1
|
Beura SK, Panigrahi AR, Yadav P, Kulkarni PP, Lakhanpal V, Singh B, Singh SK. Role of Thrombosis in Neurodegenerative Diseases: An Intricate Mechanism of Neurovascular Complications. Mol Neurobiol 2025; 62:4802-4836. [PMID: 39482419 DOI: 10.1007/s12035-024-04589-4] [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: 04/11/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024]
Abstract
Thrombosis, the formation of blood clots in arteries or veins, poses a significant health risk by disrupting the blood flow. It can potentially lead to major cardiovascular complications such as acute myocardial infarction or ischemic stroke (arterial thrombosis) and deep vein thrombosis or pulmonary embolism (venous thrombosis). Nevertheless, over the course of several decades, researchers have observed an association between different cardiovascular events and neurodegenerative diseases, which progressively harm and impair parts of the nervous system, particularly the brain. Furthermore, thrombotic complications have been identified in numerous clinical instances of neurodegenerative diseases, particularly Alzheimer's disease, Parkinson's disease, multiple sclerosis, and Huntington's disease. Substantial research indicates that endothelial dysfunction, vascular inflammation, coagulation abnormalities, and platelet hyperactivation are commonly observed in these conditions, collectively contributing to an increased risk of thrombosis. Thrombosis can, in turn, contribute to the onset, pathogenesis, and severity of these neurological disorders. Hence, this concise review comprehensively explores the correlation between cardiovascular diseases and neurodegenerative diseases, elucidating the cellular and molecular mechanisms of thrombosis in these neurodegenerative diseases. Additionally, a detailed discussion is provided on the commonly employed antithrombotic medications in the context of these neuronal diseases.
Collapse
Affiliation(s)
- Samir Kumar Beura
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401
| | | | - Pooja Yadav
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401
| | - Paresh P Kulkarni
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Vikas Lakhanpal
- Department of Neurology, All India Institute of Medical Sciences, Bathinda, Punjab, India, 151001
| | - Bhupinder Singh
- Department of Cardiology, All India Institute of Medical Sciences, Bathinda, Punjab, India, 151001
| | - Sunil Kumar Singh
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401.
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India, 151401.
| |
Collapse
|
2
|
Ercan H, Reumiller CM, Mühlberger J, Hsu F, Schmidt GJ, Umlauf E, Miller I, Rappold E, Attems J, Oehler R, Zellner M. Platelets mirror changes in the frontal lobe antioxidant system in Alzheimer's disease. Alzheimers Dement 2025; 21:e70117. [PMID: 40189792 PMCID: PMC11972982 DOI: 10.1002/alz.70117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 04/10/2025]
Abstract
INTRODUCTION Blood biomarkers reflecting Alzheimer's disease (AD) pathophysiology can improve diagnosis and treatment. METHODS We applied top-down proteomics to compare frontal lobe from 17 AD cases and 11 controls to blood platelets from a second independent study group of 124 AD patients, 61 with mild cognitive impairment (MCI), and 168 controls. Findings were immunologically validated. RESULTS Sixty AD-associated proteoforms were identified in frontal lobe, with 26 identically represented in platelets. Validation in platelet samples confirmed elevated glutathione S-transferase omega 1 (GSTO1) levels linked to single nucleotide polymorphism (SNP) rs4925 and increased superoxide dismutase 1 (SOD1) levels in AD. Bioinformatics revealed copper chaperone for superoxide dismutase (CCS) and glutathione peroxidase 1 (GPX1) as integral partners of these antioxidant enzymes. Both were detected to be reduced in frontal lobes and platelets in AD. SOD1 and CCS are already changed in MCI. DISCUSSION These four novel blood biomarkers, integrated with traditional AD biomarkers, may facilitate patient risk assessment and treatment, with SOD1 and CCS alterations in MCI offering early diagnostic potential. HIGHLIGHTS Platelets mirror several Alzheimer's disease (AD)-dependent neuronal changes, valuable for blood tests. As a start, 60 AD-associated frontal lobe proteins were identified by top-down proteomics. Fifty percent of these 60 AD-affected brain proteins are represented identically in platelets. Among these, glutathione S-transferase omega 1 (GSTO1), superoxide dismutase 1 (SOD1), copper chaperone for superoxide dismutase (CCS), and glutathione peroxidase 1 (GPX1) are identically AD related in brain and platelets. SOD1 and its crucial activating partner CCS are altered in the platelets of patients with mild cognitive impairment.
Collapse
Affiliation(s)
- Huriye Ercan
- Institute of Vascular Biology and Thrombosis ResearchCentre for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Christina Maria Reumiller
- Institute of Vascular Biology and Thrombosis ResearchCentre for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Jacqueline Mühlberger
- Institute of Vascular Biology and Thrombosis ResearchCentre for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Felicia Hsu
- Institute of Vascular Biology and Thrombosis ResearchCentre for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | | | - Ellen Umlauf
- Institute of Vascular Biology and Thrombosis ResearchCentre for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Ingrid Miller
- Department of Biological Sciences and PathobiologyUniversity of Veterinary Medicine ViennaViennaAustria
| | - Eduard Rappold
- Institute of Vascular Biology and Thrombosis ResearchCentre for Physiology and PharmacologyMedical University of ViennaViennaAustria
| | - Johannes Attems
- Translational and Clinical Research InstituteCampus for Ageing and VitalityNewcastle UniversityNewcastleUK
| | - Rudolf Oehler
- Department of General SurgeryDivision of Visceral SurgeryMedical University of ViennaViennaAustria
| | - Maria Zellner
- Institute of Vascular Biology and Thrombosis ResearchCentre for Physiology and PharmacologyMedical University of ViennaViennaAustria
| |
Collapse
|
3
|
Goubran H, Ahmed S, Ragab G, Seghatchian J, Burnouf T. Platelet proteomics: Clinical implications - Decoding the black box! Transfus Apher Sci 2025; 64:104060. [PMID: 39719751 DOI: 10.1016/j.transci.2024.104060] [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: 12/26/2024]
Abstract
Platelets are anucleate blood cells traditionally associated with hemostasis but now increasingly recognized for their multifaceted roles in immunity, inflammation, and tissue repair. Advances in platelet proteomics, employing high-throughput techniques such as mass spectrometry, have significantly enhanced our understanding of platelet biology and its clinical implications in transfusion medicine. Platelet proteomics offers a retrospective view of physiological and pathological changes over the platelet's 7-10-day lifespan, making it a unique tool for studying cumulative biological events. Recent applications include the identification of biomarkers for cardiovascular, infectious, autoimmune diseases and cancer. In neurodegeneration and aging, platelets have been explored for their shared molecular pathways with neurons, with findings implicating Tau, amyloid-beta, and alpha-synuclein as potential biomarkers. Proteomics is also emerging as an important factor in the development of evidence-based, tailor-made platelet-derived therapies. While promising, platelet proteomics requires further standardization and computational advances to support transitioning from research to routine clinical practice.
Collapse
Affiliation(s)
- Hadi Goubran
- Saskatoon Cancer Centre, Saskatoon, SK, Canada; Department of Oncology, College of Medicine, University of Saskatchewan, Saskatchewan, Canada
| | - Shahid Ahmed
- Saskatoon Cancer Centre, Saskatoon, SK, Canada; Department of Oncology, College of Medicine, University of Saskatchewan, Saskatchewan, Canada
| | - Gaafar Ragab
- Rheumatology and Immunology Unit, Internal Medicine Department, Cairo University, Egypt
| | - Jerard Seghatchian
- International Consultancy in Modern Personalized Blood Components Therapies and Innovative DDR Strategies, London, England, UK
| | - Thierry Burnouf
- Graduate Institute of Biological Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan; International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
4
|
Dhauria M, Mondal R, Deb S, Shome G, Chowdhury D, Sarkar S, Benito-León J. Blood-Based Biomarkers in Alzheimer's Disease: Advancing Non-Invasive Diagnostics and Prognostics. Int J Mol Sci 2024; 25:10911. [PMID: 39456697 PMCID: PMC11507237 DOI: 10.3390/ijms252010911] [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: 09/14/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD), the most prevalent form of dementia, is expected to rise dramatically in incidence due to the global population aging. Traditional diagnostic approaches, such as cerebrospinal fluid analysis and positron emission tomography, are expensive and invasive, limiting their routine clinical use. Recent advances in blood-based biomarkers, including amyloid-beta, phosphorylated tau, and neurofilament light, offer promising non-invasive alternatives for early AD detection and disease monitoring. This review synthesizes current research on these blood-based biomarkers, highlighting their potential to track AD pathology and enhance diagnostic accuracy. Furthermore, this review uniquely integrates recent findings on protein-protein interaction networks and microRNA pathways, exploring novel combinations of proteomic, genomic, and epigenomic biomarkers that provide new insights into AD's molecular mechanisms. Additionally, we discuss the integration of these biomarkers with advanced neuroimaging techniques, emphasizing their potential to revolutionize AD diagnostics. Although large-scale validation is still needed, these biomarkers represent a critical advancement toward more accessible, cost-effective, and early diagnostic tools for AD.
Collapse
Affiliation(s)
| | - Ritwick Mondal
- Department of Clinical Pharmacology and Therapeutic Medicine, IPGMER and SSKM Hospital, Kolkata 700020, India;
| | - Shramana Deb
- Department of Stroke Medicine, Institute of Neuroscience, Kolkata 700017, India;
| | - Gourav Shome
- Department of Biological Sciences, Bose Institute, Kolkata 700054, India;
| | - Dipanjan Chowdhury
- Department of Internal Medicine, IPGMER and SSKM Hospital, Kolkata 700020, India; (D.C.); (S.S.)
| | - Shramana Sarkar
- Department of Internal Medicine, IPGMER and SSKM Hospital, Kolkata 700020, India; (D.C.); (S.S.)
| | - Julián Benito-León
- Department of Neurology, University Hospital “12 de Octubre”, ES-28041 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), ES-28041 Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ES-28029 Madrid, Spain
- Department of Medicine, Complutense University, ES-28040 Madrid, Spain
| |
Collapse
|
5
|
Hajimohammadi S, Soodi M, Hajimehdipoor H, Sefidbakht S, Mashhadi Sharif N. Ferulago Angulata methanolic extract ameliorates scopolamine-induced memory impairment through the inhibition of hippocampal monoamine oxidase activity. Metab Brain Dis 2024; 39:691-703. [PMID: 38722561 DOI: 10.1007/s11011-024-01353-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 05/04/2024] [Indexed: 07/10/2024]
Abstract
Ferulago angulata is a medicinal herb from the Apiaceae family known for its antioxidant, anti-apoptotic, and neuroprotective properties. This study aimed to assess the effects of F. angulata extract on neurobehavioral and biochemical parameters in scopolamine-induced amnesic rats. Fifty-six male Wistar rats were divided into seven groups and orally treated with F. angulata extract (100, 200, 400 mg/kg) and Rivastigmine (1.5 mg/kg) for 10 days. Starting on the sixth day of treatment, the Morris water maze behavioral study was conducted to evaluate cognitive function, with scopolamine administered 30 min before training. Biochemical assays, including monoamine oxidase and oxidative stress measures, were performed on hippocampal tissue. Results showed that extract treatment significantly attenuated scopolamine-induced memory impairment in a dose-dependent manner. Following scopolamine administration, malondialdehyde levels and monoamine oxidase A/B activity increased, while total thiol content and catalase activity decreased compared to the control group. Pretreatment with F. angulata extracts ameliorated the scopolamine-induced impairment in all factors. Toxicological evaluation of liver, lung, heart, and kidney tissues did not indicate any side effects at high doses. The total extract of F. angulata prevents scopolamine-induced learning and memory impairment through antioxidant mechanisms and inhibition of monoamine oxidase. These results suggest that F. angulata extract is effective in the scopolamine model and could be a promising agent for preventing dementia, especially Alzheimer's disease.
Collapse
Affiliation(s)
- Samaneh Hajimohammadi
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maliheh Soodi
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
- Institute for Natural Products and Medicinal Plants, Tarbiat Modares University, Tehran, Iran.
| | - Homa Hajimehdipoor
- Traditional Medicine and Materia Medica Research Center and Department of Traditional Pharmacy, School of Traditional Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Salma Sefidbakht
- Department of Pathology, School of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
6
|
Faydalı N, Arpacı ÖT. Benzimidazole and Benzoxazole Derivatives Against Alzheimer's Disease. Chem Biodivers 2024; 21:e202400123. [PMID: 38494443 DOI: 10.1002/cbdv.202400123] [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: 01/15/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 03/19/2024]
Abstract
Benzimidazole and benzoxazole derivatives are included in the category of medical drugs in a wide range of areas such as anticancer, anticoagulant, antihypertensive, anti- inflammatory, antimicrobial, antiparasitic, antiviral, antioxidant, immunomodulators, proton pump inhibitors, hormone modulators, etc. Many researchers have focused on synthesizing more effective benzimidazole and benzoxazole derivatives for screening various biological activities. In addition, there are benzimidazole and benzoxazole rings as bioisosteres of aromatic rings found in drugs used in the treatment of Alzheimer's disease. Because of the diverse activity of the benzimidazole and benzoxazole rings and bioisosteres marketed as drugs for Alzheimer Diseases, designed compounds containing these rings are likely to be effective against Alzheimer's disease. In this study, the effectiveness of compounds containing benzimidazole and benzoxazole rings against Alzheimer's disease will be examined.
Collapse
Affiliation(s)
- Nagihan Faydalı
- Department of Pharmaceutical Chemistry, Selcuk University, 42250, Konya, Turkey
- Graduate School of Health Sciences, Ankara University, 06110, Ankara, Turkey
| | - Özlem Temiz Arpacı
- Department of Pharmaceutical Chemistry, Ankara University, 06560, Ankara, Turkey
| |
Collapse
|
7
|
Xie Z, Liu Y, Huang M, Zhong S, Lai W. Effects of antidiabetic agents on platelet characteristics with implications in Alzheimer's disease: Mendelian randomization and colocalization study. Heliyon 2024; 10:e30909. [PMID: 38778961 PMCID: PMC11108824 DOI: 10.1016/j.heliyon.2024.e30909] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Background Observational studies have found a potential link between the use of thiazolidinediones (TZDs) and a lower risk of Alzheimer's disease (AD) development. Platelets were the great source of amyloid-β (Aβ) and involved in the development of AD. This study aimed to assess the correlation between antidiabetic agents and platelet characteristics, hoping to provide a potential mechanism of TZDs neuroprotection in AD. Method Drug-targeted Mendelian randomization (MR) was performed to systematically illustrate the long-term effects of antidiabetic agents on platelet characteristics. Four antidiabetic agent targets were considered. Positive control analysis for type 2 diabetes (T2D) was conducted to validate the selection of instrumental variables (IVs). Colocalization analysis was used to further strengthen the robustness of the results. Result Positive control analysis showed an association of four antidiabetic agents with lower risk of T2D, which was consistent with their mechanisms of action and previous evidence from clinical trials. Genetically proxied TZDs were associated with lower platelet count (β[IRNT] = -0.410 [95 % CI -0.533 to -0.288], P = 5.32E-11) and a lower plateletcrit (β[IRNT] = -0.344 [95 % CI -0.481 to -0.206], P = 1.04E-6). Colocalization suggested the posterior probability of hypothesis 4 (PPH4) > 0.8, which further strengthened the MR results. Conclusion Genetically proxied TZDs were causally associated with lower platelet characteristics, particularly platelet count and plateletcrit, providing insight into the involvement of platelet-related pathways in the neuroprotection of TZDs against AD. Future studies are warranted to reveal the underlying molecular mechanism of TZDs' neuroprotective effects through platelet pathways.
Collapse
Affiliation(s)
- Zhipeng Xie
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Yijie Liu
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Min Huang
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shilong Zhong
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Weihua Lai
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Pharmacy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
8
|
Colvee-Martin H, Parra JR, Gonzalez GA, Barker W, Duara R. Neuropathology, Neuroimaging, and Fluid Biomarkers in Alzheimer's Disease. Diagnostics (Basel) 2024; 14:704. [PMID: 38611617 PMCID: PMC11012058 DOI: 10.3390/diagnostics14070704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/17/2024] [Indexed: 04/14/2024] Open
Abstract
An improved understanding of the pathobiology of Alzheimer's disease (AD) should lead ultimately to an earlier and more accurate diagnosis of AD, providing the opportunity to intervene earlier in the disease process and to improve outcomes. The known hallmarks of Alzheimer's disease include amyloid-β plaques and neurofibrillary tau tangles. It is now clear that an imbalance between production and clearance of the amyloid beta protein and related Aβ peptides, especially Aβ42, is a very early, initiating factor in Alzheimer's disease (AD) pathogenesis, leading to aggregates of hyperphosphorylation and misfolded tau protein, inflammation, and neurodegeneration. In this article, we review how the AD diagnostic process has been transformed in recent decades by our ability to measure these various elements of the pathological cascade through the use of imaging and fluid biomarkers. The more recently developed plasma biomarkers, especially phosphorylated-tau217 (p-tau217), have utility for screening and diagnosis of the earliest stages of AD. These biomarkers can also be used to measure target engagement by disease-modifying therapies and the response to treatment.
Collapse
Affiliation(s)
- Helena Colvee-Martin
- Wien Center for Alzheimer’s Disease & Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (H.C.-M.); (W.B.)
| | - Juan Rayo Parra
- Human & Molecular Genetics, Florida International University, Miami, FL 33199, USA; (J.R.P.); (G.A.G.)
| | - Gabriel Antonio Gonzalez
- Human & Molecular Genetics, Florida International University, Miami, FL 33199, USA; (J.R.P.); (G.A.G.)
| | - Warren Barker
- Wien Center for Alzheimer’s Disease & Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (H.C.-M.); (W.B.)
| | - Ranjan Duara
- Wien Center for Alzheimer’s Disease & Memory Disorders, Mount Sinai Medical Center, Miami Beach, FL 33140, USA; (H.C.-M.); (W.B.)
| |
Collapse
|
9
|
Oliveira Monteiro E Pereira de Almeida MP, Valle Pedroso R, Mantellatto Grigoli M, Vicente Silva T, Manzine PR, Cominetti MR. ADAM10 as a biomarker for Alzheimer's disease: A systematic review. Rev Neurol (Paris) 2024; 180:1-11. [PMID: 37460331 DOI: 10.1016/j.neurol.2023.04.002] [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: 02/23/2022] [Revised: 01/04/2023] [Accepted: 04/18/2023] [Indexed: 02/06/2024]
Abstract
BACKGROUND Studies have shown that A Disintegrin and Metalloproteinase 10 (ADAM10) is the main α-secretase in the non-amyloidogenic cleavage of the amyloid precursor protein (APP), avoiding the production of amyloid-β peptide (Aβ), one of the pathological hallmarks of Alzheimer's disease (AD). OBJECTIVE To investigate ADAM10 from cerebrospinal fluid (CSF) and plasma/serum as a potential biomarker for AD. METHODS A systematic review was carried out in the MEDLINE/PubMed, Web of Science, Embase, and Scopus databases using the terms and Boolean operators: "Alzheimer" AND "ADAM10" AND "biomarker". Citation searching was also adopted. The inclusion criteria were original studies of ADAM10 in blood or CSF in patients with AD. The risk of bias was assessed using the Quality Assessment Tool for Observational Cohort and Cross-Sectional Studies. The analysis methods were registered in the PROSPERO database (#CRD42021274239). RESULTS Of the 97 records screened, 17 were included. There is strong evidence for lower levels of ADAM10 in platelets of persons with AD compared to cognitively healthy participants. On the other hand, higher levels of ADAM10 were found in plasma. Regarding CSF, controversial results were found with lower and higher levels of ADAM10 in persons with AD compared to healthy older adults. The differences may be due to diverse reasons, including different sample collection and processing and different antibodies, highlighting the importance of standardizing the experiments and choosing the appropriate antibodies for ADAM10 detection. CONCLUSION Evidence shows that ADAM10 levels are altered in platelets, plasma, serum, and CSF of individuals with AD. The alteration was evident in all stages of the disease, and therefore, the protein may represent a complementary biomarker for the disease. However, more studies must be performed to establish cut-off values for ADAM10 levels to discriminate AD participants from cognitively unimpaired older adults.
Collapse
Affiliation(s)
| | - R Valle Pedroso
- Department of Gerontology, Federal University of São Carlos. Rod. Washington Luis, km 235, 13565-905 São Carlos, SP, Brazil
| | - M Mantellatto Grigoli
- Department of Gerontology, Federal University of São Carlos. Rod. Washington Luis, km 235, 13565-905 São Carlos, SP, Brazil
| | - T Vicente Silva
- Department of Gerontology, Federal University of São Carlos. Rod. Washington Luis, km 235, 13565-905 São Carlos, SP, Brazil
| | - P R Manzine
- Department of Gerontology, Federal University of São Carlos. Rod. Washington Luis, km 235, 13565-905 São Carlos, SP, Brazil
| | - M R Cominetti
- Department of Gerontology, Federal University of São Carlos. Rod. Washington Luis, km 235, 13565-905 São Carlos, SP, Brazil; Trinity College Dublin, Dublin, Ireland; Global Brain Health Institute, Dublin, Ireland.
| |
Collapse
|
10
|
Gao Y, Yu H, Liu Y, Xu Z, He B, Liu H, Wang Y, Zhang Y, Liang Y, Yang Y, Zheng J, Wang J. GSK-3β activation mediates apolipoprotein E4-associated cognitive impairment in type 2 diabetes mellitus: A multicenter, cross-sectional study. J Diabetes 2024; 16:e13470. [PMID: 37700547 PMCID: PMC10809305 DOI: 10.1111/1753-0407.13470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/17/2023] [Accepted: 08/16/2023] [Indexed: 09/14/2023] Open
Abstract
AIM Both the activation of glycogen synthase kinase-3β (GSK-3β) and the presence of ApoE ε4 genotype have been found to respectively correlate with cognitive decline in patients with type 2 diabetes mellitus (T2DM), who further show a high incidence of developing Alzheimer's disease. However, the relationship between ApoE ε4 and GSK-3β in the cognitive impairment of T2DM patients remains unclear. METHODS ApoE genotypes and platelet GSK-3β level were measured in 1139 T2DM patients recruited from five medical centers in Wuhan, China. Cognitive functions were assessed by Mini-Mental State Examination (MMSE). The association and the relationships among apolipoprotein E (ApoE) genotypes, GSK-3β activity and cognitive function were analyzed by regression and mediating effect analyses, respectively. RESULTS T2DM patients with ApoE ε4 but not ApoE ε2 haplotype showed poorer cognitive function and elevated platelet GSK-3β activity, when using ApoE ε3 as reference. The elevation of GSK-3β activity was positively correlated the diabetes duration, as well as plasma glycated hemoglobin (HbA1c) and glucose levels. Moreover, correlation and regression analysis also revealed significant pairwise correlations among GSK-3β activity, ApoE gene polymorphism and cognitive function. Lastly, using Baron and Kenny modeling, we unveiled a mediative role of GSK-3β activity between ApoE ε4 and cognitive impairment. CONCLUSION We reported here that the upregulation of GSK-3β activity mediates the exacerbation of cognitive impairment by ApoE ε4-enhanced cognitive impairment in T2DM patients, suggesting GSK-3β inhibitors as promising drugs for preserving cognitive function in T2DM patients, especially to those with ApoE ε4 genotype.
Collapse
Affiliation(s)
- Yang Gao
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Department of RadiologyWuhan Brain HospitalWuhanChina
| | - Haitao Yu
- Department of Fundamental Medicine, Wuxi School of MedicineJiangnan UniversityWuxiChina
| | - Yanchao Liu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Zhipeng Xu
- Department of NeurologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Benrong He
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Honghai Liu
- School of Medicine and Health Management, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuying Wang
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yao Zhang
- Li‐Yuan Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yi Liang
- Department of RadiologyWuhan Brain HospitalWuhanChina
| | - Ying Yang
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical SciencesPeking University; Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking UniversityBeijingChina
| | - Jian‐Zhi Wang
- Department of Pathophysiology, School of Basic Medicine, Ministry of Education Key Laboratory for Neurological Disorders, Hubei Key Laboratory for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Co‐innovation Center of NeuroregenerationNantong UniversityNantongChina
| |
Collapse
|
11
|
Grari O, Elmoujtahide D, Sebbar E, Choukri M. The Biochemistry Behind Cognitive Decline: Biomarkers of Alzheimer's Disease. EJIFCC 2023; 34:276-283. [PMID: 38303754 PMCID: PMC10828533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent type of dementia. Pathologically, the disease is marked by neurofibrillary tangles (NFT), which are aberrant accumulations of the tau protein that develop inside neurons, and extracellular plaque deposits of the amyloid β peptide (Aβ). These pathological lesions are present in the brain before the beginning of clinical manifestations. However, despite advancements in the comprehension of AD pathophysiology, timely and accurate clinical diagnosis remains challenging. Therefore, developing biomarkers capable of detecting AD during the preclinical phase holds enormous promise for precise diagnosis since detecting the disease early is crucial because it enables interventions when treatments may be more effective. This article intends to provide a comprehensive review of AD biomarkers, discussing their significance, classification, and recent developments in the field.
Collapse
Affiliation(s)
- O. Grari
- : Faculty of Medicine and Pharmacy, Mohammed I University, Oujda, Morocco
- : Department of Biochemistry, Mohammed VI University Hospital, Oujda, Morocco
| | - D. Elmoujtahide
- : Faculty of Medicine and Pharmacy, Mohammed I University, Oujda, Morocco
- : Department of Biochemistry, Mohammed VI University Hospital, Oujda, Morocco
| | - E. Sebbar
- : Faculty of Medicine and Pharmacy, Mohammed I University, Oujda, Morocco
- : Department of Biochemistry, Mohammed VI University Hospital, Oujda, Morocco
| | - M. Choukri
- : Faculty of Medicine and Pharmacy, Mohammed I University, Oujda, Morocco
- : Department of Biochemistry, Mohammed VI University Hospital, Oujda, Morocco
| |
Collapse
|
12
|
Beura SK, Dhapola R, Panigrahi AR, Yadav P, Kumar R, Reddy DH, Singh SK. Antiplatelet drugs: Potential therapeutic options for the management of neurodegenerative diseases. Med Res Rev 2023; 43:1835-1877. [PMID: 37132460 DOI: 10.1002/med.21965] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/13/2023] [Accepted: 04/12/2023] [Indexed: 05/04/2023]
Abstract
The blood platelet plays an important role but often remains under-recognized in several vascular complications and associated diseases. Surprisingly, platelet hyperactivity and hyperaggregability have often been considered the critical risk factors for developing vascular dysfunctions in several neurodegenerative diseases (NDDs) like Alzheimer's disease, Parkinson's disease, Huntington's disease, and multiple sclerosis. In addition, platelet structural and functional impairments promote prothrombotic and proinflammatory environment that can aggravate the progression of several NDDs. These findings provide the rationale for using antiplatelet agents not only to prevent morbidity but also to reduce mortality caused by NDDs. Therefore, we thoroughly review the evidence supporting the potential pleiotropic effects of several novel classes of synthetic antiplatelet drugs, that is, cyclooxygenase inhibitors, adenosine diphosphate receptor antagonists, protease-activated receptor blockers, and glycoprotein IIb/IIIa receptor inhibitors in NDDs. Apart from this, the review also emphasizes the recent developments of selected natural antiplatelet phytochemicals belonging to key classes of plant-based bioactive compounds, including polyphenols, alkaloids, terpenoids, and flavonoids as potential therapeutic candidates in NDDs. We believe that the broad analysis of contemporary strategies and specific approaches for plausible therapeutic treatment for NDDs presented in this review could be helpful for further successful research in this area.
Collapse
Affiliation(s)
- Samir K Beura
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Rishika Dhapola
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Abhishek R Panigrahi
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Pooja Yadav
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Reetesh Kumar
- Department of Agricultural Sciences, Institute of Applied Sciences and Humanities, GLA University, Mathura, Uttar Pradesh, India
| | - Dibbanti H Reddy
- Department of Pharmacology, School of Health Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| | - Sunil K Singh
- Department of Zoology, School of Biological Sciences, Central University of Punjab, Ghudda, Bathinda, Punjab, India
| |
Collapse
|
13
|
Pais MV, Forlenza OV, Diniz BS. Plasma Biomarkers of Alzheimer's Disease: A Review of Available Assays, Recent Developments, and Implications for Clinical Practice. J Alzheimers Dis Rep 2023; 7:355-380. [PMID: 37220625 PMCID: PMC10200198 DOI: 10.3233/adr-230029] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 05/25/2023] Open
Abstract
Recently, low-sensitive plasma assays have been replaced by new ultra-sensitive assays such as single molecule enzyme-linked immunosorbent assay (Simoa), the Mesoscale Discovery (MSD) platform, and immunoprecipitation-mass spectrometry (IP-MS) with higher accuracy in the determination of plasma biomarkers of Alzheimer's disease (AD). Despite the significant variability, many studies have established in-house cut-off values for the most promising available biomarkers. We first reviewed the most used laboratory methods and assays to measure plasma AD biomarkers. Next, we review studies focused on the diagnostic performance of these biomarkers to identify AD cases, predict cognitive decline in pre-clinical AD cases, and differentiate AD cases from other dementia. We summarized data from studies published until January 2023. A combination of plasma Aβ42/40 ratio, age, and APOE status showed the best accuracy in diagnosing brain amyloidosis with a liquid chromatography-mass spectrometry (LC-MS) assay. Plasma p-tau217 has shown the best accuracy in distinguishing Aβ-PET+ from Aβ-PET-even in cognitively unimpaired individuals. We also summarized the different cut-off values for each biomarker when available. Recently developed assays for plasma biomarkers have undeniable importance in AD research, with improved analytical and diagnostic performance. Some biomarkers have been extensively used in clinical trials and are now clinically available. Nonetheless, several challenges remain to their widespread use in clinical practice.
Collapse
Affiliation(s)
- Marcos V. Pais
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo, SP, Brazil
| | - Orestes V. Forlenza
- Laboratory of Neuroscience (LIM-27), Departamento e Instituto de Psiquiatria, Faculdade de Medicina, Universidade de Sao Paulo (FMUSP), Sao Paulo, SP, Brazil
| | - Breno S. Diniz
- UConn Center on Aging, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
14
|
Yu H, Li M, Pan Q, Liu Y, Zhang Y, He T, Yang H, Xiao Y, Weng Y, Gao Y, Ke D, Chai G, Wang J. Integrated analyses of brain and platelet omics reveal their common altered and driven molecules in Alzheimer's disease. MedComm (Beijing) 2022; 3:e180. [PMID: 36254251 PMCID: PMC9560744 DOI: 10.1002/mco2.180] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/08/2022] Open
Abstract
Platelets may serve as a perfect peripheral source for exploring diagnostic biomarkers for Alzheimer's disease (AD); however, the molecular linkage between platelet and the brain is missing. To find the common altered and driving molecules in both brain and the platelet, we performed an integrated analysis of our platelet omics and brain omics reported in the literature, and analyzed their correlations with AD-specific pathology and cognitive impairment. By integrating the gene and protein expression profiles from 269 AD patients, we deduced 239 differentially expressed proteins (DEPs) appeared in both brain and the platelet, and 70.3% of them had consistent changes. Further analysis demonstrated that the altered brain and peripheral regulations were pinpointed into 10 imbalanced pathways. We also found that 117 DEPs, including ADAM10, were closely associated to the AD-specific β-amyloid and tau pathologies; and the changes of IDH3B and RTN1 had a potential diagnostic value for cognitive impairment analyzed by machine learning. Finally, we identified that HMOX2 and SERPINA3 could serve as driving molecules in neurodegeneration, and they were increased and decreased in AD patients, respectively. Together, this integrated brain and platelet omics provides a valuable resource for establishing efficient peripheral diagnostic biomarkers and potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Haitao Yu
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry of China/Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Basic MedicineWuxi School of MedicineJiangnan UniversityWuxiJiangsuChina
| | - Mengzhu Li
- Department of NeurosurgeryWuhan Central Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Qihang Pan
- Department of NeurosurgeryWuhan Central Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yanchao Liu
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry of China/Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of NeurosurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yao Zhang
- Department of EndocrinologyLiyuan HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ting He
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry of China/Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Huisheng Yang
- Institute of Acupuncture and MoxibustionChina Academy of Chinese Medical SciencesBeijingChina
| | - Yue Xiao
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry of China/Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ying Weng
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry of China/Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yang Gao
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry of China/Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Dan Ke
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry of China/Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Gaoshang Chai
- Department of Basic MedicineWuxi School of MedicineJiangnan UniversityWuxiJiangsuChina
| | - Jian‐Zhi Wang
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry of China/Hubei Province for Neurological DisordersTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantongChina
| |
Collapse
|
15
|
Platelets’ Nanomechanics and Morphology in Neurodegenerative Pathologies. Biomedicines 2022; 10:biomedicines10092239. [PMID: 36140340 PMCID: PMC9496241 DOI: 10.3390/biomedicines10092239] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
The imaging and force–distance curve modes of atomic force microscopy (AFM) are explored to compare the morphological and mechanical signatures of platelets from patients diagnosed with classical neurodegenerative diseases (NDDs) and healthy individuals. Our data demonstrate the potential of AFM to distinguish between the three NDDs—Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS) and Alzheimer’s disease (AD), and normal healthy platelets. The common features of platelets in the three pathologies are reduced membrane surface roughness, area and height, and enhanced nanomechanics in comparison with healthy cells. These changes might be related to general phenomena associated with reorganization in the platelet membrane morphology and cytoskeleton, a key factor for all platelets’ functions. Importantly, the platelets’ signatures are modified to a different extent in the three pathologies, most significant in ALS, less pronounced in PD and the least in AD platelets, which shows the specificity associated with each pathology. Moreover, different degree of activation, distinct pseudopodia and nanocluster formation characterize ALS, PD and AD platelets. The strongest alterations in the biophysical properties correlate with the highest activation of ALS platelets, which reflect the most significant changes in their nanoarchitecture. The specific platelet signatures that mark each of the studied pathologies can be added as novel biomarkers to the currently used diagnostic tools.
Collapse
|
16
|
The multifaceted role of platelets in mediating brain function. Blood 2022; 140:815-827. [PMID: 35609283 PMCID: PMC9412009 DOI: 10.1182/blood.2022015970] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Platelets, the small, anucleate blood cells that originate from megakaryocytes in the bone marrow, are typically associated with coagulation. However, it is now apparent that platelets are more multifaceted than originally thought, with their function extending beyond their traditional role in hemostasis to acting as important mediators of brain function. In this review, we outline the broad repertoire of platelet function in the central nervous system, focusing on the similarities between platelets and neurons. We also summarize the role that platelets play in the pathophysiology of various neurological diseases, with a particular focus on neuroinflammation and neurodegeneration. Finally, we highlight the exciting prospect of harnessing the unique features of the platelet proteome and extracellular vesicles, which are rich in neurotrophic, antioxidative, and antiinflammatory factors, for the development of novel neuroprotective and neuroregenerative interventions to treat various neurodegenerative and traumatic pathologies.
Collapse
|
17
|
Ramos‐Cejudo J, Johnson AD, Beiser A, Seshadri S, Salinas J, Berger JS, Fillmore NR, Do N, Zheng C, Kovbasyuk Z, Ardekani BA, Pomara N, Bubu OM, Parekh A, Convit A, Betensky RA, Wisniewski TM, Osorio RS. Platelet Function Is Associated With Dementia Risk in the Framingham Heart Study. J Am Heart Assoc 2022; 11:e023918. [PMID: 35470685 PMCID: PMC9238609 DOI: 10.1161/jaha.121.023918] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 02/01/2022] [Indexed: 12/15/2022]
Abstract
Background Vascular function is compromised in Alzheimer disease (AD) years before amyloid and tau pathology are detected and a substantial body of work shows abnormal platelet activation states in patients with AD. The aim of our study was to investigate whether platelet function in middle age is independently associated with future risk of AD. Methods and Results We examined associations of baseline platelet function with incident dementia risk in the community-based FHS (Framingham Heart Study) longitudinal cohorts. The association between platelet function and risk of dementia was evaluated using the cumulative incidence function and inverse probability weighted Cox proportional cause-specific hazards regression models, with adjustment for demographic and clinical covariates. Platelet aggregation response was measured by light transmission aggregometry. The final study sample included 1847 FHS participants (average age, 53.0 years; 57.5% women). During follow-up (median, 20.5 years), we observed 154 cases of incident dementia, of which 121 were AD cases. Results from weighted models indicated that platelet aggregation response to adenosine diphosphate 1.0 µmol/L was independently and positively associated with dementia risk, and it was preceded in importance only by age and hypertension. Sensitivity analyses showed associations with the same directionality for participants defined as adenosine diphosphate hyper-responders, as well as the platelet response to 0.1 µmol/L epinephrine. Conclusions Our study shows individuals free of antiplatelet therapy with a higher platelet response are at higher risk of dementia in late life during a 20-year follow-up, reinforcing the role of platelet function in AD risk. This suggests that platelet phenotypes may be associated with the rate of dementia and potentially have prognostic value.
Collapse
Affiliation(s)
- Jaime Ramos‐Cejudo
- Department of PsychiatryNew York University (NYU) Grossman School of MedicineNew YorkNY
- VA Boston Cooperative Studies ProgramMAVERICVA Boston Healthcare SystemBostonMA
| | - Andrew D. Johnson
- Population Sciences BranchDivision of Intramural ResearchNational Heart, Lung, and Blood InstituteFraminghamMA
- The Framingham StudyBostonMA
| | - Alexa Beiser
- The Framingham StudyBostonMA
- Department of BiostatisticsBoston University School of Public HealthBostonMA
- Department of NeurologyBoston University School of MedicineBostonMA
| | - Sudha Seshadri
- The Framingham StudyBostonMA
- Department of NeurologyBoston University School of MedicineBostonMA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative DiseasesUniversity of Texas Health Sciences CenterSan AntonioTX
| | - Joel Salinas
- The Framingham StudyBostonMA
- Department of NeurologyCenter for Cognitive NeurologyNYU Grossman School of MedicineNew YorkNY
| | - Jeffrey S. Berger
- Division of Vascular SurgeryDepartment of SurgeryNYU Grossman School of MedicineNew YorkNY
- Divisions of Cardiology and HematologyDepartment MedicineNYU Grossman School of MedicineNew YorkNY
- Center for the Prevention of Cardiovascular DiseaseNYU Grossman School of MedicineNew YorkNY
| | - Nathanael R. Fillmore
- VA Boston Cooperative Studies ProgramMAVERICVA Boston Healthcare SystemBostonMA
- Harvard Medical SchoolBostonMA
| | - Nhan Do
- VA Boston Cooperative Studies ProgramMAVERICVA Boston Healthcare SystemBostonMA
- Boston University School of MedicineBostonMA
| | - Chunlei Zheng
- VA Boston Cooperative Studies ProgramMAVERICVA Boston Healthcare SystemBostonMA
- Boston University School of MedicineBostonMA
| | - Zanetta Kovbasyuk
- Department of PsychiatryNew York University (NYU) Grossman School of MedicineNew YorkNY
| | - Babak A. Ardekani
- Department of PsychiatryNew York University (NYU) Grossman School of MedicineNew YorkNY
- Nathan Kline InstituteOrangeburgNY
| | - Nunzio Pomara
- Department of PsychiatryNew York University (NYU) Grossman School of MedicineNew YorkNY
- Nathan Kline InstituteOrangeburgNY
| | - Omonigho M. Bubu
- Department of PsychiatryNew York University (NYU) Grossman School of MedicineNew YorkNY
| | - Ankit Parekh
- Division of PulmonaryCritical Care, and Sleep MedicineIcahn School of Medicine at Mount SinaiNew YorkNY
| | - Antonio Convit
- Department of PsychiatryNew York University (NYU) Grossman School of MedicineNew YorkNY
- Nathan Kline InstituteOrangeburgNY
| | - Rebecca A. Betensky
- Department of BiostatisticsNew York University School of Global Public HealthNew YorkNY
| | - Thomas M. Wisniewski
- Department of PsychiatryNew York University (NYU) Grossman School of MedicineNew YorkNY
- Department of NeurologyCenter for Cognitive NeurologyNYU Grossman School of MedicineNew YorkNY
- Department of PathologyNYU Grossman School of MedicineNew YorkNY
| | - Ricardo S. Osorio
- Department of PsychiatryNew York University (NYU) Grossman School of MedicineNew YorkNY
- Nathan Kline InstituteOrangeburgNY
| |
Collapse
|
18
|
Li S, An N, Chen N, Wang Y, Yang L, Wang Y, Yao Z, Hu B. The impact of Alzheimer's disease susceptibility loci on lateral ventricular surface morphology in older adults. Brain Struct Funct 2022; 227:913-924. [PMID: 35028746 DOI: 10.1007/s00429-021-02429-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: 01/22/2021] [Accepted: 11/13/2021] [Indexed: 11/25/2022]
Abstract
The enlargement of ventricular volume is a general trend in the elderly, especially in patients with Alzheimer's disease (AD). Multiple susceptibility loci have been reported to have an increased risk for AD and the morphology of brain structures are affected by the variations in the risk loci. Therefore, we hypothesized that genes contributed significantly to the ventricular surface, and the changes of ventricular surface were associated with the impairment of cognitive functions. After the quality controls (QC) and genotyping, a lateral ventricular segmentation method was employed to obtain the surface features of lateral ventricle. We evaluated the influence of 18 selected AD susceptibility loci on both volume and surface morphology across 410 subjects from Alzheimer's Disease Neuroimaging Initiative (ADNI). Correlations were conducted between radial distance (RD) and Montreal Cognitive Assessment (MoCA) subscales. Only the C allele at the rs744373 loci in BIN1 gene significantly accelerated the atrophy of lateral ventricle, including the anterior horn, body, and temporal horn of left lateral ventricle. No significant effect on lateral ventricle was found at other loci. Our results revealed that most regions of the bilateral ventricular surface were significantly negatively correlated with cognitive scores, particularly in delayed recall. Besides, small areas of surface were negatively correlated with language, orientation, and visuospatial scores. Together, our results indicated that the genetic variation affected the localized areas of lateral ventricular surface, and supported that lateral ventricle was an important brain structure associated with cognition in the elderly.
Collapse
Affiliation(s)
- Shan Li
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China
| | - Na An
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China
| | - Nan Chen
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China
| | - Yin Wang
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China
| | - Lin Yang
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China
| | - Yalin Wang
- School of Computing, Informatics, and Decision Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Zhijun Yao
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China.
| | - Bin Hu
- Gansu Provincial Key Laboratory of Wearable Computing, School of Information Science and Engineering, Lanzhou University, No. 222 South Tianshui Road, Lanzhou, 730000, Gansu Province, People's Republic of China.
- CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, ShangHai, China.
- Joint Research Center for Cognitive Neurosensor Technology of Lanzhou University and Institute of Semiconductors, Chinese Academy of Sciences, LanZhou, China.
- Engineering Research Center of Open Source Software and Real-Time System, Ministry of Education, Lanzhou University, Lanzhou, China.
| |
Collapse
|
19
|
Zago E, Dal Molin A, Dimitri GM, Xumerle L, Pirazzini C, Bacalini MG, Maturo MG, Azevedo T, Spasov S, Gómez-Garre P, Periñán MT, Jesús S, Baldelli L, Sambati L, Calandra-Buonaura G, Garagnani P, Provini F, Cortelli P, Mir P, Trenkwalder C, Mollenhauer B, Franceschi C, Liò P, Nardini C. Early downregulation of hsa-miR-144-3p in serum from drug-naïve Parkinson's disease patients. Sci Rep 2022; 12:1330. [PMID: 35079043 PMCID: PMC8789812 DOI: 10.1038/s41598-022-05227-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/28/2021] [Indexed: 12/18/2022] Open
Abstract
Advanced age represents one of the major risk factors for Parkinson's Disease. Recent biomedical studies posit a role for microRNAs, also known to be remodelled during ageing. However, the relationship between microRNA remodelling and ageing in Parkinson's Disease, has not been fully elucidated. Therefore, the aim of the present study is to unravel the relevance of microRNAs as biomarkers of Parkinson's Disease within the ageing framework. We employed Next Generation Sequencing to profile serum microRNAs from samples informative for Parkinson's Disease (recently diagnosed, drug-naïve) and healthy ageing (centenarians) plus healthy controls, age-matched with Parkinson's Disease patients. Potential microRNA candidates markers, emerging from the combination of differential expression and network analyses, were further validated in an independent cohort including both drug-naïve and advanced Parkinson's Disease patients, and healthy siblings of Parkinson's Disease patients at higher genetic risk for developing the disease. While we did not find evidences of microRNAs co-regulated in Parkinson's Disease and ageing, we report that hsa-miR-144-3p is consistently down-regulated in early Parkinson's Disease patients. Moreover, interestingly, functional analysis revealed that hsa-miR-144-3p is involved in the regulation of coagulation, a process known to be altered in Parkinson's Disease. Our results consistently show the down-regulation of hsa-mir144-3p in early Parkinson's Disease, robustly confirmed across a variety of analytical and experimental analyses. These promising results ask for further research to unveil the functional details of the involvement of hsa-mir144-3p in Parkinson's Disease.
Collapse
Affiliation(s)
| | | | - Giovanna Maria Dimitri
- Computer Laboratory, Department of Computer Science and Technology, University of Cambridge, Cambridge, UK
| | | | - Chiara Pirazzini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | | | - Maria Giovanna Maturo
- Personal Genomics S.R.L., Verona, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Tiago Azevedo
- Computer Laboratory, Department of Computer Science and Technology, University of Cambridge, Cambridge, UK
| | - Simeon Spasov
- Computer Laboratory, Department of Computer Science and Technology, University of Cambridge, Cambridge, UK
| | - Pilar Gómez-Garre
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - María Teresa Periñán
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Silvia Jesús
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Luca Baldelli
- Department of Biomedical and NeuroMotor Sciences (DiBiNeM), University of Bologna, Bologna, Italy
| | - Luisa Sambati
- Department of Biomedical and NeuroMotor Sciences (DiBiNeM), University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Giovanna Calandra-Buonaura
- Department of Biomedical and NeuroMotor Sciences (DiBiNeM), University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Paolo Garagnani
- Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden
- Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy
| | - Federica Provini
- Department of Biomedical and NeuroMotor Sciences (DiBiNeM), University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Pietro Cortelli
- Department of Biomedical and NeuroMotor Sciences (DiBiNeM), University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Pablo Mir
- Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kliniktstrasse 16, 34128, Kassel, Germany
- Department of Neurosurgery, University Medical Center Göttingen, Göttingen, Germany
| | - Brit Mollenhauer
- Paracelsus-Elena-Klinik, Kliniktstrasse 16, 34128, Kassel, Germany
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Claudio Franceschi
- Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhny Novgorod, Russia.
| | - Pietro Liò
- Computer Laboratory, Department of Computer Science and Technology, University of Cambridge, Cambridge, UK
| | - Christine Nardini
- Personal Genomics S.R.L., Verona, Italy.
- Consiglio Nazionale delle Ricerche, Istituto per le Applicazioni del Calcolo "Mauro Picone", 00185, Rome, Italy.
| |
Collapse
|
20
|
Fang Y, Doyle MF, Alosco ML, Mez J, Satizabal CL, Qiu WQ, Lunetta KL, Murabito JM. Cross-Sectional Association Between Blood Cell Phenotypes, Cognitive Function, and Brain Imaging Measures in the Community-Based Framingham Heart Study. J Alzheimers Dis 2022; 87:1291-1305. [PMID: 35431244 PMCID: PMC9969805 DOI: 10.3233/jad-215533] [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 Peripheral inflammation is associated with increased risk for dementia. Neutrophil to lymphocyte ratio (NLR), red cell distribution width (RDW), and mean platelet volume (MPV), are easily measured circulating blood cell phenotypes reflecting chronic peripheral inflammation, but their association with dementia status is unclear. OBJECTIVE We sought to investigate the cross-sectional association of these inflammatory measures with neuropsychological (NP) test performance, and brain magnetic resonance imaging (MRI) measures in the Framingham Heart Study (FHS) Offspring, Third-generation, and Omni cohorts. METHODS We identified FHS participants who attended an exam that included a complete blood cell count (CBC) and underwent NP testing (n = 3,396) or brain MRI (n = 2,770) within five years of blood draw. We investigated the association between NLR, RDW, and MPV and NP test performance and structural MRI-derived volumetric measurements using linear mixed effect models accounting for family relationships and adjusting for potential confounders. RESULTS Participants were on average 60 years old, 53% female, and about 80% attended some college. Higher NLR was significantly associated with poorer performance on visual memory, and visuospatial abilities, as well as with larger white matter hyperintensity volume. We also observed associations for higher RDW with poorer executive function and smaller total cerebral brain volume. CONCLUSION Chronic peripheral inflammation as measured by NLR and RDW was associated with worse cognitive function, reduced brain volume, and greater microvascular disease in FHS participants. If confirmed in other samples, CBC may provide informative and cost-effective biomarkers of abnormal brain aging in the community.
Collapse
Affiliation(s)
- Yuan Fang
- Boston University School of Public Health, Department of Biostatistics, Boston, MA, USA
| | - Margaret F. Doyle
- University of Vermont, Larner College of Medicine, Department of Pathology and Laboratory Medicine, Burlington, VT
| | - Michael L. Alosco
- Boston University School of Medicine, Boston University Alzheimer’s Disease Research Center and CTE Center, Boston, MA, USA.,Boston University School of Medicine, Department of Neurology, Boston, MA, USA
| | - Jesse Mez
- Boston University School of Medicine, Boston University Alzheimer’s Disease Research Center and CTE Center, Boston, MA, USA.,Boston University School of Medicine, Department of Neurology, Boston, MA, USA.,Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, MA, USA
| | - Claudia L. Satizabal
- Boston University School of Medicine, Department of Neurology, Boston, MA, USA.,University of Texas Health Science Center at San Antonio, Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, San Antonio, USA
| | - Wei Qiao Qiu
- Boston University School of Medicine, Boston University Alzheimer’s Disease Research Center and CTE Center, Boston, MA, USA.,Boston University School of Medicine, Department of Psychiatry, Boston, MA, USA.,Boston University School of Medicine, Department of Pharmacology & Experimental Therapeutics, Boston, MA, USA
| | - Kathryn L. Lunetta
- Boston University School of Public Health, Department of Biostatistics, Boston, MA, USA
| | - Joanne M. Murabito
- Framingham Heart Study, National Heart, Lung, and Blood Institute and Boston University School of Medicine, Framingham, MA, USA.,Boston University School of Medicine, Department of Medicine, Section of General Internal Medicine, Boston, MA, USA
| |
Collapse
|
21
|
Page MJ, Pretorius E. Platelet Behavior Contributes to Neuropathologies: A Focus on Alzheimer's and Parkinson's Disease. Semin Thromb Hemost 2021; 48:382-404. [PMID: 34624913 DOI: 10.1055/s-0041-1733960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The functions of platelets are broad. Platelets function in hemostasis and thrombosis, inflammation and immune responses, vascular regulation, and host defense against invading pathogens, among others. These actions are achieved through the release of a wide set of coagulative, vascular, inflammatory, and other factors as well as diverse cell surface receptors involved in the same activities. As active participants in these physiological processes, platelets become involved in signaling pathways and pathological reactions that contribute to diseases that are defined by inflammation (including by pathogen-derived stimuli), vascular dysfunction, and coagulation. These diseases include Alzheimer's and Parkinson's disease, the two most common neurodegenerative diseases. Despite their unique pathological and clinical features, significant shared pathological processes exist between these two conditions, particularly relating to a central inflammatory mechanism involving both neuroinflammation and inflammation in the systemic environment, but also neurovascular dysfunction and coagulopathy, processes which also share initiation factors and receptors. This triad of dysfunction-(neuro)inflammation, neurovascular dysfunction, and hypercoagulation-illustrates the important roles platelets play in neuropathology. Although some mechanisms are understudied in Alzheimer's and Parkinson's disease, a strong case can be made for the relevance of platelets in neurodegeneration-related processes.
Collapse
Affiliation(s)
- Martin J Page
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland, South Africa
| |
Collapse
|
22
|
Yu H, Liu Y, He T, Zhang Y, He J, Li M, Jiang B, Gao Y, Chen C, Ke D, Liu J, He B, Yang X, Wang J. Platelet biomarkers identifying mild cognitive impairment in type 2 diabetes patients. Aging Cell 2021; 20:e13469. [PMID: 34528736 PMCID: PMC8520722 DOI: 10.1111/acel.13469] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/09/2021] [Accepted: 07/21/2021] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is an independent risk factor of Alzheimer's disease (AD). Therefore, identifying periphery biomarkers correlated with mild cognitive impairment (MCI) is of importance for early diagnosis of AD. Here, we performed platelet proteomics in T2DM patients with MCI (T2DM-MCI) and without MCI (T2DM-nMCI). Pearson analysis of the omics data with MMSE (mini-mental state examination), Aβ1-42/Aβ1-40 (β-amyloid), and rGSK-3β(T/S9) (total to Serine-9-phosphorylated glycogen synthase kinase-3β) revealed that mitophagy/autophagy-, insulin signaling-, and glycolysis/gluconeogenesis pathways-related proteins were most significantly involved. Among them, only the increase of optineurin, an autophagy-related protein, was simultaneously correlated with the reduced MMSE score, and the increased Aβ1-42/Aβ1-40 and rGSK-3β(T/S9), and the optineurin alone could discriminate T2DM-MCI from T2DM-nMCI. Combination of the elevated platelet optineurin and rGSK-3β(T/S9) enhanced the MCI-discriminating efficiency with AUC of 0.927, specificity of 86.7%, sensitivity of 85.3%, and accuracy of 0.859, which is promising for predicting cognitive decline in T2DM patients.
Collapse
Affiliation(s)
- Haitao Yu
- Department of PathophysiologyKey Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
| | - Yanchao Liu
- Department of PathophysiologyKey Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of NeurosurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ting He
- Department of PathophysiologyKey Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yao Zhang
- Key Laboratory of Ministry of Education for Neurological DisordersLi Yuan HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jiahua He
- School of PhysicsHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Mengzhu Li
- Department of NeurosurgeryWuhan Central Hospital Affiliated to Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Bijun Jiang
- Department of PhysiologySchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Yang Gao
- Department of PathophysiologyKey Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chongyang Chen
- Department of PathophysiologyKey Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
| | - Dan Ke
- Department of PathophysiologyKey Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jianjun Liu
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
| | - Benrong He
- Department of PathophysiologyKey Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
| | - Jian‐Zhi Wang
- Department of PathophysiologyKey Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Co‐innovation Center of NeuroregenerationNantong UniversityNantongChina
| |
Collapse
|
23
|
Babić Leko M, Nikolac Perković M, Nedić Erjavec G, Klepac N, Švob Štrac DK, Borovečki F, Pivac N, Hof PR, Šimić G. Association of the MAOB rs1799836 Single Nucleotide Polymorphism and APOE ɛ4 Allele in Alzheimer's Disease. Curr Alzheimer Res 2021; 18:585-594. [PMID: 34533445 DOI: 10.2174/1567205018666210917162843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/01/2021] [Accepted: 08/22/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The dopaminergic system is functionally compromised in Alzheimer's dis-ease (AD). The activity of monoamine oxidase B (MAOB), the enzyme involved in the degradation of dopamine, is increased during AD. Also, increased expression of MAOB occurs in the post-mortem hippocampus and neocortex of patients with AD. The MAOB rs1799836 polymorphism modulates MAOB transcription, consequently influencing protein translation and MAOB activity. We recently showed that cerebrospinal fluid levels of amyloid β1-42 are decreased in patients carry- ing the A allele in MAOB rs1799836 polymorphism. OBJECTIVE The present study compares MAOB rs1799836 polymorphism and APOE, the only con- firmed genetic risk factor for sporadic AD. METHOD We included 253 participants, 127 of whom had AD, 57 had mild cognitive impairment, 11 were healthy controls, and 58 suffered from other primary causes of dementia. MAOB and APOE polymorphisms were determined using TaqMan SNP Genotyping Assays. RESULTS We observed that the frequency of APOE ɛ4/ɛ4 homozygotes and APOE ɛ4 carriers is sig- nificantly increased among patients carrying the AA MAOB rs1799836 genotype. CONCLUSION These results indicate that the MAOB rs1799836 polymorphism is a potential genetic biomarker of AD and a potential target for the treatment of decreased dopaminergic transmission and cognitive deterioration in AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | | | | | - Nataša Klepac
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | | | - Fran Borovečki
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Nela Pivac
- Department of Molecular Medicine, Institute Ruđer Bošković, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| |
Collapse
|
24
|
Uzbekov MG. Monoamine Oxidase as a Potential Biomarker of the Efficacy of Treatment of Mental Disorders. BIOCHEMISTRY (MOSCOW) 2021; 86:773-783. [PMID: 34225599 DOI: 10.1134/s0006297921060146] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The review summarizes the results of our own studies and published data on the biological markers of psychiatric disorders, with special emphasis on the activity of platelet monoamine oxidase. Pharmacotherapy studies in patients with the mixed anxiety-depressive disorder and first episode of schizophrenia have shown that the activity of platelet monoamine oxidase could serve as a potential biomarker of the efficacy of therapeutic interventions in these diseases.
Collapse
Affiliation(s)
- Marat G Uzbekov
- Moscow Research Institute of Psychiatry, Branch of Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, 107076, Russia.
| |
Collapse
|
25
|
Kopeikina E, Ponomarev ED. The Role of Platelets in the Stimulation of Neuronal Synaptic Plasticity, Electric Activity, and Oxidative Phosphorylation: Possibilities for New Therapy of Neurodegenerative Diseases. Front Cell Neurosci 2021; 15:680126. [PMID: 34335186 PMCID: PMC8318360 DOI: 10.3389/fncel.2021.680126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/23/2021] [Indexed: 12/04/2022] Open
Abstract
The central nervous system (CNS) is highly vascularized where neuronal cells are located in proximity to endothelial cells, astroglial limitans, and neuronal processes constituting integrated neurovascular units. In contrast to many other organs, the CNS has a blood-brain barrier (BBB), which becomes compromised due to infection, neuroinflammation, neurodegeneration, traumatic brain injury, and other reasons. BBB disruption is presumably involved in neuronal injury during epilepsy and psychiatric disorders. Therefore, many types of neuropsychological disorders are accompanied by an increase in BBB permeability leading to direct contact of circulating blood cells in the capillaries with neuronal cells in the CNS. The second most abundant type of blood cells are platelets, which come after erythrocytes and outnumber ~100-fold circulating leukocytes. When BBB becomes compromised, platelets swiftly respond to the vascular injury and become engaged in thrombosis and hemostasis. However, more recent studies demonstrated that platelets could also enter CNS parenchyma and directly interact with neuronal cells. Within CNS, platelets become activated by recognizing major brain gangliosides on the surface of astrocytes and neurons and releasing a milieu of pro-inflammatory mediators, neurotrophic factors, and neurotransmitters. Platelet-derived factors directly stimulate neuronal electric and synaptic activity and promote the formation of new synapses and axonal regrowth near the site of damage. Despite such active involvement in response to CNS damage, the role of platelets in neurological disorders was not extensively studied, which will be the focus of this review.
Collapse
Affiliation(s)
| | - Eugene D. Ponomarev
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
26
|
Yu H, Liu Y, He B, He T, Chen C, He J, Yang X, Wang J. Platelet biomarkers for a descending cognitive function: A proteomic approach. Aging Cell 2021; 20:e13358. [PMID: 33942972 PMCID: PMC8135080 DOI: 10.1111/acel.13358] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 02/23/2021] [Accepted: 03/15/2021] [Indexed: 12/31/2022] Open
Abstract
Memory loss is the most common clinical sign in Alzheimer's disease (AD); thus, searching for peripheral biomarkers to predict cognitive decline is promising for early diagnosis of AD. As platelets share similarities to neuron biology, it may serve as a peripheral matrix for biomarkers of neurological disorders. Here, we conducted a comprehensive and in-depth platelet proteomic analysis using TMT-LC-MS/MS in the populations with mild cognitive impairment (MCI, MMSE = 18-23), severe cognitive impairments (AD, MMSE = 2-17), and the age-/sex-matched normal cognition controls (MMSE = 29-30). A total of 360 differential proteins were detected in MCI and AD patients compared with the controls. These differential proteins were involved in multiple KEGG pathways, including AD, AMP-activated protein kinase (AMPK) pathway, telomerase RNA localization, platelet activation, and complement activation. By correlation analysis with MMSE score, three positively correlated pathways and two negatively correlated pathways were identified to be closely related to cognitive decline in MCI and AD patients. Partial least squares discriminant analysis (PLS-DA) showed that changes of nine proteins, including PHB, UQCRH, CD63, GP1BA, FINC, RAP1A, ITPR1/2, and ADAM10 could effectively distinguish the cognitively impaired patients from the controls. Further machine learning analysis revealed that a combination of four decreased platelet proteins, that is, PHB, UQCRH, GP1BA, and FINC, was most promising for predicting cognitive decline in MCI and AD patients. Taken together, our data provide a set of platelet biomarkers for predicting cognitive decline which may be applied for the early screening of AD.
Collapse
Affiliation(s)
- Haitao Yu
- Key Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineDepartment of PathophysiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
| | - Yanchao Liu
- Key Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineDepartment of PathophysiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Benrong He
- Key Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineDepartment of PathophysiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ting He
- Key Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineDepartment of PathophysiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chongyang Chen
- Key Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineDepartment of PathophysiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
| | - Jiahua He
- School of PhysicsHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
| | - Jian‐Zhi Wang
- Key Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineDepartment of PathophysiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Co‐innovation Center of NeuroregenerationNantong UniversityNantongChina
| |
Collapse
|
27
|
Babić Leko M, Nikolac Perković M, Klepac N, Švob Štrac D, Borovečki F, Pivac N, Hof PR, Šimić G. Relationships of Cerebrospinal Fluid Alzheimer's Disease Biomarkers and COMT, DBH, and MAOB Single Nucleotide Polymorphisms. J Alzheimers Dis 2021; 73:135-145. [PMID: 31771069 PMCID: PMC7029364 DOI: 10.3233/jad-190991] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The noradrenergic and dopaminergic systems are affected in Alzheimer’s disease (AD). Polymorphisms in genes encoding enzymes and proteins that are components of these systems can affect products of transcription and translation and lead to altered enzymatic activity and alterations in overall dopamine and noradrenaline levels. Catechol-O-methyltransferase (COMT) and monoamine oxidase B (MAOB) are the enzymes that regulate degradation of dopamine, while dopamine β-hydroxylase (DBH) is involved in synthesis of noradrenaline. COMT Val158Met (rs4680), DBH rs1611115 (also called –1021C/T or –970C/T), and MAOB rs1799836 (also called A644G) polymorphisms have been previously associated with AD. We assessed whether these polymorphisms are associated with cerebrospinal fluid (CSF) AD biomarkers including total tau (t-tau), phosphorylated tau proteins (p-tau181, p-tau199, and p-tau231), amyloid-β42 (Aβ42), and visinin-like protein 1 (VILIP-1) to test possible relationships of specific genotypes and pathological levels of CSF AD biomarkers. The study included 233 subjects: 115 AD, 53 mild cognitive impairment, 54 subjects with other primary causes of dementia, and 11 healthy controls. Significant decrease in Aβ42 levels was found in patients with GG compared to AG COMT Val158Met genotype, while t-tau and p-tau181 levels were increased in patients with AA compared to AG COMT Val158Met genotype. Aβ42 levels were also decreased in carriers of A allele in MAO-B rs1799836 polymorphism, while p-tau181 levels were increased in carriers of T allele in DBH rs1611115 polymorphism. These results indicate that COMT Val158Met, DBH rs1611115, and MAOB rs1799836 polymorphisms deserve further investigation as genetic markers of AD.
Collapse
Affiliation(s)
- Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | | | - Nataša Klepac
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | | | - Fran Borovečki
- Department of Neurology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Nela Pivac
- Department of Molecular Medicine, Institute Ruđer Bošković, Zagreb, Croatia
| | - Patrick R Hof
- Nash Family Department of Neuroscience, Friedman Brain Institute, and Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| |
Collapse
|
28
|
Hendrickx JO, Martinet W, Van Dam D, De Meyer GRY. Inflammation, Nitro-Oxidative Stress, Impaired Autophagy, and Insulin Resistance as a Mechanistic Convergence Between Arterial Stiffness and Alzheimer's Disease. Front Mol Biosci 2021; 8:651215. [PMID: 33855048 PMCID: PMC8039307 DOI: 10.3389/fmolb.2021.651215] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
The average age of the world's elderly population is steadily increasing. This unprecedented rise in the aged world population will increase the prevalence of age-related disorders such as cardiovascular disease (CVD) and neurodegeneration. In recent years, there has been an increased interest in the potential interplay between CVDs and neurodegenerative syndromes, as several vascular risk factors have been associated with Alzheimer's disease (AD). Along these lines, arterial stiffness is an independent risk factor for both CVD and AD. In this review, we discuss several inflammaging-related disease mechanisms including acute tissue-specific inflammation, nitro-oxidative stress, impaired autophagy, and insulin resistance which may contribute to the proposed synergism between arterial stiffness and AD.
Collapse
Affiliation(s)
- Jhana O. Hendrickx
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Debby Van Dam
- Laboratory of Neurochemistry and Behavior, Institute Born-Bunge, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- Department of Neurology and Alzheimer Research Center, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Guido R. Y. De Meyer
- Laboratory of Physiopharmacology, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
29
|
Shi Y, Gu L, Wang Q, Gao L, Zhu J, Lu X, Zhou F, Zhu D, Zhang H, Xie C, Zhang Z. Platelet Amyloid-β Protein Precursor (AβPP) Ratio and Phosphorylated Tau as Promising Indicators for Early Alzheimer's Disease. J Gerontol A Biol Sci Med Sci 2021; 75:664-670. [PMID: 31336382 DOI: 10.1093/gerona/glz005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Indexed: 12/19/2022] Open
Abstract
To identify whether platelet amyloid-β protein precursor (AβPP) ratio, phosphorylated-tau (P-tau) 231, P-tau181, and serine 396 and 404 (Ser396/404) phosphorylated tau are potential peripheral indicators for early Alzheimer's disease (AD). Forty-three amnesic mild cognitive impairment (aMCI) patients and 45 normal controls were recruited. Peripheral venous blood was drawn and platelets were collected and evaluated for potential indicators by Western blot analysis. Subsequent meta-analysis was completed on these selected indicators. In platelets of aMCI patients, the AβPP ratio level was significantly lower and levels of P-tau231 and Ser396/404 phosphorylated tau were significantly higher. Moreover, in aMCI patients, a negative correlation was observed between platelet P-tau231 level and the Trail Making Tests A score, and it was found that higher platelet P-tau231 levels significantly associated with a worse performance of information processing speed. Furthermore, values of the area under the curve of platelet P-tau231 and Ser396/404 phosphorylated tau were 0.624 and 0.657, respectively. Finally, a meta-analysis indicated platelet AβPP ratio level was significantly lower in MCI cohorts. In conclusion, platelets of aMCI subjects showed a lower AβPP ratio and higher levels of P-tau231 and Ser396/404 phosphorylated tau when compared to normal controls, which may be critical in identifying early AD.
Collapse
Affiliation(s)
- Yachen Shi
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Lihua Gu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Qing Wang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Lijuan Gao
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Jianli Zhu
- Department of Psychology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiang Lu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Fangfang Zhou
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Dan Zhu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Haisan Zhang
- Department of Psychology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Chunming Xie
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China.,Department of Psychology, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
30
|
Sakatani K, Oyama K, Hu L. Deep Learning-Based Screening Test for Cognitive Impairment Using Basic Blood Test Data for Health Examination. Front Neurol 2021; 11:588140. [PMID: 33381075 PMCID: PMC7769169 DOI: 10.3389/fneur.2020.588140] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/17/2020] [Indexed: 11/13/2022] Open
Abstract
Background: In order to develop a new screening test of cognitive impairment, we studied whether cognitive function can be estimated from basic blood test data by applying deep learning models. This model was constructed based on the effects of systemic metabolic disorders on cognitive function. Methods: We employed a deep neural network (DNN) to predict cognitive function based on subject's age and blood test items (23 items). We included 202 patients (73.48 ± 13.1 years) with various systemic metabolic disorders for training of the DNN model, and the following groups for validation of the model: (1) Patient group, 65 patients (73.6 ± 11.0 years) who were hospitalized for rehabilitation after stroke; (2) Healthy group, 37 subjects (62.0 ± 8.6 years); (3) Health examination group, 165 subjects (54.0 ± 8.6 years) admitted for a health examination. The subjects underwent the Mini-Mental State Examination (MMSE). Results: There were significant positive correlations between the predicted MMSE scores and ground truth scores in the Patient and Healthy groups (r = 0.66, p < 0.001). There were no significant differences between the predicted MMSE scores and ground truth scores in the Patient group (p > 0.05); however, in the Healthy group, the predicted MMSE scores were slightly, but significantly, lower than the ground truth scores (p < 0.05). In the Health examination group, the DNN model classified 94 subjects as normal (MMSE = 27-30), 67 subjects as having mild cognitive impairment (24-26), and four subjects as having dementia (≤ 23). In 37 subjects in the Health examination group, the predicted MMSE scores were slightly lower than the ground truth MMSE (p < 0.05). In contrast, in the subjects with neurological disorders, such as subarachnoid hemorrhage, the ground truth MMSE scores were lower than the predicted scores. Conclusions: The DNN model could predict cognitive function accurately. The predicted MMSE scores were significantly lower than the ground truth scores in the Healthy and Health examination groups, while there was no significant difference in the Patient group. We suggest that the difference between the predicted and ground truth MMSE scores was caused by changes in atherosclerosis with aging, and that applying the DNN model to younger subjects may predict future cognitive impairment after the onset of atherosclerosis.
Collapse
Affiliation(s)
- Kaoru Sakatani
- Department of Human and Engineered Environmental Studies, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan.,Institute for Healthcare Robotics, Future Robotic Organization, Waseda University, Tokyo, Japan
| | - Katsunori Oyama
- Department of Computer Science, College of Engineering, Nihon University, Tokyo, Japan
| | - Lizhen Hu
- Department of Human and Engineered Environmental Studies, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
31
|
Beyond Haemostasis and Thrombosis: Platelets in Depression and Its Co-Morbidities. Int J Mol Sci 2020; 21:ijms21228817. [PMID: 33233416 PMCID: PMC7700239 DOI: 10.3390/ijms21228817] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022] Open
Abstract
Alongside their function in primary haemostasis and thrombo-inflammation, platelets are increasingly considered a bridge between mental, immunological and coagulation-related disorders. This review focuses on the link between platelets and the pathophysiology of major depressive disorder (MDD) and its most frequent comorbidities. Platelet- and neuron-shared proteins involved in MDD are functionally described. Platelet-related studies performed in the context of MDD, cardiovascular disease, and major neurodegenerative, neuropsychiatric and neurodevelopmental disorders are transversally presented from an epidemiological, genetic and functional point of view. To provide a complete scenario, we report the analysis of original data on the epidemiological link between platelets and depression symptoms suggesting moderating and interactive effects of sex on this association. Epidemiological and genetic studies discussed suggest that blood platelets might also be relevant biomarkers of MDD prediction and occurrence in the context of MDD comorbidities. Finally, this review has the ambition to formulate some directives and perspectives for future research on this topic.
Collapse
|
32
|
Advantages and Pitfalls in Fluid Biomarkers for Diagnosis of Alzheimer's Disease. J Pers Med 2020; 10:jpm10030063. [PMID: 32708853 PMCID: PMC7563364 DOI: 10.3390/jpm10030063] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 01/08/2023] Open
Abstract
Alzheimer’s disease (AD) is a commonly occurring neurodegenerative disease in the advanced-age population, with a doubling of prevalence for each 5 years of age above 60 years. In the past two decades, there has been a sustained effort to find suitable biomarkers that may not only aide with the diagnosis of AD early in the disease process but also predict the onset of the disease in asymptomatic individuals. Current diagnostic evidence is supportive of some biomarker candidates isolated from cerebrospinal fluid (CSF), including amyloid beta peptide (Aβ), total tau (t-tau), and phosphorylated tau (p-tau) as being involved in the pathophysiology of AD. However, there are a few biomarkers that have been shown to be helpful, such as proteomic, inflammatory, oral, ocular and olfactory in the early detection of AD, especially in the individuals with mild cognitive impairment (MCI). To date, biomarkers are collected through invasive techniques, especially CSF from lumbar puncture; however, non-invasive (radio imaging) methods are used in practice to diagnose AD. In order to reduce invasive testing on the patients, present literature has highlighted the potential importance of biomarkers in blood to assist with diagnosing AD.
Collapse
|
33
|
Lee BK, Kim MH, Lee SY, Son SJ, Hong CH, Jung YS. Downregulated Platelet miR-1233-5p in Patients with Alzheimer's Pathologic Change with Mild Cognitive Impairment is Associated with Aβ-Induced Platelet Activation via P-Selectin. J Clin Med 2020; 9:E1642. [PMID: 32485903 PMCID: PMC7357133 DOI: 10.3390/jcm9061642] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) have been proposed as a promising biomarker for various diseases including Alzheimer's disease (AD). More attention has recently been focused on the diagnosis and treatment at earlier stage of mild cognitive impairment (MCI) for preventing its progression to AD. To identify potential pathologic markers for Aβ(+)MCI (Alzheimer's pathologic change with MCI), we investigated miRNA expression profiles in the platelets from patients with Aβ(+)MCI, in comparison with those from Aβ(-)MCI (Non-Alzheimer's pathologic change with MCI) and CNI (cognitively normal individuals). We found that let-7i-5p, miR-125a, miR-1233-5p, and miR-6787-5p were significantly downregulated, while miR-6880-5p expression was upregulated. Of these, only miR-1233-5p was significantly downregulated by Aβ treatment in both human platelets and their precursor megakaryocytes (MEG-01 cells). We explored the role of miRNAs by using miRNA mimics or inhibitors, and found that the diminished level of miR-1233-5p was associated with Aβ-induced increase in the expression of P-selectin and cell adhesion to fibronectin. Our results further indicated that Aβ-induced increase in platelet/MEG adhesion to fibronectin is likely mediated via P-selectin. In conclusion, this study suggests the downregulation of platelet-derived miR-1233-5p as a pathologic marker for Aβ(+)MCI.
Collapse
Affiliation(s)
- Bo Kyung Lee
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (B.K.L.); or (M.H.K.)
| | - Min Hee Kim
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (B.K.L.); or (M.H.K.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
| | - Sang Yoon Lee
- Department of Biomedical Sciences, School of Medicine, Ajou University, Suwon 16499, Korea;
| | - Sang Joon Son
- Department of Psychiatry, School of Medicine, Ajou University, Suwon 16499, Korea;
| | - Chang Hyung Hong
- Department of Psychiatry, School of Medicine, Ajou University, Suwon 16499, Korea;
| | - Yi-Sook Jung
- College of Pharmacy, Ajou University, Suwon 16499, Korea; (B.K.L.); or (M.H.K.)
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Korea
| |
Collapse
|
34
|
Leiter O, Walker TL. Platelets in Neurodegenerative Conditions-Friend or Foe? Front Immunol 2020; 11:747. [PMID: 32431701 PMCID: PMC7214916 DOI: 10.3389/fimmu.2020.00747] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
It is now apparent that platelet function is more diverse than originally thought, shifting the view of platelets from blood cells involved in hemostasis and wound healing to major contributors to numerous regulatory processes across different tissues. Given their intriguing ability to store, produce and release distinct subsets of bioactive molecules, including intercellular signaling molecules and neurotransmitters, platelets may play an important role in orchestrating healthy brain function. Conversely, a number of neurodegenerative conditions have recently been associated with platelet dysfunction, further highlighting the tissue-independent role of these cells. In this review we summarize the requirements for platelet-neural cell communication with a focus on neurodegenerative diseases, and discuss the therapeutic potential of healthy platelets and the proteins which they release to counteract these conditions.
Collapse
Affiliation(s)
- Odette Leiter
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Tara L Walker
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
35
|
Clinical implications of oxidative stress in schizophrenia: Acute relapse and chronic stable phase. Prog Neuropsychopharmacol Biol Psychiatry 2020; 99:109868. [PMID: 31954755 DOI: 10.1016/j.pnpbp.2020.109868] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 12/03/2019] [Accepted: 01/15/2020] [Indexed: 12/22/2022]
Abstract
Several studies have suggested a higher oxidative stress in schizophrenia. However, the implications of oxidative stress on clinical symptoms remain unclear. This study aimed to investigate the platelet oxidative stress in different stages of schizophrenia (i.e., chronic stable and acute relapse) in order to clarify the clinical implications of oxidative stress and the treatment effects. We recruited 43 chronic stable patients with schizophrenia and 48 non-psychiatric controls. Platelets were collected for measuring the levels of nitric oxide (NO), lipid peroxidation (LPO), and glutathione (GSH) and the activity of GSH peroxidase (GPx) and superoxide dismutase (SOD). The levels and activity were compared between patients and controls and were examined for their relationship with clinical severity. Further, we evaluated the changes of levels and activity before and after treatment in an independent sample with acute relapse (N = 19). Patients with chronic stable schizophrenia had lower SOD activity compared to non-psychiatric controls. In chronic stable patients, NO level was positively correlated with positive and disorganized symptoms, while the GPx activity were negatively correlated with excitement. In patients with acute relapse, the levels and activity were not different before and after four weeks of antipsychotic treatment, but LPO level was negatively correlated with pretreatment disorganized symptoms. The change of LPO can also predict the change of disorganized symptoms and negative symptoms. Our findings suggest that platelet SOD was lower in chronic stable schizophrenia. Platelet LPO may be associated with less disorganized symptoms in acute relapse patients and better treatment response.
Collapse
|
36
|
Kopeikina E, Dukhinova M, Yung AWY, Veremeyko T, Kuznetsova IS, Lau TYB, Levchuk K, Ponomarev ED. Platelets promote epileptic seizures by modulating brain serotonin level, enhancing neuronal electric activity, and contributing to neuroinflammation and oxidative stress. Prog Neurobiol 2020; 188:101783. [PMID: 32142857 DOI: 10.1016/j.pneurobio.2020.101783] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 02/04/2020] [Accepted: 02/24/2020] [Indexed: 11/28/2022]
Abstract
The drugs currently available for treating epilepsy are only partially effective in managing this condition. Therefore, it is crucial to investigate new pathways that induce and promote epilepsy development. Previously, we found that platelets interact with neuronal glycolipids and actively secrete pro-inflammatory mediators during central nervous system (CNS) pathological conditions such as neuroinflammation and traumatic brain injury (TBI). These factors increase the permeability of the blood-brain barrier (BBB), which may create a predisposition to epileptic seizures. In this study, we demonstrated that platelets substantially enhanced epileptic seizures in a mouse model of pentylenetetrazole (PTZ) -induced seizures. We found that platelets actively secreted serotonin, contributed to increased BBB permeability, and were present in the CNS parenchyma during epileptic seizures. Furthermore, platelets directly stimulated neuronal electric activity and induced the expression of specific genes related to early neuronal response, neuroinflammation, and oxidative phosphorylation, leading to oxidative stress in neurons. The intracranial injection of physiological numbers of platelets that mimicked TBI-associated bleeding was sufficient to induce severe seizures, which resembled conventional PTZ-induced epileptic activity. These findings highlight a conceptually new role of platelets in the development of epileptic seizures, and indicate a potential new therapeutic approach targeting platelets to prevent and treat epilepsy.
Collapse
Affiliation(s)
- Ekaterina Kopeikina
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Marina Dukhinova
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Amanda W Y Yung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Tatyana Veremeyko
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Inna S Kuznetsova
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Thomas Y B Lau
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Kseniia Levchuk
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Eugene D Ponomarev
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong.
| |
Collapse
|
37
|
dos Santos GAA, Pardi PC. Biomarkers in Alzheimer's disease: Evaluation of platelets, hemoglobin and vitamin B12. Dement Neuropsychol 2020; 14:35-40. [PMID: 32206196 PMCID: PMC7077854 DOI: 10.1590/1980-57642020dn14-010006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 12/21/2019] [Indexed: 01/09/2023] Open
Abstract
Currently, the most likely hypotheses as the cause of Alzheimer's disease are deposition of amyloid beta peptide in the cerebral cortex and hyperphosphorylation of Tau protein. The diagnosis of Alzheimer's disease is based on the exclusion of other diseases, behavioral assessments, and blood and imaging tests. Biotechnology has created interesting perspectives for the early detection of Alzheimer's disease through blood analysis, with special attention to platelets, hemoglobin and vitamin B12. OBJECTIVE To evaluate the concentrations of platelets, hemoglobin and vitamin B12 in the blood of older adults with and without dementia of Alzheimer's disease. METHODS A case-control study involving 120 individuals was conducted, seeking to establish a correlation between changes in platelet, hemoglobin and vitamin B12 concentrations in patients with confirmed AD and in individuals in the inclusion group without AD. The study met the established ethical requirements. RESULTS Hemoglobin and platelet levels were statistically lower in patients with AD. The biochemical evaluation in AD patient and healthy groups for vitamin B12 showed a decrease in the levels of this compound in patients with AD. CONCLUSION We demonstrated the feasibility of the use of blood biomarkers as predictive markers for the diagnosis of AD.
Collapse
Affiliation(s)
- Gustavo Alves Andrade dos Santos
- Universidade de São Paulo – USP. Faculdade de Medicina de Ribeirão
Preto. Departamento de Anatomia e Cirurgia. Ribeirão Preto, SP, Brazil
- Centro Universitário do Senac – Unidade Tiradentes. Departamento de
Pós-graduação em Farmácia
| | - Paulo Celso Pardi
- Universidade Anhanguera Guarulhos. Departamento de Biomedicina, São
Paulo, SP, Brazil
| |
Collapse
|
38
|
Bram JMDF, Talib LL, Joaquim HPG, Sarno TA, Gattaz WF, Forlenza OV. Protein levels of ADAM10, BACE1, and PSEN1 in platelets and leukocytes of Alzheimer's disease patients. Eur Arch Psychiatry Clin Neurosci 2019; 269:963-972. [PMID: 29845446 DOI: 10.1007/s00406-018-0905-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 05/22/2018] [Indexed: 12/31/2022]
Abstract
The clinical diagnosis of Alzheimer's disease (AD) is a probabilistic formulation that may lack accuracy particularly at early stages of the dementing process. Abnormalities in amyloid-beta precursor protein (APP) metabolism and in the level of APP secretases have been demonstrated in platelets, and to a lesser extent in leukocytes, of AD patients, with conflicting results. The aim of the present study was to compare the protein level of the APP secretases A-disintegrin and metalloprotease 10 (ADAM10), Beta-site APP-cleaving enzyme 1 (BACE1), and presenilin-1 (PSEN1) in platelets and leukocytes from 20 non-medicated older adults with AD and 20 healthy elders, and to determine the potential use of these biomarkers to discriminate cases of AD from controls. The protein levels of all APP secretases were significantly higher in platelets compared to leukocytes. We found statistically a significant decrease in ADAM10 (52.5%, p < 0.0001) and PSEN1 (32%, p = 0.02) in platelets from AD patients compared to controls, but not in leukocytes. Combining all three secretases to generate receiver-operating characteristic (ROC) curves, we found a good discriminatory effect (AD vs. controls) when using platelets (the area under the curve-AUC-0.90, sensitivity 88.9%, specificity 66.7%, p = 0.003), but not in leukocytes (AUC 0.65, sensitivity 77.8%, specificity 50.0%, p = 0.2). Our findings indicate that platelets represent a better biological matrix than leukocytes to address the peripheral level of APP secretases. In addition, combining the protein level of ADAM10, BACE1, and PSEN1 in platelets, yielded a good accuracy to discriminate AD from controls.
Collapse
Affiliation(s)
- Jessyka Maria de França Bram
- Laboratorio de Neurociencias (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da USP (HCFMUSP), Faculdade de Medicina, Universidade de Sao Paulo, Rua Doutor Ovídio Pires de Campos 785, São Paulo, SP, 05403-010, Brazil
| | - Leda Leme Talib
- Laboratorio de Neurociencias (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da USP (HCFMUSP), Faculdade de Medicina, Universidade de Sao Paulo, Rua Doutor Ovídio Pires de Campos 785, São Paulo, SP, 05403-010, Brazil
| | - Helena Passarelli Giroud Joaquim
- Laboratorio de Neurociencias (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da USP (HCFMUSP), Faculdade de Medicina, Universidade de Sao Paulo, Rua Doutor Ovídio Pires de Campos 785, São Paulo, SP, 05403-010, Brazil
| | - Tamires Alves Sarno
- Laboratorio de Neurociencias (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da USP (HCFMUSP), Faculdade de Medicina, Universidade de Sao Paulo, Rua Doutor Ovídio Pires de Campos 785, São Paulo, SP, 05403-010, Brazil
| | - Wagner Farid Gattaz
- Laboratorio de Neurociencias (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da USP (HCFMUSP), Faculdade de Medicina, Universidade de Sao Paulo, Rua Doutor Ovídio Pires de Campos 785, São Paulo, SP, 05403-010, Brazil
| | - Orestes Vicente Forlenza
- Laboratorio de Neurociencias (LIM-27), Departamento e Instituto de Psiquiatria, Hospital das Clínicas da Faculdade de Medicina da USP (HCFMUSP), Faculdade de Medicina, Universidade de Sao Paulo, Rua Doutor Ovídio Pires de Campos 785, São Paulo, SP, 05403-010, Brazil.
| |
Collapse
|
39
|
Melchinger H, Jain K, Tyagi T, Hwa J. Role of Platelet Mitochondria: Life in a Nucleus-Free Zone. Front Cardiovasc Med 2019; 6:153. [PMID: 31737646 PMCID: PMC6828734 DOI: 10.3389/fcvm.2019.00153] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/08/2019] [Indexed: 12/19/2022] Open
Abstract
Platelets are abundant, small, anucleate circulating cells, serving many emerging pathophysiological roles beyond hemostasis; including active critical roles in thrombosis, injury response, and immunoregulation. In the absence of genomic DNA transcriptional regulation (no nucleus), platelets require strategic prepackaging of all the needed RNA and organelles from megakaryocytes, to sense stress (e.g., hyperglycemia), to protect themselves from stress (e.g., mitophagy), and to communicate a stress response to other cells (e.g., granule and microparticle release). Distinct from avian thrombocytes that have a nucleus, the absence of a nucleus allows the mammalian platelet to maintain its small size, permits morphological flexibility, and may improve speed and efficiency of protein expression in response to stress. In the absence of a nucleus, platelet lifespan of 7–10 days, is largely determined by the mitochondria. The packaging of 5–8 mitochondria is critical in aerobic respiration and yielding metabolic substrates needed for function and survival. Mitochondria damage or dysfunction, as observed with several disease processes, results in greatly attenuated platelet survival and increased risk for thrombovascular events. Here we provide insights into the emerging roles of platelets despite the lack of a nucleus, and the key role played by mitochondria in platelet function and survival both in health and disease.
Collapse
Affiliation(s)
- Hannah Melchinger
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Kanika Jain
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - Tarun Tyagi
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| | - John Hwa
- Department of Internal Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
40
|
Akingbade OES, Gibson C, Kalaria RN, Mukaetova-Ladinska EB. Platelets: Peripheral Biomarkers of Dementia? J Alzheimers Dis 2019; 63:1235-1259. [PMID: 29843245 DOI: 10.3233/jad-180181] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Dementia continues to be the most burdening neurocognitive disorder, having a negative impact on the lives of millions. The search for biomarkers to improve the clinical diagnosis of dementia is ongoing, with the focus on effective use of readily accessible peripheral markers. In this review, we concentrate on platelets as biomarkers of dementia and analyze their potential as easily-accessible clinical biomarkers for various subtypes of dementia. Current platelet protein biomarkers that have been investigated for their clinical utility in the diagnosis of dementia, in particular Alzheimer's disease, include amyloid-β protein precursor (AβPP), the AβPP secretases (BACE1 and ADAM10), α-synuclein, tau protein, serotonin, cholesterol, phospholipases, clusterin, IgG, surface receptors, MAO-B, and coated platelets. Few of them, i.e., platelet tau, AβPP (particularly with regards to coated platelets) and secreted ADAM10 and BACE1 show the most promise to be taken forward into clinical setting to diagnose dementia. Aside from protein biomarkers, changes in factors such as mean platelet volume have the potential to play a very specific role in both the dementia diagnosis and prognosis. This review raises a number of research questions for consideration before application of the above biomarkers to routine clinical setting. It is without doubt that there is a need for more clarification on the effects of dementia on platelet morphology and protein content before these changes can be clinically applied as dementia biomarkers and explored further in differentiating distinct dementia subtypes.
Collapse
Affiliation(s)
- Oluwatomi E S Akingbade
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK.,School of Life Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Claire Gibson
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| | - Raj N Kalaria
- Institute of Neuroscience, Newcastle University, Newcastle, UK
| | - Elizabeta B Mukaetova-Ladinska
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK.,Evington Centre, Leicester General Hospital, Leicester, UK
| |
Collapse
|
41
|
de Castro AA, Soares FV, Pereira AF, Polisel DA, Caetano MS, Leal DHS, da Cunha EFF, Nepovimova E, Kuca K, Ramalho TC. Non-conventional compounds with potential therapeutic effects against Alzheimer’s disease. Expert Rev Neurother 2019; 19:375-395. [DOI: 10.1080/14737175.2019.1608823] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Alexandre A. de Castro
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Flávia V. Soares
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Ander F. Pereira
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Daniel A. Polisel
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Melissa S. Caetano
- Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Daniel H. S. Leal
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
- Department of Health Sciences, Federal University of Espírito Santo, São Mateus, Brazil
| | - Elaine F. F. da Cunha
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Teodorico C. Ramalho
- Laboratory of Molecular Modeling, Department of Chemistry, Federal University of Lavras, Lavras, Brazil
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
42
|
Pluta R, Ułamek-Kozioł M. Lymphocytes, Platelets, Erythrocytes, and Exosomes as Possible Biomarkers for Alzheimer’s Disease Clinical Diagnosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1118:71-82. [DOI: 10.1007/978-3-030-05542-4_4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
43
|
Dukhinova M, Kuznetsova I, Kopeikina E, Veniaminova E, Yung AWY, Veremeyko T, Levchuk K, Barteneva NS, Wing-Ho KK, Yung WH, Liu JYH, Rudd J, Yau SSY, Anthony DC, Strekalova T, Ponomarev ED. Platelets mediate protective neuroinflammation and promote neuronal plasticity at the site of neuronal injury. Brain Behav Immun 2018; 74:7-27. [PMID: 30217533 DOI: 10.1016/j.bbi.2018.09.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 09/10/2018] [Accepted: 09/10/2018] [Indexed: 01/05/2023] Open
Abstract
It is generally accepted that inflammation within the CNS contributes to neurodegeneration after traumatic brain injury (TBI), but it is not clear how inflammation is initiated in the absence of infection and whether this neuroinflammation is predominantly beneficial or detrimental. We have previously found that brain-enriched glycosphingolipids within neuronal lipid rafts (NLR) induced platelet degranulation and secretion of neurotransmitters and pro-inflammatory factors. In the present study, we compared TBI-induced inflammation and neurodegeneration in wild-type vs. St3gal5 deficient (ST3-/-) mice that lack major CNS-specific glycosphingolipids. After TBI, microglial activation and CNS macrophage infiltration were substantially reduced in ST3-/- animals. However, ST3-/- mice had a larger area of CNS damage with marked neuronal/axonal loss. The interaction of platelets with NLR stimulated neurite growth, increased the number of PSD95-positive dendritic spines, and intensified neuronal activity. Adoptive transfer and blocking experiments provide further that platelet-derived serotonin and platelet activating factor plays a key role in the regulation of sterile neuroinflammation, hemorrhage and neuronal plasticity after TBI.
Collapse
Affiliation(s)
- Marina Dukhinova
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Inna Kuznetsova
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Ekaterina Kopeikina
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Ekaterina Veniaminova
- Department of Neuroscience, Maastricht University, Universiteitssingel 40, NL 6229ER, Maastricht, Netherlands; Institute of General Pathology and Pathophysiology, Baltiiskaya str, 8, Moscow, 125315, Russia; Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Laboratory of Psychiatric Neurobiology, Trubetskaya Street 8-2, 119991, Moscow, Russia
| | - Amanda W Y Yung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Tatyana Veremeyko
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Kseniia Levchuk
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Natasha S Barteneva
- Program in Cellular and Molecular Medicine, Children's Hospital Boston and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Kenny Kam Wing-Ho
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Wing-Ho Yung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - Julia Y H Liu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong
| | - John Rudd
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong; Brain and Mind Institute, The Chinese University of Hong Kong, Shatin NT, Hong Kong
| | - Sonata S Y Yau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Oxford OX1 3QT, UK
| | - Tatyana Strekalova
- Department of Neuroscience, Maastricht University, Universiteitssingel 40, NL 6229ER, Maastricht, Netherlands; Institute of General Pathology and Pathophysiology, Baltiiskaya str, 8, Moscow, 125315, Russia; Sechenov First Moscow State Medical University, Institute of Molecular Medicine, Laboratory of Psychiatric Neurobiology, Trubetskaya Street 8-2, 119991, Moscow, Russia
| | - Eugene D Ponomarev
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin N.T., Hong Kong; Kunming Institute of Zoology and Chinese University of Hong Kong Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunmin-Hong Kong, China.
| |
Collapse
|
44
|
The Correlations of Plasma and Cerebrospinal Fluid Amyloid-Beta Levels with Platelet Count in Patients with Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7302045. [PMID: 30410937 PMCID: PMC6205107 DOI: 10.1155/2018/7302045] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/30/2018] [Indexed: 11/18/2022]
Abstract
Purpose Recent study shows that blood-derived amyloid-beta (Aβ) can induce cerebral amyloidosis and is involved in the pathogenesis of Alzheimer's disease (AD). The vast majority of blood Aβ is generated from platelet. Whether blood Aβ levels are associated with the count of platelets remains unknown. Methods 58 clinically diagnosed AD patients, 18 11C-PIB-PET diagnosed AD patients, and 61 age- and gender-matched cognitively normal controls were included to analyze the correlation of plasma Aβ levels with platelet count. 13 AD patients and 40 controls with cerebrospinal fluid (CSF) samples were included to further analyze the correlation of CSF Aβ levels with platelet count. Aβ40 and Aβ42 levels in plasma and CSF were measured by ELISA kits. Results The plasma Aβ42 level was positively correlated with platelet count in both AD patients and control group, especially in AD patients with positive PIB-PET, while there was no correlation as to Aβ40. The CSF Aβ levels also had no significant correlation with platelet count. Conclusion It suggests that platelets may be involved in the pathogenesis of AD and become a potential peripheral biomarker for AD.
Collapse
|
45
|
Daniele S, Pietrobono D, Fusi J, Lo Gerfo A, Cerri E, Chico L, Iofrida C, Petrozzi L, Baldacci F, Giacomelli C, Galetta F, Siciliano G, Bonuccelli U, Trincavelli ML, Franzoni F, Martini C. α-Synuclein Aggregated with Tau and β-Amyloid in Human Platelets from Healthy Subjects: Correlation with Physical Exercise. Front Aging Neurosci 2018; 10:17. [PMID: 29441013 PMCID: PMC5797553 DOI: 10.3389/fnagi.2018.00017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 01/15/2018] [Indexed: 12/19/2022] Open
Abstract
The loss of protein homeostasis that has been associated with aging leads to altered levels and conformational instability of proteins, which tend to form toxic aggregates. In particular, brain aging presents characteristic patterns of misfolded oligomers, primarily constituted of β-amyloid (Aβ), tau, and α-synuclein (α-syn), which can accumulate in neuronal membranes or extracellular compartments. Such aging-related proteins can also reach peripheral compartments, thus suggesting the possibility to monitor their accumulation in more accessible fluids. In this respect, we have demonstrated that α-syn forms detectable hetero-aggregates with Aβ or tau in red blood cells (RBCs) of healthy subjects. In particular, α-syn levels and its heteromeric interactions are modulated by plasma antioxidant capability (AOC), which increases in turn with physical activity. In order to understand if a specific distribution of misfolded proteins can occur in other blood cells, a cohort of human subjects was enrolled to establish a correlation among AOC, the level of physical exercise and the concentrations of aging-related proteins in platelets. The healthy subjects were divided depending on their level of physical exercise (i.e., athletes and sedentary subjects) and their age (young and older subjects). Herein, aging-related proteins (i.e., α-syn, tau and Aβ) were confirmed to be present in human platelets. Among such proteins, platelet tau concentration was demonstrated to decrease in athletes, while α-syn and Aβ did not correlate with physical exercise. For the first time, α-syn was shown to directly interact with Aβ and tau in platelets, forming detectable hetero-complexes. Interestingly, α-syn interaction with tau was inversely related to plasma AOC and to the level of physical activity. These results suggested that α-syn heterocomplexes, particularly with tau, could represent novel indicators to monitor aging-related proteins in platelets.
Collapse
Affiliation(s)
| | | | - Jonathan Fusi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Annalisa Lo Gerfo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eugenio Cerri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Lucia Chico
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Lucia Petrozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Fabio Galetta
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Ferdinando Franzoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | |
Collapse
|
46
|
Reumiller CM, Schmidt GJ, Dhrami I, Umlauf E, Rappold E, Zellner M. Gender-related increase of tropomyosin-1 abundance in platelets of Alzheimer's disease and mild cognitive impairment patients. J Proteomics 2017; 178:73-81. [PMID: 29278785 DOI: 10.1016/j.jprot.2017.12.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/19/2017] [Accepted: 12/22/2017] [Indexed: 01/13/2023]
Abstract
The incidence of Alzheimer's disease (AD) is higher in elderly women than in men. The molecular background of this gender-related risk, however, is largely unknown. In a previous proteomics study, we identified significantly elevated levels of monoamine oxidase-B and tropomyosin-1 in AD patients, together with significant changes of the genetic AD risk factors apolipoprotein E4 (APOE4) and glutathione S-transferase omega 1 (GSTO1), in platelets - a promising source for AD blood biomarkers. The present study aimed to investigate the gender-specificity as well as the disease-stage dependency of these biomarkers in AD patients and those with mild cognitive impairment (MCI). Tropomyosin-1 and monoamine oxidase-B protein levels were quantified by 2-D DIGE and 1-D Western blotting. Here, for the first time, we revealed a significant increase of 38&39kDa tropomyosin-1 protein levels in female but not male AD (+56%; p=0.008) and MCI patients (+46%; p=0.041) measured by 1-D WB. In contrast, levels of monoamine oxidase-B were, independently of gender, elevated in AD patients (+52%; p=0.009) but unaltered in MCI compared to control subjects. Moreover, we confirmed that APOE4-positive females are at a higher risk (OR=18.7; p=9.7E-09) of developing AD compared to APOE4-positive males (OR=6.5; p=5.9E-04). No gender-related effects were observed for GSTO1. SIGNIFICANCE Platelet tropomyosin-1 constitutes a gender-related and stage-dependent protein in cognitive impairment. In contrast, platelet monoamine oxidase-B, frequently described to be increased in platelets and brains of AD patients, shows a gender-independent but stage-related increase since it is unaltered in MCI subjects. A blood biomarker test for this preceding stage of AD that considers gender-specificity is not yet available. The newly described AD-related platelet protein profiles might refine and facilitate routine diagnosis and enable early as well as tailored interventions.
Collapse
Affiliation(s)
- Christina Maria Reumiller
- Center of Physiology and Pharmacology, Institute of Physiology, Medical University of Vienna, Vienna, Austria
| | | | - Ina Dhrami
- Center of Physiology and Pharmacology, Institute of Physiology, Medical University of Vienna, Vienna, Austria
| | - Ellen Umlauf
- Center of Physiology and Pharmacology, Institute of Physiology, Medical University of Vienna, Vienna, Austria
| | - Eduard Rappold
- Center of Physiology and Pharmacology, Institute of Physiology, Medical University of Vienna, Vienna, Austria
| | - Maria Zellner
- Center of Physiology and Pharmacology, Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
47
|
Identification of post-translational modifications of Aβ peptide in platelet membranes from patients with cerebral amyloid angiopathy. J Neurol Sci 2017; 383:11-17. [DOI: 10.1016/j.jns.2017.08.3269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/28/2017] [Accepted: 08/31/2017] [Indexed: 01/13/2023]
|
48
|
Kumar S, Chowdhury S, Kumar S. In silico repurposing of antipsychotic drugs for Alzheimer's disease. BMC Neurosci 2017; 18:76. [PMID: 29078760 PMCID: PMC5660441 DOI: 10.1186/s12868-017-0394-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 10/19/2017] [Indexed: 12/16/2022] Open
Abstract
Background Alzheimer’s disease (AD) is the most prevalent form of dementia and represents one of the highest unmet requirements in medicine today. There is shortage of novel molecules entering into market because of poor pharmacokinetic properties and safety issues. Drug repurposing offers an opportunity to reinvigorate the slowing drug discovery process by finding new uses for existing drugs. The major advantage of the drug repurposing approach is that the safety issues are already investigated in the clinical trials and the drugs are commercially available in the marketplace. As this approach provides an effective solution to hasten the process of providing new alternative drugs for AD, the current study shows the molecular interaction of already known antipsychotic drugs with the different protein targets implicated in AD using in silico studies. Result A computational method based on ligand–protein interaction was adopted in present study to explore potential antipsychotic drugs for the treatment of AD. The screening of approximately 150 antipsychotic drugs was performed on five major protein targets (AChE, BuChE, BACE 1, MAO and NMDA) by molecular docking. In this study, for each protein target, the best drug was identified on the basis of dock score and glide energy. The top hits were then compared with the already known inhibitor of the respective proteins. Some of the drugs showed relatively better docking score and binding energies as compared to the already known inhibitors of the respective targets. Molecular descriptors like molecular weight, number of hydrogen bond donors, acceptors, predicted octanol/water partition coefficient and percentage human oral absorption were also analysed to determine the in silico ADME properties of these drugs and all were found in the acceptable range and follows Lipinski’s rule. Conclusion The present study have led to unravel the potential of leading antipsychotic drugs such as pimozide, bromperidol, melperone, anisoperidone, benperidol and anisopirol against multiple targets associated with AD. Benperidol was found to be the best candidate drug interacting with different target proteins involved in AD.
Collapse
Affiliation(s)
- Shivani Kumar
- University School of Biotechnology, GGS Indraprastha University, Sector-16C, Dwarka, New Delhi, 110075, India
| | - Suman Chowdhury
- University School of Biotechnology, GGS Indraprastha University, Sector-16C, Dwarka, New Delhi, 110075, India
| | - Suresh Kumar
- University School of Biotechnology, GGS Indraprastha University, Sector-16C, Dwarka, New Delhi, 110075, India.
| |
Collapse
|
49
|
Wojsiat J, Laskowska-Kaszub K, Mietelska-Porowska A, Wojda U. Search for Alzheimer's disease biomarkers in blood cells: hypotheses-driven approach. Biomark Med 2017; 11:917-931. [PMID: 28976776 DOI: 10.2217/bmm-2017-0041] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Current Alzheimer's disease (AD) diagnostics is based on cognitive testing, and detecting amyloid Aβ and τ pathology by brain imaging and assays of cerebrospinal fluid. However, biomarkers identifying complex pathways contributing to pathology are lacking, especially for early AD. Preferably, such biomarkers should be more cost-effective and present in easily available diagnostic tissues, such as blood. Here, we summarize the recent findings of potential early AD molecular diagnostic biomarkers in blood platelets, lymphocytes and erythrocytes. We review molecular alterations which refer to such main hypotheses of AD pathogenesis as amyloid cascade, oxidative and mitochondrial stress, inflammation and alterations in cell cycle regulatory molecules. The major advantage of such biomarkers is the potential ability to indicate individualized therapies in AD patients.
Collapse
Affiliation(s)
- Joanna Wojsiat
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology, Polish Academy of Science, Pasteur 3 St., 02-093 Warsaw, Poland
| | - Katarzyna Laskowska-Kaszub
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology, Polish Academy of Science, Pasteur 3 St., 02-093 Warsaw, Poland
| | - Anna Mietelska-Porowska
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology, Polish Academy of Science, Pasteur 3 St., 02-093 Warsaw, Poland
| | - Urszula Wojda
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology, Polish Academy of Science, Pasteur 3 St., 02-093 Warsaw, Poland
| |
Collapse
|
50
|
Stevenson A, Lopez D, Khoo P, Kalaria RN, Mukaetova-Ladinska EB. Exploring Erythrocytes as Blood Biomarkers for Alzheimer’s Disease. J Alzheimers Dis 2017; 60:845-857. [DOI: 10.3233/jad-170363] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Anna Stevenson
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
- The School of Life Sciences, University of Glasgow, University Avenue, Glasgow, UK
| | - Dianne Lopez
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Paul Khoo
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Rajesh N. Kalaria
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | | |
Collapse
|