is Pathogenic, from Both Virus and Vaccine mRNA. Biomedicines 2023; 11:2287. [PMID: 37626783 PMCID: PMC10452662 DOI: 10.3390/biomedicines11082287] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
The COVID-19 pandemic caused much illness, many deaths, and profound disruption to society. The production of 'safe and effective' vaccines was a key public health target. Sadly, unprecedented high rates of adverse events have overshadowed the benefits. This two-part narrative review presents evidence for the widespread harms of novel product COVID-19 mRNA and adenovectorDNA vaccines and is novel in attempting to provide a thorough overview of harms arising from the new technology in vaccines that relied on human cells producing a foreign antigen that has evidence of pathogenicity. This first paper explores peer-reviewed data counter to the 'safe and effective' narrative attached to these new technologies. Spike protein pathogenicity, termed 'spikeopathy', whether from the SARS-CoV-2 virus or produced by vaccine gene codes, akin to a 'synthetic virus', is increasingly understood in terms of molecular biology and pathophysiology. Pharmacokinetic transfection through body tissues distant from the injection site by lipid-nanoparticles or viral-vector carriers means that 'spikeopathy' can affect many organs. The inflammatory properties of the nanoparticles used to ferry mRNA; N1-methylpseudouridine employed to prolong synthetic mRNA function; the widespread biodistribution of the mRNA and DNA codes and translated spike proteins, and autoimmunity via human production of foreign proteins, contribute to harmful effects. This paper reviews autoimmune, cardiovascular, neurological, potential oncological effects, and autopsy evidence for spikeopathy. With many gene-based therapeutic technologies planned, a re-evaluation is necessary and timely.
Collapse
Affiliation(s)
- Peter I. Parry
- Children’s Health Research Clinical Unit, Faculty of Medicine, The University of Queensland, South Brisbane, QLD 4101, Australia
- Department of Psychiatry, College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | - Astrid Lefringhausen
- Children’s Health Defence (Australia Chapter), Huskisson, NSW 2540, Australia; (A.L.); (R.C.); (J.G.)
| | - Conny Turni
- Microbiology Research, QAAFI (Queensland Alliance for Agriculture and Food Innovation), The University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Christopher J. Neil
- Department of Medicine, University of Melbourne, Melbourne, VIC 3010, Australia;
| | - Robyn Cosford
- Children’s Health Defence (Australia Chapter), Huskisson, NSW 2540, Australia; (A.L.); (R.C.); (J.G.)
| | - Nicholas J. Hudson
- School of Agriculture and Food Science, The University of Queensland, Brisbane, QLD 4072, Australia;
| | - Julian Gillespie
- Children’s Health Defence (Australia Chapter), Huskisson, NSW 2540, Australia; (A.L.); (R.C.); (J.G.)
| |
Collapse
|
46
|
Kell DB, Pretorius E. Are fibrinaloid microclots a cause of autoimmunity in Long Covid and other post-infection diseases? Biochem J 2023; 480:1217-1240. [PMID: 37584410 DOI: 10.1042/bcj20230241] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/17/2023]
Abstract
It is now well established that the blood-clotting protein fibrinogen can polymerise into an anomalous form of fibrin that is amyloid in character; the resultant clots and microclots entrap many other molecules, stain with fluorogenic amyloid stains, are rather resistant to fibrinolysis, can block up microcapillaries, are implicated in a variety of diseases including Long COVID, and have been referred to as fibrinaloids. A necessary corollary of this anomalous polymerisation is the generation of novel epitopes in proteins that would normally be seen as 'self', and otherwise immunologically silent. The precise conformation of the resulting fibrinaloid clots (that, as with prions and classical amyloid proteins, can adopt multiple, stable conformations) must depend on the existing small molecules and metal ions that the fibrinogen may (and is some cases is known to) have bound before polymerisation. Any such novel epitopes, however, are likely to lead to the generation of autoantibodies. A convergent phenomenology, including distinct conformations and seeding of the anomalous form for initiation and propagation, is emerging to link knowledge in prions, prionoids, amyloids and now fibrinaloids. We here summarise the evidence for the above reasoning, which has substantial implications for our understanding of the genesis of autoimmunity (and the possible prevention thereof) based on the primary process of fibrinaloid formation.
Collapse
Affiliation(s)
- Douglas B Kell
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kemitorvet 200, 2800 Kgs Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| |
Collapse
|
47
|
Chesney AD, Maiti B, Hansmann UHE. SARS-COV-2 spike protein fragment eases amyloidogenesis of α-synuclein. J Chem Phys 2023; 159:015103. [PMID: 37409768 PMCID: PMC10328560 DOI: 10.1063/5.0157331] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 06/19/2023] [Indexed: 07/07/2023] Open
Abstract
Parkinson's disease is accompanied by the presence of amyloids in the brain that are formed of α-synuclein chains. The correlation between COVID-19 and the onset of Parkinson's disease led to the idea that amyloidogenic segments in SARS-COV-2 proteins can induce aggregation of α-synuclein. Using molecular dynamic simulations, we show that the fragment FKNIDGYFKI of the spike protein, which is unique for SARS-COV-2, preferentially shifts the ensemble of α-synuclein monomer toward rod-like fibril seeding conformations and, at the same time, differentially stabilizes this polymorph over the competing twister-like structure. Our results are compared with earlier work relying on a different protein fragment that is not specific for SARS-COV-2.
Collapse
Affiliation(s)
- Andrew D. Chesney
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Buddhadev Maiti
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, USA
| | - Ulrich H. E. Hansmann
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, USA
| |
Collapse
|
48
|
Milton NGN. SARS-CoV-2 amyloid, is COVID-19-exacerbated dementia an amyloid disorder in the making? FRONTIERS IN DEMENTIA 2023; 2:1233340. [PMID: 39081980 PMCID: PMC11285677 DOI: 10.3389/frdem.2023.1233340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/26/2023] [Indexed: 08/02/2024]
|
49
|
Nunes JM, Kell DB, Pretorius E. Cardiovascular and haematological pathology in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS): A role for viruses. Blood Rev 2023; 60:101075. [PMID: 36963989 PMCID: PMC10027292 DOI: 10.1016/j.blre.2023.101075] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
ME/CFS is a debilitating chronic condition that often develops after viral or bacterial infection. Insight from the study of Long COVID/Post Acute Sequelae of COVID-19 (PASC), the post-viral syndrome associated with SARS-CoV-2 infection, might prove to be useful for understanding pathophysiological mechanisms of ME/CFS. Disease presentation is similar between the two conditions, and a subset of Long COVID patients meet the diagnostic criteria for ME/CFS. Since Long COVID is characterized by significant vascular pathology - including endothelial dysfunction, coagulopathy, and vascular dysregulation - the question of whether or not the same biological abnormalities are of significance in ME/CFS arises. Cardiac abnormalities have for a while now been documented in ME/CFS cohorts, with recent studies demonstrating major deficits in cerebral blood flow, and hence vascular dysregulation. A growing body of research is demonstrating that ME/CFS is accompanied by platelet hyperactivation, anomalous clotting, a procoagulant phenotype, and endothelial dysfunction. Endothelial damage and dysregulated clotting can impair substance exchange between blood and tissues, and result in hypoperfusion, which may contribute to the manifestation of certain ME/CFS symptoms. Here we review the ME/CFS literature to summarize cardiovascular and haematological findings documented in patients with the condition, and, in this context, briefly discuss the potential role of previously-implicated pathogens. Overall, cardiac and haematological abnormalities are present within ME/CFS cohorts. While atherosclerotic heart disease is not significantly associated with ME/CFS, suboptimal cardiovascular function defined by reduced cardiac output, impaired cerebral blood flow, and vascular dysregulation are, and these abnormalities do not appear to be influenced by deconditioning. Rather, these cardiac abnormalities may result from dysfunction in the (autonomic) nervous system. Plenty of recently published studies are demonstrating significant platelet hyperactivity and endothelial dysfunction in ME/CFS, as well as anomalous clotting processes. It is of particular importance to determine to what extent these cardiovascular and haematological abnormalities contribute to symptom severity, and if these two systems can be targeted for therapeutic purposes. Viral reservoirs of herpesviruses exist in ME/CFS, and most likely contribute to cardiovascular and haematological dysfunction directly or indirectly. This review highlights the potential of studying cardiac functioning, the vasculature, and coagulation system in ME/CFS.
Collapse
Affiliation(s)
- Jean M Nunes
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa.
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa; Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool L69 7ZB, UK; The Novo Nordisk Foundation Centre for Biosustainability, Building 220, Chemitorvet 200, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa; Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool L69 7ZB, UK.
| |
Collapse
|
50
|
Del Carpio-Orantes L. Etiopathogenic theories about long COVID. World J Virol 2023; 12:204-208. [PMID: 37396704 PMCID: PMC10311581 DOI: 10.5501/wjv.v12.i3.204] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/08/2023] [Accepted: 05/12/2023] [Indexed: 06/21/2023] Open
Abstract
The main etiopathogenic theories of long coronavirus disease (COVID) are listed and a conjunction of them is carried out with the objective of deciphering the pathophysiology of the entity, finally the main lines of treatment existing in real life are discussed (Paxlovid, use of antibiotics in dysbiosis, triple anticoagulant therapy, temelimab).
Collapse
Affiliation(s)
- Luis Del Carpio-Orantes
- Department of Internal Medicine, Virology, Research, Study Group for the Diagnosis and Treatment of COVID-19 in Veracruz, Veracruz 91900, Veracruz, Mexico
| |
Collapse
|