1
|
Praveen A, Dougnon G, Matsui H. Exploring α-Syn's Functions Through Ablation Models: Physiological and Pathological Implications. Cell Mol Neurobiol 2025; 45:44. [PMID: 40389720 PMCID: PMC12089638 DOI: 10.1007/s10571-025-01560-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 05/07/2025] [Indexed: 05/21/2025]
Abstract
A significant advancement in neurodegenerative research was the discovery that α-synuclein (α-Syn/SNCA) plays a part in the pathophysiology of Parkinson's disease (PD). Decades later, the protein's significant impacts on various brain disorders are still being extensively explored. In disease conditions, α-Syn misfolds and forms abnormal aggregates that accumulate in neurons, thus triggering various organellar dysfunctions and ultimately neurodegeneration. These misfolded forms are highly heterogeneous and vary significantly among different synucleinopathies, such as PD, Multiple System Atrophy, or Dementia with Lewy bodies. Though initially believed to be exclusively localized in the brain, numerous pieces of evidence suggest that α-Syn functions transcend the central nervous system, with roles in peripheral functions, such as modulation of immune responses, hematopoiesis, and gastrointestinal regulation. Here, we aim to provide a detailed compilation of cellular functions and pathological phenotypes that are altered upon attenuation of α-Syn function in vitro and in vivo and explore the effects of SNCA gene silencing in healthy and disease states using cellular and animal models.
Collapse
Affiliation(s)
- Anjali Praveen
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Godfried Dougnon
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Hideaki Matsui
- Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan.
| |
Collapse
|
2
|
Lungu R, Fernandes FF, Pires Monteiro S, Outeiro TF, Shemesh N. Neural and vascular contributions to sensory impairments in a human alpha-synuclein transgenic mouse model of Parkinson's disease. J Cereb Blood Flow Metab 2025:271678X251338952. [PMID: 40334688 PMCID: PMC12058711 DOI: 10.1177/0271678x251338952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/06/2025] [Accepted: 04/14/2025] [Indexed: 05/09/2025]
Abstract
Parkinson's disease (PD) is a complex progressive neurodegenerative disorder involving hallmarks such as α -Synuclein (α Syn) aggregation and dopaminergic dysfunction that affect brain-wide neural activity. Although movement disorders are prominent in PD, sensory impairments also occur relatively early on, mainly in olfactory and, to a lesser extent visual systems. While these deficits have been described mainly at the behavioral and molecular levels, the underlying network-level activity remains poorly understood. Here, we harnessed a human α Syn transgenic mouse model of PD with in vivo functional MRI (fMRI) to map evoked activity in the visual and olfactory pathways, along with pseudo-Continuous Arterial Spin Labeling (pCASL) and c-FOS measurements to disentangle vascular from neuronal effects. Upon stimulation with either odors or flickering lights, we found significant decreases in fMRI responses along both olfactory and visual pathways, in multiple cortical and subcortical sensory areas. Average Cerebral Blood Flow rates were decreased by ∼10% in the α Syn group, while c-FOS levels were reduced by over 50%, suggesting a strong neural driver for the dysfunction, along with more modest vascular contributions. Our study provides insight into brain-level activity in an α Syn-based model, and suggests a novel target for biomarking via quantification of simple sensory evoked responses.
Collapse
Affiliation(s)
- Ruxanda Lungu
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| | | | - Sara Pires Monteiro
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
- Institute for Systems and Robotics - Lisboa and Department of Bioengineering, Instituto Superior Técnico – Universidade de Lisboa, Lisbon, Portugal
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, University Medical Center Göttingen, Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
- Scientific Employee with an Honorary Contract at German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
| | - Noam Shemesh
- Champalimaud Research, Champalimaud Foundation, Lisbon, Portugal
| |
Collapse
|
3
|
Singh KS, Verma R, Singh N, Singh LR, Gupta A. Factors responsible for alpha-Synuclein aggregation. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 211:271-292. [PMID: 39947752 DOI: 10.1016/bs.pmbts.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Aggregation of α-Synuclein (α-Syn) is the hallmark of the pathophysiology of Parkinson's disease. Apart from aggregates, α-Syn can exist in multiple abnormal forms such as oligomers, protofibrils, fibrils amorphous aggregates etc. These forms initiate aggressive, selective and progressive neuronal atrophy through various modes such as mitochondrial dysfunction, lysosomal malfunction, and disruption of calcium homeostasis in various α-Syn-related neurodegenerative disorders. Structurally α-Syn is divided into three domains: N-terminal region made by amino acids1-67 (amphipathic, lysine-rich and interacts with acidic lipid membranes), Non-amyloid-β component (NAC) region made by amino acids 67-95 (hydrophobic region, central to α-syn aggregation) and C-terminal region made by amino acids 96-140 (acidic and proline-rich region responsible for interaction with other proteins). α-Syn follows the pattern of a typical intrinsically disordered protein and lacks a proper folded conformation and exist majorly in a random coil form, though on lipid binding the protein assumes an α-helical structure. The central random coil region of α-Syn is involved in fibril formation transforming into β-sheet rich secondary structures which is a characteristic of amyloids. This chapter entails an elaborate explanation of factors influencing the structure, function and aggregation of α-Syn. Major factors being abnormally high physiological expression of the protein, mutations, posttranslational modifications and also interactions with small molecules such as osmolytes in the cellular milieu. Studying the factors responsible for misfolding and aggregation of α-Syn along with the mechanism involved is crucial to understanding their implications in Parkinson's disease, and will yield valuable insights into disease mechanisms, potential therapeutic strategies.
Collapse
Affiliation(s)
| | - Rahul Verma
- Dr. B. R. Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India
| | - Nagendra Singh
- Gautam Buddha University, Greater Noida, Gautam Budh Nagar, Uttar Pradesh, India
| | | | - Akshita Gupta
- Dr. B. R. Ambedkar Centre for Biomedical Research, University of Delhi, Delhi, India.
| |
Collapse
|
4
|
Mekala S, Wu Y, Li YM. Strategies of positron emission tomography (PET) tracer development for imaging of tau and α-synuclein in neurodegenerative disorders. RSC Med Chem 2024:d4md00576g. [PMID: 39678127 PMCID: PMC11638850 DOI: 10.1039/d4md00576g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder, characterized by the presence of extracellular amyloid plaques consisting of β-amyloid peptides (Aβ) and intracellular neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau (pTau) protein in the brain. Genetic and animal studies strongly indicate that Aβ, tau and neuroinflammation play important roles in the pathogenesis of AD. Several staging models showed that NFTs correlated well with the disease progression. Positron emission tomography (PET) imaging has become a widely used non-invasive technique to image NFTs for early diagnosis of AD. Despite the remarkable progress made over the past few years, tau PET imaging is still challenging due to the nature of tau pathology and the technical aspects of PET imaging. Tau pathology often coexists with other proteinopathies, such as Aβ plaques and α-synuclein aggregates. Distinguishing tau-specific signals from other overlapping pathologies is difficult, especially in the context of AD, where multiple protein aggregates are present, as well as the spectrum of different tau isoforms (3R and 4R) and conformations. Moreover, tracers should ideally have optimal pharmacokinetic properties to penetrate the blood-brain barrier (BBB) while maintaining specificity, low toxicity, low non-specific binding, rapid uptake and clearance from the brain, and formation of no radiolabeled metabolites in the brain. On the other hand, Parkinson's disease (PD) is a progressive neurodegenerative movement disorder characterized by the abnormal accumulations of α-synuclein in neurons. Heterogeneity and the unclear pathogenesis of PD hinder early and accurate diagnosis of the disease for therapeutic development in clinical use. In this review, while referring to existing reviews, we focus on the design strategies and current progress in tau (NFTs) targeting new PET tracers for AD; evolution of non-AD tau targeting PET tracers for applications including progressive supranuclear paralysis (PSP) and corticobasal degeneration (CBD); new PET tracer development for α-synuclein aggregate imaging in PD and giving an outlook for future PET tracer development.
Collapse
Affiliation(s)
- Shekar Mekala
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center 1275 York Avenue New York New York 10065 USA
| | - You Wu
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center 1275 York Avenue New York New York 10065 USA
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center New York New York 10065 USA
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan-Kettering Cancer Center 1275 York Avenue New York New York 10065 USA
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center New York New York 10065 USA
| |
Collapse
|
5
|
Fırtına ÖB, Salt Ö, Sayhan MB, Dibirdik I, Yucal A. Value of plasma alpha- and beta-synuclein levels in the diagnosis, severity, and functional outcome of acute ischemic stroke. Turk J Emerg Med 2024; 24:238-244. [PMID: 39564445 PMCID: PMC11573173 DOI: 10.4103/tjem.tjem_17_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 11/21/2024] Open
Abstract
OBJECTIVE We aimed to determine the role of plasma alpha- and beta-synuclein levels and other routine inflammatory parameters in the diagnosis, outcome, and mortality of acute ischemic stroke (AIS). METHODS In our study, serum alpha- and beta-synuclein levels and clinical data were prospectively evaluated in 93 subjects (43 controls and 50 AIS patients) admitted to the emergency department. The outcome status and prognostic classification were performed according to the modified Rankin Scale (mRS) scores on the 30th day from hospital admission. RESULTS The mean age of the subjects was 70.6 ± 11 years. Thirty-eight percentage were female. Plasma α-synuclein levels in the AIS group (33.6 ± 8.5 ng/mL) were significantly higher than those in the control group (4.22 ± 2.1 ng/mL) (P < 0.001). Plasma β-synuclein levels in the AIS group (13.07 ± 2.7 ng/mL) were significantly higher than those in the control group (2.17 ± 1.4 ng/mL) (P < 0.001). There was no significant difference in alpha- and beta-synuclein levels between the subgroups formed according to the 30th-day results of the patients using the mRS scores (P = 0.813 and 0.812, respectively). CONCLUSION The serum alpha- and beta-synuclein concentrations of patients with AIS at admission were significantly higher than the healthy control group. At admission, serum alpha- and beta-synuclein levels do not have definitive clinically predictive value in predicting stroke progression and outcome in patients with AIS.
Collapse
Affiliation(s)
- Özge Başak Fırtına
- Department of Emergency Medicine, Mardin Public Hospital, Mardin, Turkey
| | - Ömer Salt
- Department of Emergency Medicine, Kayseri City Hospital, Kayseri, Turkey
| | - Mustafa Burak Sayhan
- Department of Emergency Medicine, Trakya University School of Medicine, Edirne, Turkey
| | - Ilker Dibirdik
- Department of Medical Biochemistry, Trakya University School of Medicine, Edirne, Turkey
| | - Aykut Yucal
- Department of Emergency Medicine, Trakya University School of Medicine, Edirne, Turkey
| |
Collapse
|
6
|
Lecht S, Lahiani A, Klazas M, Naamneh MS, Rubin L, Dong J, Zheng W, Lazarovici P. Rasagiline Exerts Neuroprotection towards Oxygen-Glucose-Deprivation/Reoxygenation-Induced GAPDH-Mediated Cell Death by Activating Akt/Nrf2 Signaling. Biomedicines 2024; 12:1592. [PMID: 39062165 PMCID: PMC11275171 DOI: 10.3390/biomedicines12071592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Rasagiline (Azilect®) is a selective monoamine oxidase B (MAO-B) inhibitor that provides symptomatic benefits in Parkinson's disease (PD) treatment and has been found to exert preclinical neuroprotective effects. Here, we investigated the neuroprotective signaling pathways of acute rasagiline treatment for 22 h in PC12 neuronal cultures exposed to oxygen-glucose deprivation (OGD) for 4 h, followed by 18 h of reoxygenation (R), causing 40% aponecrotic cell death. In this study, 3-10 µM rasagiline induced dose-dependent neuroprotection of 20-80%, reduced the production of the neurotoxic reactive oxygen species by 15%, and reduced the nuclear translocation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) by 75-90%. In addition, 10 µM rasagiline increased protein kinase B (Akt) phosphorylation by 50% and decreased the protein expression of the ischemia-induced α-synuclein protein by 50% in correlation with the neuroprotective effect. Treatment with 1-5 µM rasagiline induced nuclear shuttling of transcription factor Nrf2 by 40-90% and increased the mRNA levels of the antioxidant enzymes heme oxygenase-1, (NAD (P) H- quinone dehydrogenase, and catalase by 1.8-2.0-fold compared to OGD/R insult. These results indicate that rasagiline provides neuroprotection to the ischemic neuronal cultures through the inhibition of α-synuclein and GAPDH-mediated aponecrotic cell death, as well as via mitochondrial protection, by increasing mitochondria-specific antioxidant enzymes through a mechanism involving the Akt/Nrf2 redox-signaling pathway. These findings may be exploited for neuroprotective drug development in PD and stroke therapy.
Collapse
Affiliation(s)
- Shimon Lecht
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| | - Adi Lahiani
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| | - Michal Klazas
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| | - Majdi Saleem Naamneh
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| | - Limor Rubin
- Allergy and Clinical Immunology Unit, Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel
| | - Jiayi Dong
- Center of Reproduction, Development & Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Wenhua Zheng
- Center of Reproduction, Development & Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112002, Israel
| |
Collapse
|
7
|
Wu YC, Bogale TA, Koistinaho J, Pizzi M, Rolova T, Bellucci A. The contribution of β-amyloid, Tau and α-synuclein to blood-brain barrier damage in neurodegenerative disorders. Acta Neuropathol 2024; 147:39. [PMID: 38347288 PMCID: PMC10861401 DOI: 10.1007/s00401-024-02696-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/15/2024]
Abstract
Central nervous system (CNS) accumulation of fibrillary deposits made of Amyloid β (Aβ), hyperphosphorylated Tau or α-synuclein (α-syn), present either alone or in the form of mixed pathology, characterizes the most common neurodegenerative diseases (NDDs) as well as the aging brain. Compelling evidence supports that acute neurological disorders, such as traumatic brain injury (TBI) and stroke, are also accompanied by increased deposition of toxic Aβ, Tau and α-syn species. While the contribution of these pathological proteins to neurodegeneration has been experimentally ascertained, the cellular and molecular mechanisms driving Aβ, Tau and α-syn-related brain damage remain to be fully clarified. In the last few years, studies have shown that Aβ, Tau and α-syn may contribute to neurodegeneration also by inducing and/or promoting blood-brain barrier (BBB) disruption. These pathological proteins can affect BBB integrity either directly by affecting key BBB components such as pericytes and endothelial cells (ECs) or indirectly, by promoting brain macrophages activation and dysfunction. Here, we summarize and critically discuss key findings showing how Aβ, Tau and α-syn can contribute to BBB damage in most common NDDs, TBI and stroke. We also highlight the need for a deeper characterization of the role of these pathological proteins in the activation and dysfunction of brain macrophages, pericytes and ECs to improve diagnosis and treatment of acute and chronic neurological disorders.
Collapse
Affiliation(s)
- Ying-Chieh Wu
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Tizibt Ashine Bogale
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, BS, Italy
- Department of Acute Brain and Cardiovascular Injury, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Jari Koistinaho
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Marina Pizzi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, BS, Italy
| | - Taisia Rolova
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, BS, Italy.
| |
Collapse
|
8
|
Feng L, Sharma A, Wang Z, Muresanu DF, Tian ZR, Lafuente JV, Buzoianu AD, Nozari A, Wiklund L, Sharma HS. Co-administration of Nanowired DL-3-n-Butylphthalide (DL-NBP) Together with Mesenchymal Stem Cells, Monoclonal Antibodies to Alpha Synuclein and TDP-43 (TAR DNA-Binding Protein 43) Enhance Superior Neuroprotection in Parkinson's Disease Following Concussive Head Injury. ADVANCES IN NEUROBIOLOGY 2023; 32:97-138. [PMID: 37480460 DOI: 10.1007/978-3-031-32997-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
dl-3-n-butylphthalide (dl-NBP) is one of the potent antioxidant compounds that induces profound neuroprotection in stroke and traumatic brain injury. Our previous studies show that dl-NBP reduces brain pathology in Parkinson's disease (PD) following its nanowired delivery together with mesenchymal stem cells (MSCs) exacerbated by concussive head injury (CHI). CHI alone elevates alpha synuclein (ASNC) in brain or cerebrospinal fluid (CSF) associated with elevated TAR DNA-binding protein 43 (TDP-43). TDP-43 protein is also responsible for the pathologies of PD. Thus, it is likely that exacerbation of brain pathology in PD following brain injury may be thwarted using nanowired delivery of monoclonal antibodies (mAb) to ASNC and/or TDP-43. In this review, the co-administration of dl-NBP with MSCs and mAb to ASNC and/or TDP-43 using nanowired delivery in PD and CHI-induced brain pathology is discussed based on our own investigations. Our observations show that co-administration of TiO2 nanowired dl-NBP with MSCs and mAb to ASNC with TDP-43 induced superior neuroprotection in CHI induced exacerbation of brain pathology in PD, not reported earlier.
Collapse
Affiliation(s)
- Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Zhongshan Road (West), Shijiazhuang, Hebei Province, China
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Zhenguo Wang
- Shijiazhuang Pharma Group NBP Pharmaceutical Co., Ltd., Shijiazhuang, Hebei Province, China
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania
- "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, USA
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ala Nozari
- Anesthesiology & Intensive Care, Chobanian & Avedisian School of Medicine, Boston University, Boston, MA, USA
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
9
|
Piccarducci R, Caselli MC, Zappelli E, Ulivi L, Daniele S, Siciliano G, Ceravolo R, Mancuso M, Baldacci F, Martini C. The Role of Amyloid-β, Tau, and α-Synuclein Proteins as Putative Blood Biomarkers in Patients with Cerebral Amyloid Angiopathy. J Alzheimers Dis 2022; 89:1039-1049. [PMID: 35964181 DOI: 10.3233/jad-220216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cerebral amyloid angiopathy (CAA) is a cerebrovascular disorder characterized by the deposition of amyloid-β protein (Aβ) within brain blood vessels that develops in elderly people and Alzheimer's disease (AD) patients. Therefore, the investigation of biomarkers able to differentiate CAA patients from AD patients and healthy controls (HC) is of great interest, in particular in peripheral fluids. OBJECTIVE The current study aimed to detect the neurodegenerative disease (ND)-related protein (i.e., Aβ 1 - 40, Aβ 1 - 42, tau, and α-synuclein) levels in both red blood cells (RBCs) and plasma of CAA patients and HC, evaluating their role as putative peripheral biomarkers for CAA. METHODS For this purpose, the proteins' concentration was quantified in RBCs and plasma by homemade immunoenzymatic assays in an exploratory cohort of 20 CAA patients and 20 HC. RESULTS The results highlighted a significant increase of Aβ 1 - 40 and α-synuclein concentrations in both RBCs and plasma of CAA patients, while higher Aβ 1 - 42 and t-tau levels were detected only in RBCs of CAA individuals compared to HC. Moreover, Aβ 1 - 42/Aβ 1 - 40 ratio increased in RBCs and decreased in plasma of CAA patients. The role of these proteins as candidate peripheral biomarkers easily measurable with a blood sample in CAA needs to be confirmed in larger studies. CONCLUSION In conclusion, we provide evidence concerning the possible use of blood biomarkers for contributing to CAA diagnosis and differentiation from other NDs.
Collapse
Affiliation(s)
| | - Maria Chiara Caselli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Leonardo Ulivi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Michelangelo Mancuso
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | |
Collapse
|
10
|
Galkin AP, Sysoev EI. Stress Response Is the Main Trigger of Sporadic Amyloidoses. Int J Mol Sci 2021; 22:4092. [PMID: 33920986 PMCID: PMC8071232 DOI: 10.3390/ijms22084092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 12/11/2022] Open
Abstract
Amyloidoses are a group of diseases associated with the formation of pathological protein fibrils with cross-β structures. Approximately 5-10% of the cases of these diseases are determined by amyloidogenic mutations, as well as by transmission of infectious amyloids (prions) between organisms. The most common group of so-called sporadic amyloidoses is associated with abnormal aggregation of wild-type proteins. Some sporadic amyloidoses are known to be induced only against the background of certain pathologies, but in some cases the cause of amyloidosis is unclear. It is assumed that these diseases often occur by accident. Here we present facts and hypotheses about the association of sporadic amyloidoses with vascular pathologies, trauma, oxidative stress, cancer, metabolic diseases, chronic infections and COVID-19. Generalization of current data shows that all sporadic amyloidoses can be regarded as a secondary event occurring against the background of diseases provoking a cellular stress response. Various factors causing the stress response provoke protein overproduction, a local increase in the concentration or modifications, which contributes to amyloidogenesis. Progress in the treatment of vascular, metabolic and infectious diseases, as well as cancers, should lead to a significant reduction in the risk of sporadic amyloidoses.
Collapse
Affiliation(s)
- Alexey P. Galkin
- St. Petersburg Branch, Vavilov Institute of General Genetics, 199034 St. Petersburg, Russia
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia;
| | - Evgeniy I. Sysoev
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia;
| |
Collapse
|