1
|
Templeton HN, Tobet SA, Schwerdtfeger LA. Gut neuropeptide involvement in Parkinson's disease. Am J Physiol Gastrointest Liver Physiol 2025; 328:G716-G733. [PMID: 40279198 DOI: 10.1152/ajpgi.00383.2024] [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: 12/05/2024] [Revised: 01/09/2025] [Accepted: 04/21/2025] [Indexed: 04/27/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder affecting over 10 million people. A key pathological feature of PD is the accumulation of misfolded α-synuclein (aSyn) protein in the substantia nigra pars compacta. Aggregation of aSyn can form Lewy bodies that contribute to dopaminergic neuron degeneration and motor symptoms, such as tremor, rigidity, and bradykinesia. Beyond the central nervous system, aSyn aggregates have been detected in the gastrointestinal (GI) tract, suggesting a link between peripheral aSyn and nonmotor PD symptoms. GI symptoms, often preceding motor symptoms by up to 20 years, highlight the bidirectional communication between the central nervous system and the enteric nervous system (gut-brain axis) in PD. Although microbiome alterations and intestinal inflammation have been associated with PD, functional impacts on gut-brain signaling or aSyn aggregation remain unclear. Intestinal neuropeptides are key modulators of gut-brain communication, alter immune response to pathogens and environmental toxins, and may contribute to the function of the luminal gut barrier. Dysregulation of gut neuropeptide signaling, including vasoactive intestinal peptide, neuropeptide Y, calcitonin gene-related peptide, ghrelin, cholecystokinin, glucagon-like peptide 1, and substance P, have been associated with pathologic effects of PD in animal models. Despite their potential role in pathogenesis and disease modulation, gut neuropeptide roles in PD are underexplored. This article reviews current knowledge surrounding microbial metabolite and immune influences on gut neuropeptide signaling, aSyn aggregation in the enteric nervous system, and downstream neuroimmune pathway alterations within the context of PD and its mouse models.
Collapse
Affiliation(s)
- Hayley N Templeton
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
| | - Stuart A Tobet
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado, United States
| | - Luke A Schwerdtfeger
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States
- Ann Romney Center for Neurological Disease, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
2
|
Sohail MU, Batool RM, Aamir J, Saad M, Aisha E, Jain H, Arshad MS, Ahmed R. Trends in Type 2 Diabetes Mellitus and Parkinson's Disease Related Mortality in the United States from 1999 to 2020. Diabetes Res Clin Pract 2025; 224:112239. [PMID: 40345594 DOI: 10.1016/j.diabres.2025.112239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 05/01/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Emerging evidence indicates that individuals with Type 2 Diabetes Mellitus (T2DM) are at an elevated risk of Parkinson's Disease (PD). While mortality trends for each condition have been studied individually, the combined burden of T2DM- and PD-related mortality remains poorly understood. This study aims to evaluate national trends and disparities in T2DM and PD related mortality among older adults in the United States (U.S.). from 1999 to 2020. METHODS Using CDC WONDER database, we analyzed deaths among U.S. residents aged 65 + from 1999 to 2020, identifying T2DM (ICD-10: E11) and PD (ICD-10: G20) related deaths. Crude and age-adjusted mortality rates (AAMR) per 100,000 individuals were calculated, and Joinpoint regression analysis was employed to estimate the annual percent change (APC). RESULTS Between 1999 and 2020, a total of 26,020 deaths occurred among older adults with T2DM and PD. The AAMR increased from 1.65 in 1999 to 5.61 in 2020, with a sharp rise between 2015 and 2020 (APC: +14.42 %; 95 % CI: 11.22 to 20.80). Males experienced higher AAMRs than females (4.51 vs. 1.80). Across racial groups, Hispanic or Latino individuals exhibited the highest AAMR (3.61), while non-Hispanic Black Americans had the lowest (1.96). Non-metropolitan areas experienced higher AAMRs than metropolitan areas (3.64 vs. 2.71). CONCLUSIONS T2DM and PD-related mortality has surged over the past two decades, particularly since 2015, with significant racial, sex-based, and regional disparities. Targeted public health strategies are needed to address these growing health concerns.
Collapse
Affiliation(s)
| | | | - Jazza Aamir
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Muhammad Saad
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan.
| | - Eliza Aisha
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Hritvik Jain
- All India Institute of Medical Sciences, Jodhpur, India
| | | | - Raheel Ahmed
- National Heart and Lung Institute, Imperial College London, London, United Kingdom.
| |
Collapse
|
3
|
Berezhnoy G, Bae G, Wüst L, Schulte C, Cannet C, Wurster I, Zimmermann M, Jäck A, Spruth EJ, Hellmann-Regen J, Roeske S, Pürner D, Glanz W, Maass F, Hufschmidt F, Kilimann I, Dinter E, Kimmich O, Gamez A, Levin J, Priller J, Peters O, Wagner M, Storch A, Lingor P, Düzel E, van Riesen C, Wüllner U, Teipel S, Falkenburger B, Bähr M, Zerr I, Petzold GC, Spottke A, Rizzu P, Brosseron F, Schäfer H, Gasser T, Trautwein C. Application of IVDr NMR spectroscopy to stratify Parkinson's disease with absolute quantitation of blood serum metabolites and lipoproteins. Sci Rep 2025; 15:17738. [PMID: 40404791 PMCID: PMC12098827 DOI: 10.1038/s41598-025-01352-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 05/06/2025] [Indexed: 05/24/2025] Open
Abstract
The challenge of early detection and stratification in Parkinson's disease (PD) is urgent due to the current emergence of mechanism-based disease-modifying treatments. In here, metabolomic and lipidomic parameters obtained by a standardized and targeted in vitro diagnostic research (IVDr) platform have a significant potential to address therapy-related questions and generate improved biomarker panels. Our study aimed to use IVDr nuclear magnetic resonance (NMR) spectroscopy to quantify metabolites and lipoproteins in PD blood serum from different cohorts to stratify metabolically driven subtypes of idiopathic and genetic PD. Serum aliquots from three neurodegeneration biobank cohorts (287 samples in total, including 62 PD patient samples with GBA mutation, 98/43 PD patient samples of early/late stages of disease duration, 20 PD samples from patients with mutations in recessive PD genes and some smaller subgroups of mitochondrial and double mutation cases) were prepared and analyzed with IVDr NMR spectroscopy, covering 39 blood serum metabolites and 112 lipoprotein parameters. Uni- and multivariate statistics were used to identify metabolism-driven changes under consideration of typical confounders such as age, sex and disease duration and set into context with clinical biomarkers such as CSF concentrations of alpha-synuclein, neurofilament light chain, and tau protein. Based on the different PD subgroups we performed a total of eight different comparisons. Highlights from these comparisons include increased citrate and dimethylglycine with a decrease of creatinine and methionine in healthy controls and early PD group compared to GBA, PD late and recessive PD. We furthermore identified decreased HDL-3 free cholesterol in genetic PD cases compared to sporadic subject samples (sum of the PD early and PD late groups). Considering medication, we found that the levodopa equivalent daily dose (LEDD) is mostly positively correlated with tyrosine and citrate in sporadic PD compared to pyruvate and phenylalanine in genetic PD. Cerebrospinal fluid levels of alpha-synuclein were negatively correlated with alanine. Further metabolites and lipoproteins with discriminatory power for double mutation PD cases involved ornithine, 2-aminobutyrate and 2-hydroxybutyrate as well as for mitochondrial phenotypes via LDL phospholipid, apolipoprotein and cholesterol subfractions. Quantitative IVDr NMR serum spectroscopy is able to stratify PD patient samples of different etiology and can contribute to a wider understanding of the underlying metabolism-driven alterations e.g. in energy, amino acid, and lipoprotein metabolism. Though our overall cohort was large, major confounders such as age, sex and medication have a strong impact. That is why absolute quantification and detailed patient knowledge about metabolic confounders, is a premise for future translation of NMR serum spectroscopy to routine PD diagnostics.
Collapse
Affiliation(s)
- Georgy Berezhnoy
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Gyuntae Bae
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Leonie Wüst
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Claudia Schulte
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Claire Cannet
- Bruker BioSpin GmbH & Co. KG (AIC Division), Ettlingen, Germany
| | - Isabel Wurster
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Milan Zimmermann
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Alexander Jäck
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Eike Jakob Spruth
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Neuropsychiatry and Laboratory of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julian Hellmann-Regen
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Neurosciences, Charité Universitätsmedizin Berlin, Berlin, Germany
- ECRC Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sandra Roeske
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Dominik Pürner
- Department of Neurology, School of Medicine, University Hospital München rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Clinic for Neurology, Medical Faculty, University Hospital Magdeburg, Magdeburg, Germany
| | - Fabian Maass
- Department of Neurology, University Medical Center, Georg August University, Göttingen, Germany
| | - Felix Hufschmidt
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn and University of Bonn, Bonn, Germany
| | - Ingo Kilimann
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Elisabeth Dinter
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Okka Kimmich
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Vascular Neurology, University Hospital Bonn, Bonn, Germany
| | - Anna Gamez
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, University Hospital of Munich, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
| | - Josef Priller
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Neuropsychiatry and Laboratory of Molecular Psychiatry, Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Psychiatry and Psychotherapy, School of Medicine and Health, Technical University of Munich, German Center for Mental Health (DZPG), Munich, Germany
- University of Edinburgh and UK DRI, Edinburgh, UK
| | - Oliver Peters
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin-Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Michael Wagner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn and University of Bonn, Bonn, Germany
| | - Alexander Storch
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Germany
- Department of Neurology, University Medical Centre, Rostock, Germany
| | - Paul Lingor
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Neurology, School of Medicine, University Hospital München rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany
- Institute of Cognitive Neuroscience, University College London, London, UK
| | - Christoph van Riesen
- Department of Neurology, University Medical Center, Georg August University, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
| | - Ullrich Wüllner
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn and University of Bonn, Bonn, Germany
| | - Stefan Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock-Greifswald, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Center, Rostock, Germany
| | - Björn Falkenburger
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Department of Neurology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center, Georg August University, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
| | - Inga Zerr
- Department of Neurology, University Medical Center, Georg August University, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Goettingen, Germany
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Vascular Neurology, University Hospital Bonn, Bonn, Germany
| | - Annika Spottke
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Patricia Rizzu
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | | - Hartmut Schäfer
- Bruker BioSpin GmbH & Co. KG (AIC Division), Ettlingen, Germany
| | - Thomas Gasser
- Hertie Institute for Clinical Brain Research, Department of Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
| | - Christoph Trautwein
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany.
- M3 Research Center for Malignome, Metabolome and Microbiome, Medical Faculty, University of Tübingen, Tübingen, Germany.
- Core Facility Metabolomics, Medical Faculty, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
4
|
Ramchandra J, Inca-Martinez M, Leal TP, Chaparro-Solano HM, Salim A, Gatto EM, Rojas NG, Da Prat G, Micheli F, Santos-Lobato BL, Cardoso FEC, Camargos S, Letro GH, Braga-Neto P, Peixoto VMT, Schuh AFS, Tumas V, Brito MM, Borges V, Candeias da Silva C, Ferraz HB, Chana-Cuevas P, Saffie-Awad P, Olguin P, Colombo A, de la Cerda A, Farías GA, Nuñez JC, Arboleda G, Arboleda H, Fernandez W, Arboleda-Bustos CE, Orozco JL, Muñoz-Ospina B, Velez-Pardo C, Jiménez-Del-Río M, Lopera F, Moreno S, Pineda DA, Buritica O, Torrealba-Acosta G, Medina Escobar A, Rodríguez-Violante M, Hernández-Medrano AJ, Martinez-Ramirez D, González-González M, Rentería ME, Alcauter S, Reyes-Pérez P, Medina-Rivera A, Vazquez-Guevara D, de María Ugalde-Mejía L, Valadez M MJ, Cárdenas-Sáenz O, Rodríguez-Leyva I, Guerra-Galicia CM, Gandarilla-Martínez NA, Matuk-Pérez Y, Morelos-Figaredo E, Salinas-Barboza K, Isais-Millán S, Pérez-Torres T, Deras Gaucin DC, Ruiz-Contreras AE, Estrada-Bellmann I, Rios-Pinto J, Cornejo-Olivas M, Cosentino C, Torres Ramirez L, Mori N, Mejía-Rojas K, Medina ÁC, Cornejo-Herrera I, Ochoa EM, Viñuela Á, Dieguez E, Amorín I, Lescano A, Mata IF. Sex differences in the diagnosis latency of Parkinson's disease in Latin America. Parkinsonism Relat Disord 2025; 134:107344. [PMID: 40058072 DOI: 10.1016/j.parkreldis.2025.107344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 01/19/2025] [Accepted: 02/20/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Age and sex are known risk factors for Parkinson's Disease (PD), but it remains controversial if there are sex differences in the diagnosis latency. The objective of this study was to examine these sex differences in Latin America. METHODS The Latin American Research Consortium on the Genetics of PD (LARGE-PD) includes PD patients from countries across Latin America who were diagnosed using the UK Brain Bank criteria. Ages at onset (AAO; N = 2,792), diagnosis (AAD; N = 1,416), and calculated diagnosis latency (N = 1,416) were extracted from the LARGE-PD database and compared for both males and females overall, by country, and decade-long age ranges. A cohort was created based on available data for motor sign at onset (N = 492). Regressions examining diagnosis latency as a factor of sex, country, and motor subtype were performed. Two-tailed t-tests at 95 % confidence intervals were used to identify differences in mean AAOs, AADs, and diagnosis latencies between the sexes. RESULTS Across the LARGE-PD cohort, lower AAD was observed in males. Per country, AAO was lower for males in Mexico and diagnosis latency was shorter for males in Chile. Overall, younger females (≤39) and older males (≥70) are likely to experience longer latencies. CONCLUSIONS Our results suggest that there may be country and age dependent sex differences in AAO, AAD, and diagnosis latency of PD in Latin America. Interestingly, the mean AAO of LARGE-PD is approximately 6 years younger than studies done with European populations. Analyses with additional data are needed to determine the influence of other factors.
Collapse
Affiliation(s)
- Janvi Ramchandra
- Genomic Medicine Institute, Cleveland Clinic Foundation Lerner Research Institute, Cleveland, USA; Department of Biochemistry, Case Western Reserve University, Cleveland, USA
| | - Miguel Inca-Martinez
- Genomic Medicine Institute, Cleveland Clinic Foundation Lerner Research Institute, Cleveland, USA
| | - Thiago Peixoto Leal
- Genomic Medicine Institute, Cleveland Clinic Foundation Lerner Research Institute, Cleveland, USA
| | - Henry Mauricio Chaparro-Solano
- Genomic Medicine Institute, Cleveland Clinic Foundation Lerner Research Institute, Cleveland, USA; Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Department of Molecular Medicine, Cleveland, USA
| | - Amira Salim
- Genomic Medicine Institute, Cleveland Clinic Foundation Lerner Research Institute, Cleveland, USA; Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Department of Molecular Medicine, Cleveland, USA
| | - Emilia M Gatto
- Department of Neurology, Affiliated University of Buenos Aires, Buenos Aires, Argentina
| | - Natalia González Rojas
- Department of Neurology, Institute of Neuroscience of Buenos Aires (INEBA), Sanatorio de la Trinidad Mitre, Buenos Aires University, Buenos Aires, Argentina
| | - Gustavo Da Prat
- Department of Neurology, Institute of Neuroscience of Buenos Aires (INEBA), Sanatorio de la Trinidad Mitre, Buenos Aires University, Buenos Aires, Argentina
| | - Federico Micheli
- Parkinson's Disease and Movement Disorders Center, University of Buenos Aires, Buenos Aires, Argentina; Centro de Parkinson y Movimientos Anormales, Fundación San Gabriel, Buenos Aires, Argentina
| | - Bruno Lopes Santos-Lobato
- Hospital Ophir Loyola, Belém, Brazil; Laboratório de Neuropatología Experimental, Universidade Federal do Pará, Belém, Brazil
| | - Francisco E C Cardoso
- Movement Disorders Unit, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sarah Camargos
- Movement Disorders Unit, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Grace H Letro
- Departamento de Neurología, Hospital de Pontifícia Católica de Campinas, Campinas, Brazil
| | - Pedro Braga-Neto
- Centro de Ciências da Saúde, Universidade Estadual do Ceará, Fortaleza, Brazil; Neurology Service, Hospital Universitário Walter Cantídio, Universidade Federal do Ceará, Fortaleza, Brazil
| | | | - Artur F S Schuh
- Departamento de Farmacologia, Universidade Federal do Rio Grande do Sul, São Paulo, Brazil; Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Vitor Tumas
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Riberão Preto, Brazil
| | - Manuelina M Brito
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Riberão Preto, Brazil
| | - Vanderci Borges
- Movement Disorders Unit, Department of Neurology and Neurosurgery, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Carolina Candeias da Silva
- Movement Disorders Unit, Department of Neurology and Neurosurgery, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Henrique B Ferraz
- Movement Disorders Unit, Department of Neurology and Neurosurgery, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Pedro Chana-Cuevas
- Centro de Trastornos del Movimiento, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago, Chile
| | - Paula Saffie-Awad
- Centro de Trastornos del Movimiento, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Santiago, Chile; Programa de Pós-Graduação em Ciências Médicas, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Chile Clínica Santa María, Santiago, Chile
| | - Patricio Olguin
- Human Genetics Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago, Chile; Department of Neuroscience, Biomedical Neuroscience Institute (BNI), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alicia Colombo
- Departament of Pathological Anatomy, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Pathological Anatomy Service, Hospital Clínico de la Universidad de Chile, Santiago, Chile
| | - Andres de la Cerda
- Clínica Dávila, Corporación de Investigación en Neurología, Santiago, Chile
| | - Gonzalo A Farías
- Facultad de Medicina, Universidad de Chile, Santiago, Chile; Departamento de Neurología y Neurocirugía Norte, Hospital Clínico Universidad de Chile, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Juan Cristobal Nuñez
- Departamento de Neurología y Neurocirugía Norte, Hospital Clínico Universidad de Chile, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Gonzalo Arboleda
- Department of Neurosciences, Universidad Nacional de Colombia, Facultad de Medicina, Bogotá, Colombia
| | - Humberto Arboleda
- Neurosciences and Cell Death Research Groups, Medical School and Genetic Institute, Universidad Nacional de Colombia, Facultad de Medicina, Bogotá, Colombia
| | - William Fernandez
- Department of Neurosciences, Universidad Nacional de Colombia, Facultad de Medicina, Bogotá, Colombia
| | - Carlos E Arboleda-Bustos
- Neurosciences and Cell Death Research Groups, Medical School and Genetic Institute, Universidad Nacional de Colombia, Facultad de Medicina, Bogotá, Colombia
| | - Jorge L Orozco
- Departamento de Neurología, Fundación Valle del Lili, Cali, Colombia; Universidad Icesi, Cali, Colombia
| | | | - Carlos Velez-Pardo
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Marlene Jiménez-Del-Río
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Francisco Lopera
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Sonia Moreno
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - David A Pineda
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Omar Buritica
- Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Gabriel Torrealba-Acosta
- Neurosciences Research Center, Universidad Nacional de Costa Rica, San José, Costa Rica; Department of Neurology, Duke University Medical Center, Durham, USA
| | - Alex Medina Escobar
- Department of Neurology, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Mayela Rodríguez-Violante
- Movement Disorders Clinic, Instituto Nacional de Neurología y Neurocirugía (INNN), Mexico City, Mexico
| | - Ana Jimena Hernández-Medrano
- Clinical Laboratory of Neurodegenerative Diseases, Instituto Nacional de Neurología y Neurocirugía (INNN), Mexico City, Mexico
| | | | | | - Miguel E Rentería
- Mental Health & Neuroscience Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Sarael Alcauter
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Paula Reyes-Pérez
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Alejandra Medina-Rivera
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | | | | | | | | | - Ildefonso Rodríguez-Leyva
- Facultad de Medicina de la Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico; Hospital Central "Dr Igacio Morones Prieto", San Luis Potosí, Mexico
| | | | | | - Yamil Matuk-Pérez
- Universidad Autónoma de Querétaro, Querétaro, Mexico; Unidad de Neurociencias y Medicina Interna, Hospital Angeles Centro Sur, Querétaro, Mexico
| | | | | | | | - Teresa Pérez-Torres
- Neurólogo Guadalajara - Neuromuscular. Dra. Teresa Pérez Torres, Guadalajara, Mexico
| | - Diana C Deras Gaucin
- Departamento de Neurología, Hospital Medica de la Ciudad Mazatlán, Mazatlán, Mexico
| | | | - Ingrid Estrada-Bellmann
- Movement Disorders Clinic, Neurology Division, Internal Medicine Department, University Hospital "Dr. José E. González", Universidad Autónoma de Nuevo León, Monterrey, Mexico
| | - Julia Rios-Pinto
- Universidad Peruana Los Andes, Huancayo, Peru; Neurogenetics Research Center, Instituto Nacional de Ciencias Neurológicas, Lima, Peru
| | - Mario Cornejo-Olivas
- Neurogenetics Research Center, Instituto Nacional de Ciencias Neurológicas, Lima, Peru; Neurogenetics Working Group, Universidad Científica del Sur, Lima, Peru
| | - Carlos Cosentino
- Movement Disorders Unit, Instituto Nacional de Ciencias Neurológicas, Lima, Peru; School of Medicine, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Luis Torres Ramirez
- Movement Disorders Unit, Instituto Nacional de Ciencias Neurológicas, Lima, Peru
| | - Nicanor Mori
- Neurology Department, Hospital Nacional Daniel Alcides Carrion, Bellavista, Peru
| | - Koni Mejía-Rojas
- Neurology Department, Hospital Nacional Daniel Alcides Carrion, Bellavista, Peru; Educación Médica Continua (EDMECON), Lima, Peru
| | | | | | | | - Ángel Viñuela
- Instituto de Neurociencias, Manatí Medical Center, Manatí, Puerto Rico
| | - Elena Dieguez
- Neurology Institute, Universidad de la República, Montevideo, Uruguay
| | - Ignacio Amorín
- Neurology Institute, Universidad de la República, Montevideo, Uruguay
| | - Andrés Lescano
- Neurology Institute, Universidad de la República, Montevideo, Uruguay
| | - Ignacio F Mata
- Genomic Medicine Institute, Cleveland Clinic Foundation Lerner Research Institute, Cleveland, USA; Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Department of Molecular Medicine, Cleveland, USA.
| |
Collapse
|
5
|
Soto Linan V, Rioux V, Peralta M, Dupré N, Hébert M, Lévesque M. Early detection of Parkinson's disease: Retinal functional impairments as potential biomarkers. Neurobiol Dis 2025; 208:106872. [PMID: 40090470 DOI: 10.1016/j.nbd.2025.106872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/09/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025] Open
Abstract
BACKGROUND Parkinson's disease is typically diagnosed after substantial neurodegeneration despite early non-motor symptoms manifesting decades earlier. These changes offer a promising avenue for diagnostic exploration, especially within the eye, which has been proposed as a "window to the brain." OBJECTIVE The aim was to identify biomarkers by validating the use of electroretinography, a non-invasive technique, to detect early retinal function anomalies reflecting central dysfunction. METHODS Homozygous M83 transgenic mice (n = 10 males, 11 females), overexpressing human A53T α-synuclein, underwent behavioral tests and electroretinography measurements. Histological evaluation was performed at four months to analyze synucleinopathies and neurodegeneration. Electroretinography was also conducted with idiopathic PD patients (mean age 63.35 ± 7.73; disease duration 4.15 ± 2.06; H&Y score 2.07 ± 0.59; n = 12 males, 8 females) and healthy age-matched controls (mean age 61.65 ± 8.39; n = 9 males, 11 females). RESULTS Rodent electroretinography revealed reduced photopic b-wave, PhNR b-wave, and PhNR-wave amplitudes at two and four months, particularly in females, indicating bipolar and retinal ganglion cell impairment. Based on retinal histological assessment, these changes might arise from α-synuclein pathology occurring in outer retinal layers. Likewise, the scotopic b-wave and PhNR waveform were similarly impaired in female participants with Parkinson's disease. The scotopic oscillatory potentials isolated further identified an attenuated amacrine cell output in females. CONCLUSIONS Findings from both mice and human cohorts indicate that retinal functional impairments can be detected early in the progression of Parkinson's disease, particularly among females. These tools show promise in facilitating early diagnosis, disease monitoring, therapeutic intervention, and ultimately enhancing patient outcomes.
Collapse
Affiliation(s)
- Victoria Soto Linan
- Department of Psychiatry and Neurosciences, Université Laval, 1050 Av. de la Médecine, Québec G1V 0A6, Québec, Canada; Integrative Neuroscience and Experimental Therapies Axis, CERVO Brain Research Center, 2301 Av. D'Estimauville, Québec G1E 1T2, Québec, Canada.
| | - Véronique Rioux
- Integrative Neuroscience and Experimental Therapies Axis, CERVO Brain Research Center, 2301 Av. D'Estimauville, Québec G1E 1T2, Québec, Canada.
| | - Modesto Peralta
- Integrative Neuroscience and Experimental Therapies Axis, CERVO Brain Research Center, 2301 Av. D'Estimauville, Québec G1E 1T2, Québec, Canada.
| | - Nicolas Dupré
- Neuroscience Axis, CHU de Québec-Université Laval, 2705 Bd Laurier, Québec G1V 4G2, Québec, Canada; Department of Medicine, Faculty of Medicine, Université Laval, 1050 Av. de la Médecine, Québec G1V 0A6, Québec, Canada.
| | - Marc Hébert
- Department of Ophthalmology and Otorhinolaryngology, Université Laval, 1050 Av. de la Médecine, Québec G1V 0A6, Québec, Canada; Clinical and Cognitive Neuroscience Axis, CERVO Brain Research Center, 2301 Av. D'Estimauville, Québec G1E 1T2, Québec, Canada.
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Université Laval, 1050 Av. de la Médecine, Québec G1V 0A6, Québec, Canada; Integrative Neuroscience and Experimental Therapies Axis, CERVO Brain Research Center, 2301 Av. D'Estimauville, Québec G1E 1T2, Québec, Canada.
| |
Collapse
|
6
|
Bentall L, Parr‐Brownlie L. Sexual Dimorphism in Levodopa-Induced Dyskinesia Following Parkinson's Disease: Uncharted Territory. Eur J Neurosci 2025; 61:e70144. [PMID: 40360439 PMCID: PMC12075048 DOI: 10.1111/ejn.70144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 04/12/2025] [Accepted: 05/01/2025] [Indexed: 05/15/2025]
Abstract
Sexual dimorphism is well-documented in Parkinson's disease (PD); however, when it comes to levodopa-induced dyskinesia (LID), epidemiological and clinical findings are scarce. This is an oversight because recent studies show significant correlations between LID risk and female sex. Estrogen strongly impacts neuronal function, affecting cognitive tasks such as movement, object recognition, and reward. In movement pathways, estrogen increases dopamine synthesis, transmission, and regulation, resulting in neuroprotection for PD in women. However, following menopause, PD prevalence, symptom severity, and LID risk increase for women. Consequently, early to mid-life estrogen state is neuroprotective, but later in life becomes a risk factor for PD and LID. This review explores estrogen's action in the brain, specifically within the dopamine system. Sexual dimorphism is described for the prevalence and onset of PD and LID. We examine the cellular basis of estrogen's role in sexual dimorphism and integrate these ideas to hypothesize why the risk for LID is higher for women, than men, with PD. Lastly, this review proposes that women with PD need their symptoms to be considered and managed differently to males. Treatment of women with PD should be based on their menopausal stage, as estrogen may be masking, exacerbating, or complicating symptoms. Importantly, we present these concepts to stimulate discussion among clinical and bench scientists so that key experiments can be conducted to examine the mechanisms underlying LID, so they can be prevented to improve the quality of life for women and men living with PD in the future.
Collapse
|
7
|
Liu Y, Zhang P, Li H, Zhou L, Jiang J, Jiang Y, Ai K, Liu G, Zhang J. Sex-specific brain morphological and network differences in patients showing Parkinson's disease with and without possible rapid eye movement sleep behavior disorder. Front Neurol 2025; 16:1561555. [PMID: 40330249 PMCID: PMC12053292 DOI: 10.3389/fneur.2025.1561555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/17/2025] [Indexed: 05/08/2025] Open
Abstract
Background Sex is a crucial determinant in the clinical manifestations of diseases. However, previous studies have not clarified whether altered brain morphology shows sex-specific patterns in patients with Parkinson's disease (PD) with or without possible rapid eye movement sleep behavior disorder (RBD). This study aimed to investigate sex-specific differences in the patterns of morphological changes among different subgroups of PD. Methods High-resolution T1-weighted magnetic resonance imaging and clinical scale data were collected from 278 participants in the Parkinson's disease Progression Marker Initiative database: 93 patients with PD-pRBD (60 males, 33 females), 114 patients showing PD without RBD (PDnon-pRBD group; 68 males, 46 females), and 71 healthy controls (HCs; 44 males, 17 females). The Computational Anatomy Toolbox (CAT) 12 was utilized to collect data on gray matter volume (GMV) and cortical morphological metrics. Subsequently, individual-level morphological similarity networks were constructed on the basis of these cortical metrics. Finally, the topological properties of the network were analyzed using graph theoretical methods. Results In the PD-pRBD group, the GMV in the frontal and temporal lobes of males was lower than that of females. In contrast, the gyrification index (GI) of the frontal lobe in males was lower than that in females within the PDnon-pRBD group. Network analyses based on graph theory revealed that male PD-pRBD patients showed lower network information integration than female patients, particularly in terms of the global properties of fractal dimension (FD) networks. Moreover, in the PD-pRBD group, male patients showed a strong correlation between morphological network metrics and cognitive performance, as measured by the Hopkins Verbal Learning Test-Revised (HVLT-R) memory scores. Conclusion The presence of more significant sex-related differences in brain morphological changes in the PD-pRBD group in comparison with the PDnon-pRBD group highlights the importance of considering sex-related differences in the diagnosis and management of patients with PD-pRBD.
Collapse
Affiliation(s)
- Yang Liu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, China
- Gansu Medical MRI Equipment Application Industry Technology Center, Lanzhou, China
| | - Pengfei Zhang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, China
- Gansu Medical MRI Equipment Application Industry Technology Center, Lanzhou, China
| | - Hao Li
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, China
- Gansu Medical MRI Equipment Application Industry Technology Center, Lanzhou, China
| | - Liang Zhou
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, China
- Gansu Medical MRI Equipment Application Industry Technology Center, Lanzhou, China
| | - Jingqi Jiang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, China
- Gansu Medical MRI Equipment Application Industry Technology Center, Lanzhou, China
| | - Yanli Jiang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, China
- Gansu Medical MRI Equipment Application Industry Technology Center, Lanzhou, China
| | - Kai Ai
- Department of Clinical and Technical Support, Philips Healthcare, Xi'an, China
| | - Guangyao Liu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, China
- Gansu Medical MRI Equipment Application Industry Technology Center, Lanzhou, China
| | - Jing Zhang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou, China
- Gansu Medical MRI Equipment Application Industry Technology Center, Lanzhou, China
| |
Collapse
|
8
|
Kritzer MF, Adler A, Locklear M. Androgen effects on mesoprefrontal dopamine systems in the adult male brain. Neuroscience 2025; 568:519-534. [PMID: 38977069 DOI: 10.1016/j.neuroscience.2024.07.001] [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: 04/22/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
Epidemiological data show that males are more often and/or more severely affected by symptoms of prefrontal cortical dysfunction in schizophrenia, Parkinson's disease and other disorders in which dopamine circuits associated with the prefrontal cortex are dysregulated. This review focuses on research showing that these dopamine circuits are powerfully regulated by androgens. It begins with a brief overview of the sex differences that distinguish prefrontal function in health and prefrontal dysfunction or decline in aging and/or neuropsychiatric disease. This review article then spotlights data from human subjects and animal models that specifically identify androgens as potent modulators of prefrontal cortical operations and of closely related, functionally critical measures of prefrontal dopamine level or tone. Candidate mechanisms by which androgens dynamically control mesoprefrontal dopamine systems and impact prefrontal states of hypo- and hyper-dopaminergia in aging and disease are then considered. This is followed by discussion of a working model that identifies a key locus for androgen modulation of mesoprefrontal dopamine systems as residing within the prefrontal cortex itself. The last sections of this review critically consider the ways in which the organization and regulation of mesoprefrontal dopamine circuits differ in the adult male and female brain, and highlights gaps where more research is needed.
Collapse
Affiliation(s)
- Mary F Kritzer
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY 11794-5230, United States.
| | - Alexander Adler
- Department of Oncology and Immuno-Oncology, Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, United States
| | | |
Collapse
|
9
|
Li Q, Han X, Dong M, Bai L, Zhang W, Liu W, Wang F, Zhu X. FDA-Approved Secukinumab Alleviates Glial Activation and Immune Cell Infiltration in MPTP-Induced Mouse Model of Parkinson's Disease. Inflammation 2025:10.1007/s10753-025-02267-8. [PMID: 40011292 DOI: 10.1007/s10753-025-02267-8] [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: 10/23/2024] [Revised: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 02/28/2025]
Abstract
Interleukin-17A (IL-17A) has been implicated in the progression of neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD). However, the effect of the FDA-approved Secukinumab (SEC), an IL-17A inhibitor, on PD remains unclear. This study aimed to investigate the neuroprotective effect of SEC and its potential mechanisms in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD. Male C57BL/6 J mice were mainly assigned to three groups: Sham, MPTP, and MPTP + SEC. Motor coordination was assessed using the climbing rod and rotarod tests. Dopaminergic neurons (TH +) and glial cells (Iba-1 + , GFAP +) in the substantia nigra were evaluated using immunohistochemistry and immunofluorescence. Flow cytometry was used to analyze immune cell populations in the brain and spleen. Inflammatory cytokines and chemokines were quantified using RT-PCR. SEC treatment significantly alleviated the loss of dopaminergic neurons and improved motor coordination in MPTP mice. It also reduced the infiltration of peripheral immune cells, including CD4 + T cells, NK cells, and monocyte-macrophages into the brain. SEC attenuated glial activation (Iba-1 + , GFAP +) and decreased the expression of pro-inflammatory cytokines and chemokines (CCL2, CXCL9), which recruit immune cells into the brain. These results suggest that Secukinumab protects dopaminergic neurons and attenuates neuroinflammation in MPTP-induced model. SEC treatment in PD might be an effective therapeutic approach for clinical application in the future. HIGHLIGHTS: • Secukinumab reduces the loss of dopaminergic neurons and axons in MPTP mice. • Secukinumab inhibits the infiltration of peripheral immune cells into the brain in MPTP mice. • Secukinumab inhibits the activation of glial cells and reduces neuroinflammation in MPTP mice.
Collapse
Affiliation(s)
- Qi Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Xiaoxuan Han
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Mengmeng Dong
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Lipeng Bai
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Wei Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Wei Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Fei Wang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China
| | - Xiaodong Zhu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Anshan Road 154#, Tianjin, 300052, China.
| |
Collapse
|
10
|
Liu X, Zhao Y, Feng Y, Wang S, Luo A, Zhang J. Ovarian Aging: The Silent Catalyst of Age-Related Disorders in Female Body. Aging Dis 2025:AD.2024.1468. [PMID: 39965250 DOI: 10.14336/ad.2024.1468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025] Open
Abstract
Age-related diseases have emerged as a global concern as the population ages. Consequently, understanding the underlying causes of aging and exploring potential anti-aging interventions is imperative. In females, the ovaries serve as the principal organs responsible for ovulation and the production of female hormones. The aging ovaries are related to infertility, menopause, and associated menopausal syndromes, with menopause representing the culmination of ovarian aging. Current evidence indicates that ovarian aging may contribute to dysfunction across multiple organ systems, including, but not limited to, cognitive impairment, osteoporosis, and cardiovascular disease. Nevertheless, due to the widespread distribution of sex hormone receptors throughout the body, ovarian aging affects not only these specific organs but also influences a broader spectrum of age-related diseases in women. Despite this, the impact of ovarian aging on overall age-related diseases has been largely neglected. This review provides a thorough summary of the impact of ovarian aging on age-related diseases, encompassing the nervous, circulatory, locomotor, urinary, digestive, respiratory, and endocrine systems. Additionally, we have outlined prospective therapeutic approaches for addressing both ovarian aging and age-related diseases, with the aim of mitigating their impacts and preserving women's fertility, physical health, and psychological well-being.
Collapse
Affiliation(s)
- Xingyu Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuanqu Zhao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yanzhi Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Aiyue Luo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
11
|
Chen L, Fang MJ, Yu XE, Xu Y. Genetic analyses identify brain functional networks associated with the risk of Parkinson's disease and drug-induced parkinsonism. Cereb Cortex 2025; 35:bhae506. [PMID: 39820363 DOI: 10.1093/cercor/bhae506] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/01/2024] [Accepted: 12/31/2024] [Indexed: 01/19/2025] Open
Abstract
Brain functional networks are associated with parkinsonism in observational studies. However, the causal effects between brain functional networks and parkinsonism remain unclear. We aimed to assess the potential bidirectional causal associations between 191 brain resting-state functional magnetic resonance imaging (rsfMRI) phenotypes and parkinsonism including Parkinson's disease (PD) and drug-induced parkinsonism (DIP). We used Mendelian randomization (MR) to assess the bidirectional associations between brain rsfMRI phenotypes and parkinsonism, followed by several sensitivity analyses for robustness validation. In the forward MR analyses, we found that three rsfMRI phenotypes genetically determined the risk of parkinsonism. The connectivity in the visual network decreased the risk of PD (OR = 0.391, 95% CI = 0.235 ~ 0.649, P = 2.83 × 10-4, P_FDR = 0.039). The connectivity of salience and motor networks increased the risk of DIP (OR = 4.102, 95% CI = 1.903 ~ 8.845, P = 3.17 × 10-4, P_FDR = 0.044). The connectivity of limbic and default mode networks increased the risk of DIP (OR = 14.526, 95% CI = 3.130 ~ 67.408, P = 6.32 × 10-4, P_FDR = 0.0437). The reverse MR analysis indicated that PD and DIP had no effect on brain rsfMRI phenotypes. Our findings reveal causal relationships between brain functional networks and parkinsonism, providing important interventional and therapeutic targets for different parkinsonism.
Collapse
Affiliation(s)
- Lin Chen
- Institute of Neurology, Anhui University of Chinese Medicine, No. 357 Changjiang Middle Road, Luyang District, Hefei 230061, China
- Anhui University of Chinese Medicine, No. 350, Longzihu Road, Xinzhan District, Hefei 230012, China
| | - Ming-Juan Fang
- Anhui University of Chinese Medicine, No. 350, Longzihu Road, Xinzhan District, Hefei 230012, China
| | - Xu-En Yu
- Institute of Neurology, Anhui University of Chinese Medicine, No. 357 Changjiang Middle Road, Luyang District, Hefei 230061, China
- Anhui University of Chinese Medicine, No. 350, Longzihu Road, Xinzhan District, Hefei 230012, China
| | - Yin Xu
- Institute of Neurology, Anhui University of Chinese Medicine, No. 357 Changjiang Middle Road, Luyang District, Hefei 230061, China
- Anhui University of Chinese Medicine, No. 350, Longzihu Road, Xinzhan District, Hefei 230012, China
| |
Collapse
|
12
|
Kumar D, Bishnoi M, Kondepudi KK, Sharma SS. Gut Microbiota-Based Interventions for Parkinson's Disease: Neuroprotective Mechanisms and Current Perspective. Probiotics Antimicrob Proteins 2025:10.1007/s12602-024-10433-x. [PMID: 39809955 DOI: 10.1007/s12602-024-10433-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 01/16/2025]
Abstract
Recent evidence links gut microbiota alterations to neurodegenerative disorders, including Parkinson's disease (PD). Replenishing the abnormal composition of gut microbiota through gut microbiota-based interventions "prebiotics, probiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT)" has shown beneficial effects in PD. These interventions increase gut metabolites like short-chain fatty acids (SCFAs) and glucagon-like peptide-1 (GLP-1), which may protect dopaminergic neurons via the gut-brain axis. Neuroprotective effects of these interventions are mediated by several mechanisms, including the enhancement of neurotrophin and activation of the PI3K/AKT/mTOR signaling pathway, GLP-1-mediated gut-brain axis signaling, Nrf2/ARE pathway, and autophagy. Other pathways, such as free fatty acid receptor activation, synaptic plasticity improvement, and blood-brain and gut barrier integrity maintenance, also contribute to neuroprotection. Furthermore, the inhibition of the TLR4/NF-кB pathway, MAPK pathway, GSK-3β signaling pathway, miR-155-5p-mediated neuroinflammation, and ferroptosis could account for their protective effects. Clinical studies involving gut microbiota-based interventions have shown therapeutic benefits in PD patients, particularly in improving gastrointestinal dysfunction and some neurological symptoms. However, the effectiveness in alleviating motor symptoms remains mild. Large-scale clinical trials are still needed to confirm these findings. This review emphasizes the neuroprotective mechanisms of gut microbiota-based interventions in PD as supported by both preclinical and clinical studies.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Mahendra Bishnoi
- Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biomanufacturing Institute (NABI), Knowledge City-Sector 81, S.A.S. Nagar, Punjab, 140306, India
| | - Kanthi Kiran Kondepudi
- Centre for Excellence in Functional Foods, Division of Food and Nutritional Biotechnology, National Agri-Food Biomanufacturing Institute (NABI), Knowledge City-Sector 81, S.A.S. Nagar, Punjab, 140306, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
13
|
Weresh H, Hermann K, Al-Salahat A, Noor A, Billion T, Chen YT, Tauseef A, Abdul Jabbar AB. Trends and Disparities in Parkinson's Disease Mortality in the United States with Predictions Using Machine Learning. NEUROSCI 2025; 6:6. [PMID: 39846565 PMCID: PMC11755521 DOI: 10.3390/neurosci6010006] [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: 12/18/2024] [Revised: 01/05/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative condition characterized by the degradation of dopaminergic pathways in the brain. As the population in the United States continues to age, it is essential to understand the trends in mortality related to PD. This analysis of PD's mortality characterizes temporal shifts, examines demographic and regional differences, and provides machine-learning predictions. METHODS PD-related deaths in the United States were gathered from CDC WONDER. Age-adjusted mortality rates (AAMR) were collected, and trends were analyzed based on gender, race, region, age, and place of death. Annual percent change and average annual percent change were calculated using Joinpoint Regression program. Forecasts were obtained using the optimal Autoregressive Integrated Moving Average (ARIMA) model. RESULTS Overall mortality rate due to Parkinson's increased from 1999 to 2022. Male gender, White race, Southern region, and older ages were associated with higher mortality compared to other groups. Deaths at home decreased and hospice deaths increased during the study period. CONCLUSIONS This study highlights the increasing rate of PD AAMR and how it may become even more prevalent with time, emphasizing the value of increasing knowledge surrounding the disease and its trends to better prepare health systems and individual families for the burden of PD.
Collapse
Affiliation(s)
- Henry Weresh
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (H.W.); (K.H.); (T.B.)
| | - Kallin Hermann
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (H.W.); (K.H.); (T.B.)
| | - Ali Al-Salahat
- Neurology Department, Creighton University, Omaha, NE 68178, USA;
| | - Amna Noor
- Services Hospital, Lahore 40050, Pakistan;
| | - Taylor Billion
- School of Medicine, Creighton University, Omaha, NE 68178, USA; (H.W.); (K.H.); (T.B.)
| | - Yu-Ting Chen
- Neurology Department, Creighton University, Omaha, NE 68178, USA;
| | - Abubakar Tauseef
- Department of Medicine, Creighton University, Omaha, NE 68178, USA; (A.T.); (A.B.A.J.)
| | - Ali Bin Abdul Jabbar
- Department of Medicine, Creighton University, Omaha, NE 68178, USA; (A.T.); (A.B.A.J.)
| |
Collapse
|
14
|
Petit P, Berger F, Bonneterre V, Vuillerme N. Investigating Parkinson's disease risk across farming activities using data mining and large-scale administrative health data. NPJ Parkinsons Dis 2025; 11:13. [PMID: 39779703 PMCID: PMC11711245 DOI: 10.1038/s41531-024-00864-2] [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: 07/02/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
The risk of Parkinson's disease (PD) associated with farming has received considerable attention, in particular for pesticide exposure. However, data on PD risk associated with specific farming activities is lacking. We aimed to explore whether specific farming activities exhibited a higher risk of PD than others among the entire French farm manager (FM) population. A secondary analysis of real-world administrative insurance claim data and electronic health/medical records (TRACTOR project) was conducted to estimate PD risk for 26 farming activities using data mining. PD cases were identified through chronic disease declarations and antiparkinsonian drug claims. There were 8845 PD cases among 1,088,561 FMs. The highest-risk group included FMs engaged in pig farming, cattle farming, truck farming, fruit arboriculture, and crop farming, with mean hazard ratios (HRs) ranging from 1.22 to 1.67. The lowest-risk group included all activities involving horses and small animals, as well as gardening, landscaping and reforestation companies (mean HRs: 0.48-0.81). Our findings represent a preliminary work that suggests the potential involvement of occupational risk factors related to farming in PD onset and development. Future research focusing on farmers engaged in high-risk farming activities will allow to uncover potential occupational factors by better characterizing the farming exposome, which could improve PD surveillance among farmers.
Collapse
Affiliation(s)
- Pascal Petit
- Univ. Grenoble Alpes, AGEIS, 38000, Grenoble, France.
| | - François Berger
- Univ. Grenoble Alpes, INSERM, Unit 1205, Braintech Lab, 38000, Grenoble, France
| | - Vincent Bonneterre
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000, Grenoble, France
- CHU Grenoble Alpes, Centre Régional de Pathologies Professionnelles et Environnementales, 38000, Grenoble, France
| | - Nicolas Vuillerme
- Univ. Grenoble Alpes, AGEIS, 38000, Grenoble, France
- Institut Universitaire de France, 75000, Paris, France
| |
Collapse
|
15
|
Belbellaj W, Lona-Durazo F, Bodano C, Busseuil D, Cyr MC, Fiorillo E, Mulas A, Provost S, Steri M, Tanaka T, Vanderwerff B, Wang J, Byrne RP, Cucca F, Dubé MP, Ferrucci L, McLaughlin RL, Tardif JC, Zawistowski M, Gagliano Taliun SA. The role of genetically predicted serum iron levels on neurodegenerative and cardiovascular traits. Sci Rep 2024; 14:24588. [PMID: 39427026 PMCID: PMC11490554 DOI: 10.1038/s41598-024-76245-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024] Open
Abstract
Iron is an essential mineral that supports numerous biological functions. Studies have reported associations between iron dysregulation and certain cardiovascular and neurodegenerative diseases, but the direction of influence is not clear. Our goal was to use computational approaches to better understand the role of genetically predicted iron levels on disease risk. We meta-analyzed genome-wide association study summary statistics for serum iron levels from two cohorts and two previous meta-analyses. We then obtained summary statistics from 11 neurodegenerative, cerebrovascular, cardiovascular or lipid traits to assess global and regional genetic correlation between iron levels and these traits. We used two-sample Mendelian randomization (MR) to estimate causal effects. Sex-stratified analyses were also carried out to identify effects potentially differing by sex. Overall, we identified three significant global correlations between iron levels and (i) coronary heart disease, (ii) triglycerides, and (iii) high-density lipoprotein (HDL) cholesterol levels. A total of 194 genomic regions had significant (after correction for multiple testing) local correlations between iron levels and the 11 tested traits. MR analysis revealed two potential causal relationships, between genetically predicted iron levels and (i) total cholesterol or (ii) non-HDL cholesterol. Sex-stratified analyses suggested a potential protective effect of iron levels on Parkinson's disease risk in females, but not in males. Our results will contribute to a better understanding of the genetic basis underlying iron in cardiovascular and neurological health in aging, and to the eventual identification of new preventive interventions or therapeutic avenues for diseases which affect women and men worldwide.
Collapse
Affiliation(s)
- Wiame Belbellaj
- Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
| | - Frida Lona-Durazo
- Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
| | - Cinzia Bodano
- Institute for Genetic and Biomedical Research, National Research Council (CNR), 09042, Monserrato-Cagliari, Italy
| | - David Busseuil
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
| | - Marie-Christyne Cyr
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
- Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, QC, H1T 1C8, Canada
| | - Edoardo Fiorillo
- Institute for Genetic and Biomedical Research, National Research Council (CNR), 08045, Lanusei, Italy
| | - Antonella Mulas
- Institute for Genetic and Biomedical Research, National Research Council (CNR), 08045, Lanusei, Italy
| | - Sylvie Provost
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
- Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, QC, H1T 1C8, Canada
| | - Maristella Steri
- Institute for Genetic and Biomedical Research, National Research Council (CNR), 09042, Monserrato-Cagliari, Italy
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institutes on Aging, Baltimore, MD, USA
| | - Brett Vanderwerff
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jiongming Wang
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ross P Byrne
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, D02 DK07, Republic of Ireland
| | - Francesco Cucca
- Department of Biomedical Sciences, University of Sassari, 07100, Sassari, Italy
| | - Marie-Pierre Dubé
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
- Université de Montréal Beaulieu-Saucier Pharmacogenomics Centre, Montreal, QC, H1T 1C8, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institutes on Aging, Baltimore, MD, USA
| | - Russell L McLaughlin
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, D02 DK07, Republic of Ireland
| | - Jean-Claude Tardif
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Matthew Zawistowski
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sarah A Gagliano Taliun
- Research Centre, Montreal Heart Institute, 5000 Bélanger Street, Montreal, QC, H1T 1C8, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
| |
Collapse
|
16
|
Kochmanski J, Virani M, Kuhn NC, Boyd SL, Becker K, Adams M, Bernstein AI. Developmental origins of Parkinson's disease risk: perinatal exposure to the organochlorine pesticide dieldrin leads to sex-specific DNA modifications in critical neurodevelopmental pathways in the mouse midbrain. Toxicol Sci 2024; 201:263-281. [PMID: 38995845 PMCID: PMC11424889 DOI: 10.1093/toxsci/kfae091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024] Open
Abstract
Epidemiological studies show that exposure to the organochlorine pesticide dieldrin is associated with an increased risk of Parkinson's disease (PD). Animal studies support a link between developmental dieldrin exposure and increased neuronal susceptibility in the α-synuclein preformed fibril and MPTP models in adult male C57BL/6 mice. In a previous study, we showed that developmental dieldrin exposure was associated with sex-specific changes in DNA modifications within genes related to dopaminergic neuron development and maintenance at 12 wk of age. Here, we used capture hybridization-sequencing with custom baits to interrogate DNA modifications across the entire genetic loci of the previously identified genes at multiple time points-birth, 6, 12, and 36 wk old. We identified largely sex-specific dieldrin-induced changes in DNA modifications at each time point that annotated to pathways important for neurodevelopment, potentially related to critical steps in early neurodevelopment, dopaminergic neuron differentiation, synaptogenesis, synaptic plasticity, and glial-neuron interactions. Despite large numbers of age-specific DNA modifications, longitudinal analysis identified a small number of differential modification of cytosines with dieldrin-induced deflection of epigenetic aging. The sex-specificity of these results adds to evidence that sex-specific responses to PD-related exposures may underly sex-specific differences in disease. Overall, these data support the idea that developmental dieldrin exposure leads to changes in epigenetic patterns that persist after the exposure period and disrupt critical neurodevelopmental pathways, thereby impacting risk of late-life diseases, including PD.
Collapse
Affiliation(s)
- Joseph Kochmanski
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, United States
| | - Mahek Virani
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| | - Nathan C Kuhn
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, United States
| | - Sierra L Boyd
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, United States
| | - Katelyn Becker
- Genomics Core, Van Andel Research Institute, Grand Rapids, MI 49503, United States
| | - Marie Adams
- Genomics Core, Van Andel Research Institute, Grand Rapids, MI 49503, United States
| | - Alison I Bernstein
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, United States
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
- Environmental and Occupational Health Sciences Institute, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, United States
| |
Collapse
|
17
|
Bourque M, Morissette M, Isenbrandt A, Giatti S, Melcangi RC, Carta M, Frau R, Bortolato M, Soulet D, Di Paolo T. Effect of 5-alpha reductase inhibitors in animal models of Parkinson's disease. Front Neuroendocrinol 2024; 75:101156. [PMID: 39353534 DOI: 10.1016/j.yfrne.2024.101156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/04/2024]
Abstract
Parkinson's disease (PD) is characterized by motor symptoms due to loss of brain dopamine and non-motor symptoms, including gastrointestinal disorders. Although there is no cure for PD, symptomatic treatments are available. L-Dopa is the gold standard PD therapy, but most patients develop dyskinesias (LID), which are challenging to manage. Amantadine is recognized as the most effective drug for LID, but its adverse effects limit the use in patients. Here we review how 5α-reductase inhibitors (5ARIs), drugs used to treat benign prostatic hyperplasia and alopecia, exhibit beneficial effects in PD animal models. 5ARIs show neuroprotective properties in brain and gut dopaminergic systems, and reduce dyskinesias in rodent model of PD. Additionally, the 5ARI finasteride dampened dopaminergic-induced drug gambling in PD patients. Neuroprotection and antidyskinetic activities of 5ARIs in animal models of PD suggest their potential repurposing in men with PD to address gut dysfunction, protect brain DA and inhibit dyskinesias.
Collapse
Affiliation(s)
- Mélanie Bourque
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Marc Morissette
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Amandine Isenbrandt
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada
| | - Silvia Giatti
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Roberto Cosimo Melcangi
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Manolo Carta
- Department of Biomedical Sciences, "Guy Everett Laboratory", University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, "Guy Everett Laboratory", University of Cagliari, Cittadella Universitaria SP 8, Monserrato 09042, Italy
| | - Marco Bortolato
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, 30 S 2000 E, Salt Lake City, UT 84112, USA
| | - Denis Soulet
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada
| | - Thérèse Di Paolo
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Pharmacy, Laval University, Quebec, QC, Canada.
| |
Collapse
|
18
|
Loo RTJ, Soudy M, Nasta F, Macchi M, Glaab E. Bioinformatics approaches for studying molecular sex differences in complex diseases. Brief Bioinform 2024; 25:bbae499. [PMID: 39397573 PMCID: PMC11471957 DOI: 10.1093/bib/bbae499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/09/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024] Open
Abstract
Many complex diseases exhibit pronounced sex differences that can affect both the initial risk of developing the disease, as well as clinical disease symptoms, molecular manifestations, disease progression, and the risk of developing comorbidities. Despite this, computational studies of molecular data for complex diseases often treat sex as a confounding variable, aiming to filter out sex-specific effects rather than attempting to interpret them. A more systematic, in-depth exploration of sex-specific disease mechanisms could significantly improve our understanding of pathological and protective processes with sex-dependent profiles. This survey discusses dedicated bioinformatics approaches for the study of molecular sex differences in complex diseases. It highlights that, beyond classical statistical methods, approaches are needed that integrate prior knowledge of relevant hormone signaling interactions, gene regulatory networks, and sex linkage of genes to provide a mechanistic interpretation of sex-dependent alterations in disease. The review examines and compares the advantages, pitfalls and limitations of various conventional statistical and systems-level mechanistic analyses for this purpose, including tailored pathway and network analysis techniques. Overall, this survey highlights the potential of specialized bioinformatics techniques to systematically investigate molecular sex differences in complex diseases, to inform biomarker signature modeling, and to guide more personalized treatment approaches.
Collapse
Affiliation(s)
- Rebecca Ting Jiin Loo
- Biomedical Data Science Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6 avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Mohamed Soudy
- Biomedical Data Science Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6 avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Francesco Nasta
- Biomedical Data Science Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6 avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Mirco Macchi
- Biomedical Data Science Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6 avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Enrico Glaab
- Biomedical Data Science Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6 avenue du Swing, L-4367 Belvaux, Luxembourg
| |
Collapse
|
19
|
Shi Y, Ma J, Li S, Liu C, Liu Y, Chen J, Liu N, Liu S, Huang H. Sex difference in human diseases: mechanistic insights and clinical implications. Signal Transduct Target Ther 2024; 9:238. [PMID: 39256355 PMCID: PMC11387494 DOI: 10.1038/s41392-024-01929-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 06/26/2024] [Accepted: 07/23/2024] [Indexed: 09/12/2024] Open
Abstract
Sex characteristics exhibit significant disparities in various human diseases, including prevalent cardiovascular diseases, cancers, metabolic disorders, autoimmune diseases, and neurodegenerative diseases. Risk profiles and pathological manifestations of these diseases exhibit notable variations between sexes. The underlying reasons for these sex disparities encompass multifactorial elements, such as physiology, genetics, and environment. Recent studies have shown that human body systems demonstrate sex-specific gene expression during critical developmental stages and gene editing processes. These genes, differentially expressed based on different sex, may be regulated by androgen or estrogen-responsive elements, thereby influencing the incidence and presentation of cardiovascular, oncological, metabolic, immune, and neurological diseases across sexes. However, despite the existence of sex differences in patients with human diseases, treatment guidelines predominantly rely on male data due to the underrepresentation of women in clinical trials. At present, there exists a substantial knowledge gap concerning sex-specific mechanisms and clinical treatments for diverse diseases. Therefore, this review aims to elucidate the advances of sex differences on human diseases by examining epidemiological factors, pathogenesis, and innovative progress of clinical treatments in accordance with the distinctive risk characteristics of each disease and provide a new theoretical and practical basis for further optimizing individualized treatment and improving patient prognosis.
Collapse
Affiliation(s)
- Yuncong Shi
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Jianshuai Ma
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Sijin Li
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Chao Liu
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Yuning Liu
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China
| | - Jie Chen
- Department of Radiotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ningning Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Shiming Liu
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Hui Huang
- Department of Cardiology, the Eighth Affiliated Hospital, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, Sun Yat-sen University, Shenzhen, China.
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
20
|
Raheel K, See QR, Munday V, Fakhroo B, Ivanenko O, Salvatelli ML, Mutti C, Goadsby PJ, Delogu A, Naismith SL, Holland P, Parrino L, Chaudhuri KR, Rosenzweig I. Orexin and Sleep Disturbances in Alpha-Synucleinopathies: a Systematic Review. Curr Neurol Neurosci Rep 2024; 24:389-412. [PMID: 39031323 PMCID: PMC11349833 DOI: 10.1007/s11910-024-01359-6] [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] [Accepted: 07/02/2024] [Indexed: 07/22/2024]
Abstract
PURPOSE OF REVIEW Sleep disturbances are amongst most frequent non-motor symptoms of Parkinson's Disease (PD), and they are similarly frequently reported in other alpha-syncleinopathies, such as Dementia with Lewy Bodies (DLB) and Multiple System Atrophy (MSA). More recently, the orexin system has been implicated in control of arousal based on salient environmental set points, and its dysregulation in sleep issues in alpha-synucleinopathies suggested by the findings from the translational animal models. However, its role in the patients with alpha-synucleinopathies remains unclear. We thus set to systematically review, and to critically assess, contemporary evidence on the association of the orexinergic system and sleep disturbances in alpha-synucleinopathies. In this systematic review, studies investigating orexin and sleep in alpha-synucleinopathies (Rapid Eye Movement (REM) Behaviour Disorder (RBD), Parkinson's Disease (PD), Dementia with Lewy Bodies (DLB), Multiple System Atrophy (MSA)) were identified using electronic database searches of PubMed, Web of Science and PsychINFO using MeSH terms, keywords, and title words such as "Alpha-synucleinopathies" AND "Orexin" AND "Sleep Disturbances". RECENT FINDINGS 17 studies were included in this systemic review, of which 2 studies on RBD, 10 on PD, 4 on DLB, and 1 on MSA patients. Taken together, RBD and PD studies suggest a potential adaptive increase in orexin levels in early stages of the neurodegenerative process, with reduced levels more often reported for later, more advanced stages of illness. To date, no differences in orexin levels were demonstrated between MSA patients and healthy controls. There is a dearth of studies on the role of orexin levels in alpha-synucleinopathies. Moreover, significant methodologic limitations in the current body of work, including use of non-standardised research protocols and lack of prospective, multi-centre studies, disallow for any finite conclusion in regards to underlying pathomechanisms. Nonetheless, a picture of a complex, multifaceted relationship between the dysregulation of the orexinergic pathway and sleep disturbances in alpha-synucleinopathies is emerging. Hence, future studies disentangling orexinergic pathomechanisms of alpha-syncleinopathies are urgently needed to obtain a more comprehensive account of the role of orexinergic pathway in alpha-synucleinopathies. Pharmacological manipulations of orexins may have multiple therapeutic applications in treatment strategies, disease diagnosis, and might be effective for treating both motor and non-motor symptoms.
Collapse
Affiliation(s)
- Kausar Raheel
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Qi Rui See
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Veronica Munday
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Basma Fakhroo
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Olga Ivanenko
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Marcello Luigi Salvatelli
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, De Crespigny Park, London, SE5 8AF, UK
- Sleep Disorders Center, Department of General and Specialized Medicine, University Hospital of Parma, 43125, Parma, Italy
| | - Carlotta Mutti
- Sleep Disorders Center, Department of General and Specialized Medicine, University Hospital of Parma, 43125, Parma, Italy
| | - Peter J Goadsby
- NIHR-Wellcome Trust King's Clinical Research Facility, King's College London, London, WC2R 2LS, UK
| | - Alessio Delogu
- Basic and Clinical Neuroscience, IoPPN, King's College London, London, WC2R 2LS, UK
| | - Sharon L Naismith
- Healthy Brain Ageing Program, School of Psychology; Brain and Mind Centre, The University of Sydney, & Charles Perkins Centre, Camperdown, Sydney, Australia
| | - Phil Holland
- Basic and Clinical Neuroscience, IoPPN, King's College London, London, WC2R 2LS, UK
| | - Liborio Parrino
- Sleep Disorders Center, Department of General and Specialized Medicine, University Hospital of Parma, 43125, Parma, Italy
- Department of Medicine and Surgery, Neurology Unit, University of Parma, 43125, Parma, Italy
| | - K Ray Chaudhuri
- Movement Disorders Unit, King's College Hospital and Department of Clinical and Basic Neurosciences, Institute of Psychiatry, Psychology & Neuroscience and Parkinson Foundation Centre of Excellence, King's College London, London, UK
| | - Ivana Rosenzweig
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, De Crespigny Park, London, SE5 8AF, UK.
- Sleep Disorders Centre, Guy's and St Thomas' NHS Foundation Trust, London, UK.
| |
Collapse
|
21
|
Freuchet A, Pinçon A, Sette A, Lindestam Arlehamn CS. Inflammation and heterogeneity in synucleinopathies. Front Immunol 2024; 15:1432342. [PMID: 39281666 PMCID: PMC11392857 DOI: 10.3389/fimmu.2024.1432342] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024] Open
Abstract
Neurodegenerative diseases represent a huge healthcare challenge which is predicted to increase with an aging population. Synucleinopathies, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA), present complex challenges in understanding their onset and progression. They are characterized by the abnormal aggregation of α-synuclein in the brain leading to neurodegeneration. Accumulating evidence supports the existence of distinct subtypes based on the site of α-synuclein aggregation initiation, genetics, and, more recently, neuroinflammation. Mediated by both central nervous system-resident cells, peripheral immune cells, and gut dysbiosis, neuroinflammation appears as a key process in the onset and progression of neuronal loss. Sex-based differences add another layer of complexity to synucleinopathies, influencing disease prevalence - with a known higher incidence of PD in males compared to females - as well as phenotype and immune responses. Biological sex affects neuroinflammatory pathways and the immune response, suggesting the need for sex-specific therapeutic strategies and biomarker identification. Here, we review the heterogeneity of synucleinopathies, describing the etiology, the mechanisms by which the inflammatory processes contribute to the pathology, and the consideration of sex-based differences to highlight the need for personalized therapeutics.
Collapse
Affiliation(s)
- Antoine Freuchet
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, San Diego, CA, United States
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| | - Anaëlle Pinçon
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, San Diego, CA, United States
- Master de Biologie, Ecole Normale Superieure de Lyon, University of Lyon, Lyon, France
| | - Alessandro Sette
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, San Diego, CA, United States
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
- Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Cecilia S Lindestam Arlehamn
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, San Diego, CA, United States
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, United States
| |
Collapse
|
22
|
Li S, Song H, Yu C. Causal association between phenylalanine and Parkinson's disease: a two-sample bidirectional mendelian randomization study. Front Genet 2024; 15:1322551. [PMID: 39011398 PMCID: PMC11246959 DOI: 10.3389/fgene.2024.1322551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Background Research findings indicate a putative indirect or latent association between phenylalanine (Phe) and Parkinson's disease (PD). In this study, we aimed to analyze the causal relationship between Phe and PD by two sample Mendelian randomization (MR) analysis. Methods In this study, the PD-related dataset and Phe-related dataset were downloaded from Integrative Epidemiology U1nit (IEU) Open Genome-Wide Association Study (GWAS) database. Four algorithms (MR Egger, maximum likelihood, inverse variance weighting (IVW) and unweighted regression) were used to perform MR analysis. The sensitivity analysis (heterogeneity test, horizontal pleiotropy test and Leave-One-Out (LOO) analysis) was used to assess the reliability of MR analyses. Results In the forward MR analysis, Phe was a safety factor for PD (p-value < 0.05 and odds ratios (OR) < 1). The results of reverse MR analysis showed that there was no causal relationship between PD and Phe (p-value > 0.05). In addition, sensitivity analysis showed that MR analysis was reliable. Conclusion The results of this study revealed that Phe was a safety factor for PD, meaning that Phe reduced the risk of PD.
Collapse
Affiliation(s)
- Shiqing Li
- Nantong Haimen People’s Hospital, Neurosurgery Department, Jiangsu, China
| | | | | |
Collapse
|
23
|
Cucinotta L, Mannino D, Filippone A, Romano A, Esposito E, Paterniti I. The role of autophagy in Parkinson's disease: a gender difference overview. Front Pharmacol 2024; 15:1408152. [PMID: 38933683 PMCID: PMC11199695 DOI: 10.3389/fphar.2024.1408152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Recent studies have demonstrated dysregulation of the autophagy pathway in patients with Parkinson's disease (PD) and in animal models of PD, highlighting its emerging role in disease. In particular, several studies indicate that autophagy, which is an essential degradative process for the damaged protein homeostasis and the management of cell balance, can manifest significant variations according to gender. While some evidence suggests increased autophagic activation in men with PD, women may have distinct regulatory patterns. In this review, we examined the existing literature on gender differences in PD-associated autophagic processes, focusing on the autophagy related proteins (ATGs) and leucine rich repeat kinase 2 (LRRK2) genes. Also, this review would suggest that an in-depth understanding of these gender differences in autophagic processes could open new perspectives for personalized therapeutic strategies, promoting more effective and targeted management of PD.
Collapse
Affiliation(s)
- Laura Cucinotta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Deborah Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Adele Romano
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
24
|
Bovenzi R, Schirinzi T, Conti M, Sancesario GM, Zenuni H, Simonetta C, Bissacco J, Mascioli D, Pieri M, Cerroni R, Stefani A, Mercuri NB, Pierantozzi M. A biological characterization of patients with postmenopausal Parkinson's disease. J Neurol 2024; 271:3610-3615. [PMID: 38492015 DOI: 10.1007/s00415-024-12258-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 03/18/2024]
Abstract
Menopause increases the risk for Parkinson's disease (PD), although the underlying biological mechanisms have not been established in patients. Here, we aimed to understand the basis of menopause-related vulnerability to PD. Main motor and non-motor scores, blood levels of estradiol, testosterone, follicle-stimulating hormone, and luteinizing hormone, CSF levels of total α-synuclein, amyloid-β-42, amyloid-β-40, total tau, and phosphorylated-181-tau were examined in 45 women with postmenopausal-onset PD and 40 age-matched controls. PD patients had higher testosterone and lower estradiol levels than controls, and the residual estradiol production was associated with milder motor disturbances and lower dopaminergic requirements. In PD but not in controls, follicle-stimulating hormone levels correlated with worse cognitive scores and CSF markers of amyloidopathy and neuronal loss. In conclusion, menopause-related hormonal changes might differentially contribute to clinical-pathological trajectories of PD, accounting for the peculiar vulnerability to the disease.
Collapse
Affiliation(s)
- Roberta Bovenzi
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Tommaso Schirinzi
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.
| | - Matteo Conti
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | | | - Henri Zenuni
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Clara Simonetta
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Jacopo Bissacco
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Davide Mascioli
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Massimo Pieri
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
- Department of Clinical Biochemistry, Tor Vergata University Hospital, Rome, Italy
| | - Rocco Cerroni
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Alessandro Stefani
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
- UOSD Parkinson Centre, Tor Vergata University Hospital, Rome, Italy
| | - Nicola Biagio Mercuri
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Mariangela Pierantozzi
- Unit of Neurology, Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
25
|
Kochmanski J, Virani M, Kuhn NC, Boyd SL, Becker K, Adams M, Bernstein AI. Developmental origins of Parkinson's disease risk: perinatal exposure to the organochlorine pesticide dieldrin leads to sex-specific DNA modifications in critical neurodevelopmental pathways in the mouse midbrain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.590998. [PMID: 38746441 PMCID: PMC11092502 DOI: 10.1101/2024.04.26.590998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Epidemiological studies show that exposure to the organochlorine pesticide dieldrin is associated with increased risk of Parkinson's disease (PD). Animal studies support a link between developmental dieldrin exposure and increased neuronal susceptibility in the α-synuclein preformed fibril (α-syn PFF) and MPTP models in adult male C57BL/6 mice. In a previous study, we showed that developmental dieldrin exposure was associated with sex-specific changes in DNA modifications within genes related to dopaminergic neuron development and maintenance at 12 weeks of age. Here, we used capture hybridization-sequencing with custom baits to interrogate DNA modifications across the entire genetic loci of the previously identified genes at multiple time points - birth, 6 weeks, 12 weeks, and 36 weeks old. We identified largely sex-specific dieldrin-induced changes in DNA modifications at each time point that annotated to pathways important for neurodevelopment, potentially related to critical steps in early neurodevelopment, dopaminergic neuron differentiation, synaptogenesis, synaptic plasticity, and glial-neuron interactions. Despite large numbers of age-specific DNA modifications, longitudinal analysis identified a small number of DMCs with dieldrin-induced deflection of epigenetic aging. The sex-specificity of these results adds to evidence that sex-specific responses to PD-related exposures may underly sex-specific differences in disease. Overall, these data support the idea that developmental dieldrin exposure leads to changes in epigenetic patterns that persist after the exposure period and disrupt critical neurodevelopmental pathways, thereby impacting risk of late life diseases, including PD.
Collapse
Affiliation(s)
- Joseph Kochmanski
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| | - Mahek Virani
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ
| | - Nathan C. Kuhn
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| | - Sierra L. Boyd
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| | - Katelyn Becker
- Genomics Core, Van Andel Research Institute, Grand Rapids, MI
| | - Marie Adams
- Genomics Core, Van Andel Research Institute, Grand Rapids, MI
| | - Alison I. Bernstein
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ
- Environmental and Occupational Health Sciences Institute, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI
| |
Collapse
|
26
|
Lee J, Sohn YH, Chung SJ, Kim E, Kim Y. Presentations of nonmotor symptoms by sex and onset age in people with Parkinson's disease. Int J Nurs Pract 2024; 30:e13177. [PMID: 37394896 DOI: 10.1111/ijn.13177] [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: 01/15/2023] [Revised: 05/18/2023] [Accepted: 06/13/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND There is growing evidence that sex and onset age are important factors of clinical features in Parkinson's disease. AIM The study aimed to identify nonmotor symptoms based on sex and onset age in people with Parkinson's disease. DESIGN This is a cross-sectional descriptive study. METHODS A total of 210 participants were recruited from the university hospital and the Parkinson's disease association. This study measured the Korean version of the nonmotor symptoms questionnaire which includes gastrointestinal, urinary, apathy/attention/memory, hallucination/delusions, depression/anxiety, sexual function, cardiovascular, sleep disorder, and miscellaneous domains. RESULTS All participants reported at least one nonmotor symptom. The most commonly reported symptoms were nocturia (65.7%) and constipation (61.9%). The male participants reported more dribbling of saliva, constipation, and impaired sexual function, whereas the female reported more weight change. Young-onset people with Parkinson's disease reported more depression than late-onset people with Parkinson's disease. CONCLUSION This study contributes to the understanding of symptom experience beyond motor-related symptomatology for those with Parkinson's disease and adds to the current literature. Individualized symptom assessment and management should be provided by prioritizing prevalent sex or onset age-specific symptoms, rather than addressing with all nonmotor symptoms.
Collapse
Affiliation(s)
- JuHee Lee
- Mo-Im Kim Nursing Research Institute, Yonsei Evidence Based Nursing Centre of Korea: A Joanna Briggs Institute of Excellence, College of Nursing, Yonsei University, Seoul, South Korea
| | - Young H Sohn
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Seok Jong Chung
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, South Korea
| | - Eunyoung Kim
- College of Nursing and Brain Korea 21 FOUR Project, Yonsei University, Seoul, South Korea
| | - Yielin Kim
- Division of Nursing, Severance Hospital, Yonsei University Health System, Seoul, South Korea
| |
Collapse
|
27
|
Nordengen K, Cappelletti C, Bahrami S, Frei O, Pihlstrøm L, Henriksen SP, Geut H, Rozemuller AJM, van de Berg WDJ, Andreassen OA, Toft M. Pleiotropy with sex-specific traits reveals genetic aspects of sex differences in Parkinson's disease. Brain 2024; 147:858-870. [PMID: 37671566 PMCID: PMC10907091 DOI: 10.1093/brain/awad297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/01/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023] Open
Abstract
Parkinson's disease is an age-related neurodegenerative disorder with a higher incidence in males than females. The causes for this sex difference are unknown. Genome-wide association studies (GWAS) have identified 90 Parkinson's disease risk loci, but the genetic studies have not found sex-specific differences in allele frequency on autosomal chromosomes or sex chromosomes. Genetic variants, however, could exert sex-specific effects on gene function and regulation of gene expression. To identify genetic loci that might have sex-specific effects, we studied pleiotropy between Parkinson's disease and sex-specific traits. Summary statistics from GWASs were acquired from large-scale consortia for Parkinson's disease (n cases = 13 708; n controls = 95 282), age at menarche (n = 368 888 females) and age at menopause (n = 69 360 females). We applied the conditional/conjunctional false discovery rate (FDR) method to identify shared loci between Parkinson's disease and these sex-specific traits. Next, we investigated sex-specific gene expression differences in the superior frontal cortex of both neuropathologically healthy individuals and Parkinson's disease patients (n cases = 61; n controls = 23). To provide biological insights to the genetic pleiotropy, we performed sex-specific expression quantitative trait locus (eQTL) analysis and sex-specific age-related differential expression analysis for genes mapped to Parkinson's disease risk loci. Through conditional/conjunctional FDR analysis we found 11 loci shared between Parkinson's disease and the sex-specific traits age at menarche and age at menopause. Gene-set and pathway analysis of the genes mapped to these loci highlighted the importance of the immune response in determining an increased disease incidence in the male population. Moreover, we highlighted a total of nine genes whose expression or age-related expression in the human brain is influenced by genetic variants in a sex-specific manner. With these analyses we demonstrated that the lack of clear sex-specific differences in allele frequencies for Parkinson's disease loci does not exclude a genetic contribution to differences in disease incidence. Moreover, further studies are needed to elucidate the role that the candidate genes identified here could have in determining a higher incidence of Parkinson's disease in the male population.
Collapse
Affiliation(s)
- Kaja Nordengen
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
| | - Chiara Cappelletti
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Department of Mechanical, Electronics and Chemical Engineering, Faculty of Technology, Art and Design, OsloMet—Oslo Metropolitan University, 0130 Oslo, Norway
- Department of Research, Innovation and Education, Oslo University Hospital, 0424 Oslo, Norway
| | - Shahram Bahrami
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway
| | - Oleksandr Frei
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway
| | - Lasse Pihlstrøm
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
| | | | - Hanneke Geut
- Section of Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 Amsterdam, The Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 Amsterdam, The Netherlands
| | - Wilma D J van de Berg
- Section of Clinical Neuroanatomy and Biobanking, Department of Anatomy and Neurosciences, Amsterdam UMC, Location Vrije Universiteit Amsterdam, Amsterdam Neuroscience, 1081 Amsterdam, The Netherlands
| | - Ole A Andreassen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
- Norwegian Centre for Mental Disorders Research (NORMENT), Division of Mental Health and Addiction, Oslo University Hospital, 0450 Oslo, Norway
| | - Mathias Toft
- Department of Neurology, Oslo University Hospital, 0424 Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0372 Oslo, Norway
| |
Collapse
|
28
|
Yoon SY, Lee SC, Suh JH, Yang SN, Han K, Kim YW. Different risks of early-onset and late-onset Parkinson disease in individuals with mental illness. NPJ Parkinsons Dis 2024; 10:17. [PMID: 38195604 PMCID: PMC10776668 DOI: 10.1038/s41531-023-00621-x] [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: 09/04/2023] [Accepted: 12/08/2023] [Indexed: 01/11/2024] Open
Abstract
We aimed to investigate the association of various mental illnesses, including depression, bipolar disorder, schizophrenia, insomnia, and anxiety, with the risk of early-onset Parkinson's disease (EOPD) (age <50 years) and compare it with that of late-onset PD (LOPD) (age ≥50 years). This nationwide cohort study enrolled 9,920,522 people who underwent a national health screening examination in 2009, and followed up until 31 December 2018. There was a significantly increased risk of EOPD and LOPD in individuals with mental illness, and EOPD showed a stronger association than LOPD (EOPD, hazard ratio (HR) = 3.11, 95% CI: 2.61‒3.72; LOPD, HR = 1.70, 95% CI: 1.66‒1.74; p for interaction <0.0001). Our results suggest that people with mental illnesses aged < 50 years are at a higher risk of PD than those aged ≥50 years. Future studies are warranted to elucidate the pathomechanism of EOPD in relation to mental illness.
Collapse
Affiliation(s)
- Seo Yeon Yoon
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang Chul Lee
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jee Hyun Suh
- Department of Rehabilitation Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Seung Nam Yang
- Department of Physical Medicine & Rehabilitation, Korea University Guro Hospital, Seoul, Republic of Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea.
| | - Yong Wook Kim
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
29
|
Chen YH, Kuo TT, Wang V, Cheng PW, Huang EYK, Ma KH, Greig NH, Olson L, Hoffer BJ, Tseng KY. Serotonergic Regulation of Synaptic Dopamine Levels Mitigates L-DOPA-Induced Dyskinesia in a Mouse Model of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:941-964. [PMID: 38905058 PMCID: PMC11307072 DOI: 10.3233/jpd-240080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 06/23/2024]
Abstract
Background The serotonin (5-HT) system can manipulate the processing of exogenous L-DOPA in the DA-denervated striatum, resulting in the modulation of L-DOPA-induced dyskinesia (LID). Objective To characterize the effects of the serotonin precursor 5-hydroxy-tryptophan (5-HTP) or the serotonin transporter (SERT) inhibitor, Citalopram on L-DOPA-induced behavior, neurochemical signals, and underlying protein expressions in an animal model of Parkinson's disease. Methods MitoPark (MP) mice at 20 weeks of age, subjected to a 14-day administration of L-DOPA/Carbidopa, displayed dyskinesia, referred to as LID. Subsequent investigations explored the effects of 5-HT-modifying agents, such as 5-HTP and Citalopram, on abnormal involuntary movements (AIMs), locomotor activity, neurochemical signals, serotonin transporter activity, and protein expression in the DA-denervated striatum of LID MP mice. Results 5-HTP exhibited duration-dependent suppressive effects on developing and established LID, especially related to abnormal limb movements observed in L-DOPA-primed MP mice. However, Citalopram, predominantly suppressed abnormal axial movement induced by L-DOPA in LID MP mice. We demonstrated that 5-HTP could decrease L-DOPA-upregulation of DA turnover rates while concurrently upregulating 5-HT metabolism. Additionally, 5-HTP was shown to reduce the expressions of p-ERK and p-DARPP-32 in the striatum of LID MP mice. The effect of Citalopram in alleviating LID development may be attributed to downregulation of SERT activity in the dorsal striatum of LID MP mice. Conclusions While both single injection of 5-HTP and Citalopram effectively mitigated the development of LID, the difference in mitigation of AIM subtypes may be linked to the unique effects of these two serotonergic agents on L-DOPA-derived DA and 5-HT metabolism.
Collapse
Affiliation(s)
- Yuan-Hao Chen
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan
- National Defense Medical Center, Taipei, Taiwan
| | - Tung-Tai Kuo
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan
| | - Vicki Wang
- Doctoral Degree Program in Translational Medicine, National Defense Medical Center and Academia Sinica, Taipei, Taiwan
| | - Pin-Wen Cheng
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan
| | | | - Kuo-Hsing Ma
- Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, National Institutes of Health (NIH), Baltimore, MD, USA
| | - Lars Olson
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Barry J. Hoffer
- Department of Neurosurgery, University Hospitals of Cleveland, Cleveland, OH, USA
| | - Kuan-Yin Tseng
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan
- National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
30
|
Yoon SY, Choi JY, Nam GE, Jung JH, Han K, Kang SH, Kim CK, Kim YW, Koh SB. Association Between Body Mass Index Changes and All-Cause Mortality in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:1441-1450. [PMID: 39331108 PMCID: PMC11492103 DOI: 10.3233/jpd-240181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/12/2024] [Indexed: 09/28/2024]
Abstract
Background Whether body weight changes are associated with Parkinson's disease (PD) mortality remains uncertain. Objective To investigate the association between changes in body mass index (BMI) and all-cause mortality in patients with PD. Methods This nationwide cohort study enrolled 20,703 individuals with new-onset PD (ICD-10 code: G20 and a rare intractable disease registration code: V124) who underwent health screening program by the Korean National Health Insurance Service within two years from pre- and post-PD diagnosis. We identified nine BMI change groups based on three BMI status: underweight (BMI < 18.5 kg/m2), normal or overweight (18.5 kg/m2≤BMI < 25 kg/m2), and obese (BMI≥25 kg/m2). Results Of 20,703 individuals, 3,789 (18.0%) died during the follow-up period. Excessive weight loss to underweight in the obese group (hazard ratio [HR] = 3.36, 95% CI:1.60-7.08), weight loss in the normal to overweight group (HR = 2.04, 95% CI:1.75-2.39), sustained underweight status (HR = 2.05, 95% CI:1.67-2.52), and weight gain from underweight to normal or overweight (HR = 1.52, 95% CI:1.15-2.02) were associated with increased mortality. Sustained obese status (HR = 0.80, 95% CI:0.74-0.87) and weight gain in the normal to overweight group (HR = 0.82, 95% CI:0.71-0.95) were associated with reduced mortality. Conclusions We found that BMI change at diagnosis was associated with mortality in patients with PD. Specifically, being underweight either before or after diagnosis as well as experiencing weight loss, were associated with increased mortality. These findings provide valuable insights for weight management planning in PD, highlighting the importance of individualized approach that consider pre-diagnosis BMI.
Collapse
Affiliation(s)
- Seo Yeon Yoon
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Ja Young Choi
- Department of Physical and Rehabilitation Medicine, Chungnam National University College of Medicine, Daejeon, South Korea
| | - Ga Eun Nam
- Department of Family Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Jin-Hyung Jung
- Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, South Korea
| | - Sung Hoon Kang
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Chi Kyung Kim
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| | - Yong Wook Kim
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Seong-Beom Koh
- Department of Neurology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, South Korea
| |
Collapse
|
31
|
Khor SLQ, Ng KY, Koh RY, Chye SM. Blood-brain Barrier and Neurovascular Unit Dysfunction in Parkinson's Disease: From Clinical Insights to Pathogenic Mechanisms and Novel Therapeutic Approaches. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:315-330. [PMID: 36999187 DOI: 10.2174/1871527322666230330093829] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 04/01/2023]
Abstract
The blood-brain barrier (BBB) plays a crucial role in the central nervous system by tightly regulating the influx and efflux of biological substances between the brain parenchyma and peripheral circulation. Its restrictive nature acts as an obstacle to protect the brain from potentially noxious substances such as blood-borne toxins, immune cells, and pathogens. Thus, the maintenance of its structural and functional integrity is vital in the preservation of neuronal function and cellular homeostasis in the brain microenvironment. However, the barrier's foundation can become compromised during neurological or pathological conditions, which can result in dysregulated ionic homeostasis, impaired transport of nutrients, and accumulation of neurotoxins that eventually lead to irreversible neuronal loss. Initially, the BBB is thought to remain intact during neurodegenerative diseases, but accumulating evidence as of late has suggested the possible association of BBB dysfunction with Parkinson's disease (PD) pathology. The neurodegeneration occurring in PD is believed to stem from a myriad of pathogenic mechanisms, including tight junction alterations, abnormal angiogenesis, and dysfunctional BBB transporter mechanism, which ultimately causes altered BBB permeability. In this review, the major elements of the neurovascular unit (NVU) comprising the BBB are discussed, along with their role in the maintenance of barrier integrity and PD pathogenesis. We also elaborated on how the neuroendocrine system can influence the regulation of BBB function and PD pathogenesis. Several novel therapeutic approaches targeting the NVU components are explored to provide a fresh outlook on treatment options for PD.
Collapse
Affiliation(s)
- Sarah Lei Qi Khor
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University, 47500, Selangor, Malaysia
| | - Rhun Yian Koh
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- Division of Applied Biomedical Science and Biotechnology, School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| |
Collapse
|
32
|
Khayretdinova M, Zakharov I, Pshonkovskaya P, Adamovich T, Kiryasov A, Zhdanov A, Shovkun A. Prediction of brain sex from EEG: using large-scale heterogeneous dataset for developing a highly accurate and interpretable ML model. Neuroimage 2024; 285:120495. [PMID: 38092156 DOI: 10.1016/j.neuroimage.2023.120495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/29/2023] [Accepted: 12/10/2023] [Indexed: 12/17/2023] Open
Abstract
This study presents a comprehensive examination of sex-related differences in resting-state electroencephalogram (EEG) data, leveraging two different types of machine learning models to predict an individual's sex. We utilized data from the Two Decades-Brainclinics Research Archive for Insights in Neurophysiology (TDBRAIN) EEG study, affirming that gender prediction can be attained with noteworthy accuracy. The best performing model achieved an accuracy of 85% and an ROC AUC of 89%, surpassing all prior benchmarks set using EEG data and rivaling the top-tier results derived from fMRI studies. A comparative analysis of LightGBM and Deep Convolutional Neural Network (DCNN) models revealed DCNN's superior performance, attributed to its ability to learn complex spatial-temporal patterns in the EEG data and handle large volumes of data effectively. Despite this, interpretability remained a challenge for the DCNN model. The LightGBM interpretability analysis revealed that the most important EEG features for accurate sex prediction were related to left fronto-central and parietal EEG connectivity. We also showed the role of both low (delta and theta) and high (beta and gamma) activity in the accurate sex prediction. These results, however, have to be approached with caution, because it was obtained from a dataset comprised largely of participants with various mental health conditions, which limits the generalizability of the results and necessitates further validation in future studies. . Overall, the study illuminates the potential of interpretable machine learning for sex prediction, alongside highlighting the importance of considering individual differences in prediction sex from brain activity.
Collapse
|
33
|
Ha WS, Kim J, Hwang HW, Lee SH, Kim JI, Hong JY, Park SH, Han KD, Baek MS. The association between migraine and Parkinson's disease: a nationwide cohort study. Epidemiol Health 2023; 46:e2024010. [PMID: 38186247 PMCID: PMC10928470 DOI: 10.4178/epih.e2024010] [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: 06/30/2023] [Accepted: 11/22/2023] [Indexed: 01/09/2024] Open
Abstract
OBJECTIVES Clinical studies have suggested an association between migraine and the occurrence of Parkinson's disease (PD). However, it is unknown whether migraine affects PD risk. We aimed to investigate the incidence of PD in patients with migraine and to determine the risk factors affecting the association between migraine and PD incidence. METHODS Using the Korean National Health Insurance System database (2002-2019), we enrolled all Koreans aged ≥40 years who participated in the national health screening program in 2009. International Classification of Diseases (10th revision) diagnostic codes and Rare Incurable Diseases System diagnostic codes were used to define patients with migraine (within 12 months of enrollment) and newly diagnosed PD. RESULTS We included 214,193 patients with migraine and 5,879,711 individuals without migraine. During 9.1 years of follow-up (55,435,626 person-years), 1,973 (0.92%) and 30,664 (0.52%) individuals with and without migraine, respectively, were newly diagnosed with PD. Following covariate adjustment, patients with migraine showed a 1.35-fold higher PD risk than individuals without migraine. The incidence of PD was not significantly different between patients with migraine with aura and those without aura. In males with migraine, underlying dyslipidemia increased the risk of PD (p=0.012). In contrast, among females with migraine, younger age (<65 years) increased the risk of PD (p=0.038). CONCLUSIONS Patients with migraine were more likely to develop PD than individuals without migraine. Preventive management of underlying comorbidities and chronic migraine may affect the incidence of PD in these patients. Future prospective randomized clinical trials are warranted to clarify this association.
Collapse
Affiliation(s)
- Woo-Seok Ha
- Department of Neurology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jaeho Kim
- Department of Neurology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Korea
| | - Hee Won Hwang
- Department of Neurology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Sue Hyun Lee
- Department of Neurology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Ji In Kim
- Department of Neurology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Jin Yong Hong
- Department of Neurology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Sang Hyun Park
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea
| | - Kyung Do Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Korea
| | - Min Seok Baek
- Department of Neurology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Korea
- Research Institute of Metabolism and Inflammation, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
34
|
Buck SA, Rubin SA, Kunkhyen T, Treiber CD, Xue X, Fenno LE, Mabry SJ, Sundar VR, Yang Z, Shah D, Ketchesin KD, Becker-Krail DD, Vasylieva I, Smith MC, Weisel FJ, Wang W, Erickson-Oberg MQ, O’Leary EI, Aravind E, Ramakrishnan C, Kim YS, Wu Y, Quick M, Coleman JA, MacDonald WA, Elbakri R, De Miranda BR, Palladino MJ, McCabe BD, Fish KN, Seney ML, Rayport S, Mingote S, Deisseroth K, Hnasko TS, Awatramani R, Watson AM, Waddell S, Cheetham CEJ, Logan RW, Freyberg Z. Sexually dimorphic mechanisms of VGLUT-mediated protection from dopaminergic neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560584. [PMID: 37873436 PMCID: PMC10592912 DOI: 10.1101/2023.10.02.560584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Parkinson's disease (PD) targets some dopamine (DA) neurons more than others. Sex differences offer insights, with females more protected from DA neurodegeneration. The mammalian vesicular glutamate transporter VGLUT2 and Drosophila ortholog dVGLUT have been implicated as modulators of DA neuron resilience. However, the mechanisms by which VGLUT2/dVGLUT protects DA neurons remain unknown. We discovered DA neuron dVGLUT knockdown increased mitochondrial reactive oxygen species in a sexually dimorphic manner in response to depolarization or paraquat-induced stress, males being especially affected. DA neuron dVGLUT also reduced ATP biosynthetic burden during depolarization. RNA sequencing of VGLUT+ DA neurons in mice and flies identified candidate genes that we functionally screened to further dissect VGLUT-mediated DA neuron resilience across PD models. We discovered transcription factors modulating dVGLUT-dependent DA neuroprotection and identified dj-1β as a regulator of sex-specific DA neuron dVGLUT expression. Overall, VGLUT protects DA neurons from PD-associated degeneration by maintaining mitochondrial health.
Collapse
Affiliation(s)
- Silas A. Buck
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sophie A. Rubin
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tenzin Kunkhyen
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Christoph D. Treiber
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Lief E. Fenno
- Departments of Psychiatry and Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Samuel J. Mabry
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Varun R. Sundar
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Zilu Yang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Divia Shah
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kyle D. Ketchesin
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Darius D. Becker-Krail
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Iaroslavna Vasylieva
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Megan C. Smith
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Florian J. Weisel
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Wenjia Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - M. Quincy Erickson-Oberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Emma I. O’Leary
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Eshan Aravind
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yoon Seok Kim
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yanying Wu
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Matthias Quick
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Jonathan A. Coleman
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | - Rania Elbakri
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Briana R. De Miranda
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michael J. Palladino
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Pittsburgh Institute of Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Brian D. McCabe
- Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Kenneth N. Fish
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Marianne L. Seney
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Stephen Rayport
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Susana Mingote
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY 10031, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Thomas S. Hnasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | | | - Alan M. Watson
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Scott Waddell
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | | | - Ryan W. Logan
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
35
|
Cai J, Kim JL, Wang Y, Baumeister TR, Zhu M, Liu A, Lee S, McKeown MJ. Sex, myelin, and clinical characteristics of Parkinson's disease. Front Neurosci 2023; 17:1235524. [PMID: 37781247 PMCID: PMC10535348 DOI: 10.3389/fnins.2023.1235524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Objective To determine if there are sex differences in myelin in Parkinson's disease, and whether these explain some of the previously-described sex differences in clinical presentation. Methods Thirty-three subjects (23 males, 10 females) with Parkinson's disease underwent myelin water fraction (MWF) imaging, an MRI scanning technique of in vivo myelin content. MWF of 20 white matter regions of interest (ROIs) were assessed. Motor symptoms were assessed using the Unified Parkinson's Disease Rating Scale (UPDRS). Principal component analysis, logistic and multiple linear regressions, and t-tests were used to determine which white matter ROIs differed between sexes, the clinical features associated with these myelin changes, and if overall MWF and MWF laterality differed between males and females. Results Consistent with prior reports, tremor and bradykinesia were more likely seen in females, whereas rigidity and axial symptoms were more likely seen in males in our cohort. MWF of the thalamic radiation, cingulum, cingulum hippocampus, inferior fronto-occipital fasciculi, inferior longitudinal fasciculi, and uncinate were significant in predicting sex. Overall MWF and asymmetry of MWF was greater in males. MWF differences between sexes were associated with tremor symptomatology and asymmetry of motor performance. Conclusion Sex differences in myelin are associated with tremor and asymmetry of motor presentation. While preliminary, our results suggest that further investigation of the role of biological sex in myelin pathology and clinical presentation in Parkinson's disease is warranted.
Collapse
Affiliation(s)
- Jiayue Cai
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, China
- Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Jowon L. Kim
- Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Yuheng Wang
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Tobias R. Baumeister
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada
| | - Maria Zhu
- Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Aiping Liu
- Department of Electronic Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Soojin Lee
- Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Martin J. McKeown
- Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
36
|
Zhang X, Huangfu Z, Wang S. Review of mendelian randomization studies on age at natural menopause. Front Endocrinol (Lausanne) 2023; 14:1234324. [PMID: 37766689 PMCID: PMC10520463 DOI: 10.3389/fendo.2023.1234324] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
Menopause marks the end of the reproductive phase of life. Based on epidemiological studies, abnormal age at natural menopause (ANM) is thought to contribute to a number of adverse outcomes, such as osteoporosis, cardiovascular disease, and cancer. However, the causality of these associations remains unclear. A powerful epidemiological method known as Mendelian randomization (MR) can be used to clarify the causality between ANM and other diseases or traits. The present review describes MR studies that included ANM as an exposure, outcome and mediator. The findings of MR analyses on ANM have revealed that higher body mass index, poor educational level, early age at menarche, early age at first live birth, early age at first sexual intercourse, and autoimmune thyroid disease appear to be involved in early ANM etiology. The etiology of late ANM appears to be influenced by higher free thyroxine 4 and methylene tetrahydrofolate reductase gene mutations. Furthermore, early ANM has been found to be causally associated with an increased risk of osteoporosis, fracture, type 2 diabetes mellitus, glycosylated hemoglobin, and the homeostasis model of insulin resistance level. In addition, late ANM has been found to be causally associated with an increased systolic blood pressure, higher risk of breast cancer, endometrial cancer, endometrioid ovarian carcinoma, lung cancer, longevity, airflow obstruction, and lower risk of Parkinson's disease. ANM is also a mediator for breast cancer caused by birth weight and childhood body size. However, due to the different instrumental variables used, some results of studies are inconsistent. Future studies with more valid genetic variants are needed for traits with discrepancies between MRs or between MR and other types of epidemiological studies.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Obstetrics and Gynecology, Beijing Hospital, National Center of Gerontology, Beijing, China
- Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
- Graduate School of Peking Union Medical College, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhao Huangfu
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Shaowei Wang
- Department of Obstetrics and Gynecology, Beijing Hospital, National Center of Gerontology, Beijing, China
- Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing, China
- Graduate School of Peking Union Medical College, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
37
|
Li Z, Wang X, Wang X, Yi X, Wong YK, Wu J, Xie F, Hu D, Wang Q, Wang J, Zhong T. Research progress on the role of extracellular vesicles in neurodegenerative diseases. Transl Neurodegener 2023; 12:43. [PMID: 37697342 PMCID: PMC10494410 DOI: 10.1186/s40035-023-00375-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 08/23/2023] [Indexed: 09/13/2023] Open
Abstract
Neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease, affect millions of people worldwide. Tremendous efforts have been put into disease-related research, but few breakthroughs have been made in diagnostic and therapeutic approaches. Extracellular vesicles (EVs) are heterogeneous cell-derived membrane structures that arise from the endosomal system or are directly separated from the plasma membrane. EVs contain many biomolecules, including proteins, nucleic acids, and lipids, which can be transferred between different cells, tissues, or organs, thereby regulating cross-organ communication between cells during normal and pathological processes. Recently, EVs have been shown to participate in various aspects of neurodegenerative diseases. Abnormal secretion and levels of EVs are closely related to the pathogenesis of neurodegenerative diseases and contribute to disease progression. Numerous studies have proposed EVs as therapeutic targets or biomarkers for neurodegenerative diseases. In this review, we summarize and discuss the advanced research progress on EVs in the pathological processes of several neurodegenerative diseases. Moreover, we outline the latest research on the roles of EVs in neurodegenerative diseases and their therapeutic potential for the diseases.
Collapse
Affiliation(s)
- Zhengzhe Li
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Xiaoling Wang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Xiaoxing Wang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Xiaomei Yi
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Yin Kwan Wong
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, China
| | - Jiyang Wu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Fangfang Xie
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Die Hu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Qi Wang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
| | - Jigang Wang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
- Department of Nephrology, Shenzhen Key Laboratory of Kidney Diseases, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, China.
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Tianyu Zhong
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China.
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
38
|
Al-Zaid FS, Hurley MJ, Dexter DT, Gillies GE. Neuroprotective role for RORA in Parkinson's disease revealed by analysis of post-mortem brain and a dopaminergic cell line. NPJ Parkinsons Dis 2023; 9:119. [PMID: 37500636 PMCID: PMC10374904 DOI: 10.1038/s41531-023-00563-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 07/17/2023] [Indexed: 07/29/2023] Open
Abstract
Parkinson's disease (PD) is almost twice as prevalent in men, which has largely been attributed to neuroprotective effect of oestradiol in women. RORA (retinoic acid receptor-related orphan receptor alpha) regulates the transcription of central aromatase, the enzyme responsible for local oestradiol synthesis, simultaneously, RORA expression is regulated by sex hormones. Moreover, RORA protects neurones against oxidative stress, a key mechanism contributing to the loss of dopaminergic neurones in PD. Therefore, we hypothesized that there would be sex differences in RORA expression in the substantia nigra pars compacta (SNpc), which could contribute to sex differences observed in PD prevalence and pathogenesis. In a case control study, qPCR and western blot analyses were used to quantify gene and protein expression in the SNpc of post-mortem brains (n = 14 late-stage PD and 11 age and sex matched controls). The neuroprotective properties of a RORA agonist were then investigated directly using a cell culture toxin-based model of PD coupled with measures of viability, mitochondrial function and apoptosis. RORA was expressed at significantly higher levels in the SNpc from control females' brains compared to males. In PD, we found a significant increase in SNpc RORA expression in male PD compared to female PD. Treatment with a RORA agonist showed a significant neuroprotection in our cell culture model of PD and revealed significant effects on intracellular factors involved in neuronal survival and demise. This study is the first to demonstrate a sex specific pattern of RORA protein and gene expression in the SNpc of controls post-mortem human brains, and to show that this is differentially altered in male and female PD subjects, thus supporting a role for RORA in sex-specific aspects of PD. Furthermore, our in vitro PD model indicates mechanisms whereby a RORA agonist exerts its neuroprotective effect, thereby highlighting the translational potential for RORA ligands in PD.
Collapse
Affiliation(s)
- Felwah S Al-Zaid
- Department of Physiology, College of Medicine, King Saud University, Riyadh, KSA, Saudi Arabia.
- Department of Brain Sciences, Imperial College London, London, W12 0NN, UK.
| | - Michael J Hurley
- Department of Brain Sciences, Imperial College London, London, W12 0NN, UK
- Department of Clinical and Movement Neuroscience, UCL Queen Square Institute of Neurology, Rowland Hill Street, London, NW3 2PF, UK
| | - David T Dexter
- Department of Brain Sciences, Imperial College London, London, W12 0NN, UK
- Parkinson's UK, 215 Vauxhall Bridge Road, London, SW1V 1EJ, UK
| | - Glenda E Gillies
- Department of Brain Sciences, Imperial College London, London, W12 0NN, UK
| |
Collapse
|
39
|
Rao SC, Li Y, Lapin B, Pattipati S, Ghosh Galvelis K, Naito A, Gutierrez N, Leal TP, Salim A, Salles PA, De Leon M, Mata IF. Association of women-specific health factors in the severity of Parkinson's disease. NPJ Parkinsons Dis 2023; 9:86. [PMID: 37277346 PMCID: PMC10241917 DOI: 10.1038/s41531-023-00524-x] [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: 10/17/2022] [Accepted: 05/15/2023] [Indexed: 06/07/2023] Open
Abstract
Parkinson's disease (PD) is an age-related neurological disorder known for the observational differences in its risk, progression, and severity between men and women. While estrogen has been considered to be a protective factor in the development of PD, there is little known about the role that fluctuations in hormones and immune responses from sex-specific health experiences have in the disease's development and severity. We sought to identify women-specific health experiences associated with PD severity, after adjusting for known PD factors, by developing and distributing a women-specific questionnaire across the United States and creating multivariable models for PD severity. We created a questionnaire that addresses women's specific experiences and their PD clinical history and deployed it through The Parkinson's Foundation: PD Generation. To determine the association between women-specific health factors and PD severity, we constructed multivariable logistic regression models based on the MDS-UPDRS scale and the participants' questionnaire responses, genetics, and clinical data. For our initial launch in November 2021, we had 304 complete responses from PD GENEration. Univariate and multivariate logistic modeling found significant associations between major depressive disorder, perinatal depression, natural childbirth, LRRK2 genotype, B12 deficiency, total hysterectomy, and increased PD severity. This study is a nationally available questionnaire for women's health and PD. It shifts the paradigm in understanding PD etiology and acknowledging how sex-specific experiences may contribute to PD severity. In addition, the work in this study sets the foundation for future research to investigate the factors behind sex differences in PD.
Collapse
Affiliation(s)
- Shilpa C Rao
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yadi Li
- Center for Outcomes Research and Evaluation, Cleveland Clinic, Cleveland, OH, USA
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Brittany Lapin
- Center for Outcomes Research and Evaluation, Cleveland Clinic, Cleveland, OH, USA
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Sreya Pattipati
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | | | | | | | - Amira Salim
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Philippe A Salles
- Center for Movement Disorders CETRAM, University of Santiago de Chile, Santiago, Chile
| | - Maria De Leon
- DefeatParkinsons, Houston, TX, USA
- De Leon Enterprises, Houston, TX, USA
| | - Ignacio F Mata
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
40
|
Jiang Z, Wang X, Zhang H, Yin J, Zhao P, Yin Q, Wang Z. Ketogenic diet protects MPTP-induced mouse model of Parkinson's disease via altering gut microbiota and metabolites. MedComm (Beijing) 2023; 4:e268. [PMID: 37200942 PMCID: PMC10186339 DOI: 10.1002/mco2.268] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 04/03/2023] [Accepted: 04/12/2023] [Indexed: 05/20/2023] Open
Abstract
The ketogenic diet (KD) is a low-carbohydrate, high-fat regime that is protective against neurodegenerative diseases. However, the impact of KD on Parkinson's disease (PD) and its mechanisms remains unclear. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD was fed with KD for 8 weeks. Motor function and dopaminergic neurons were evaluated. Inflammation in the brain, plasma, and colon tissue were also measured. Fecal samples were assessed by 16S rDNA gene sequencing and untargeted metabolomics. We found that KD protected motor dysfunction, dopaminergic neuron loss, and inflammation in an MPTP mouse model of PD. 16S rDNA sequencing revealed that MPTP administration significantly increased Citrobacter, Desulfovibrio, and Ruminococcus, and decreased Dubosiella, whereas KD treatment reversed the dysbiosis. Meanwhile, KD regulated the MPTP-induced histamine, N-acetylputrescine, d-aspartic acid, and other metabolites. Fecal microbiota transplantation using feces from the KD-treated mice attenuated the motor function impairment and dopaminergic neuron loss in antibiotic-pretreated PD mice. Our current study demonstrates that KD played a neuroprotective role in the MPTP mouse model of PD through the diet-gut microbiota-brain axis, which may involve inflammation in the brain and colon. However, future research is warranted to explore the explicit anti-inflammatory mechanisms of the gut-brain axis in PD models fed with KD.
Collapse
Affiliation(s)
- Ziying Jiang
- Department of Geriatric NeurologyThe Second Medical Center & National Clinical Research Center for Geriatric DiseaseChinese PLA General HospitalBeijingChina
| | - Xinyu Wang
- Department of Geriatric NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Haoqiang Zhang
- Department of EndocrinologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Jian Yin
- Department of Bio‐Medical DiagnosticsSuzhou Institute of Biomedical Engineering and TechnologyChinese Academy of SciencesSuzhouJiangsuChina
- Department of Bio‐Medical DiagnosticsJinan Guo Ke Medical Technology Development Co. Ltd.JinanShandongChina
| | - Peiqing Zhao
- Department of Translational Medical CenterZibo Central Hospital Affiliated to Binzhou Medical UniversityZiboShandongChina
| | - Qingqing Yin
- Department of Geriatric NeurologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
| | - Zhenfu Wang
- Department of Geriatric NeurologyThe Second Medical Center & National Clinical Research Center for Geriatric DiseaseChinese PLA General HospitalBeijingChina
| |
Collapse
|
41
|
Bourque M, Morissette M, Soulet D, Di Paolo T. Impact of Sex on Neuroimmune contributions to Parkinson's disease. Brain Res Bull 2023:110668. [PMID: 37196734 DOI: 10.1016/j.brainresbull.2023.110668] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/27/2023] [Accepted: 05/13/2023] [Indexed: 05/19/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease. Inflammation has been observed in both the idiopathic and familial forms of PD. Importantly, PD is reported more often in men than in women, men having at least 1.5- fold higher risk to develop PD than women. This review summarizes the impact of biological sex and sex hormones on the neuroimmune contributions to PD and its investigation in animal models of PD. Innate and peripheral immune systems participate in the brain neuroinflammation of PD patients and is reproduced in neurotoxin, genetic and alpha-synuclein based models of PD. Microglia and astrocytes are the main cells of the innate immune system in the central nervous system and are the first to react to restore homeostasis in the brain. Analysis of serum immunoprofiles in female and male control and PD patients show that a great proportion of these markers differ between male and female. The relationship between CSF inflammatory markers and PD clinical characteristics or PD biomarkers shows sex differences. Conversely, in animal models of PD, sex differences in inflammation are well documented and the beneficial effects of endogenous and exogenous estrogenic modulation in inflammation have been reported. Targeting neuroinflammation in PD is an emerging therapeutic option but gonadal drugs have not yet been investigated in this respect, thus offering new opportunities for sex specific treatments.
Collapse
Affiliation(s)
- Mélanie Bourque
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada.
| | - Marc Morissette
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada.
| | - Denis Soulet
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada; Faculté de Pharmacie, Pavillon Ferdinand-Vandry, 1050, avenue de la Médecine, Université Laval, Québec (Québec) G1V 0A6, Canada.
| | - Thérèse Di Paolo
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada; Faculté de Pharmacie, Pavillon Ferdinand-Vandry, 1050, avenue de la Médecine, Université Laval, Québec (Québec) G1V 0A6, Canada.
| |
Collapse
|
42
|
Vila-Castelar C, Udeh-Momoh C, Aggarwal NT, Mielke MM. Sex and gender considerations in dementia: a call for global research. NATURE AGING 2023; 3:463-465. [PMID: 37202511 PMCID: PMC10331726 DOI: 10.1038/s43587-023-00374-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Studies have identified sex and/or gender differences in Alzheimer’s disease, but few have examined other dementias. We highlight sex and gender differences in other dementias, discuss sociocultural factors and provide a framework for future global studies.
Collapse
Affiliation(s)
- Clara Vila-Castelar
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Chinedu Udeh-Momoh
- Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, UK
- Department of Neurobiology, Care Sciences and Society (NVS), Division of Clinical Geriatrics, Karolinska Institute, Stockholm, Sweden
- Global Brain Health Institute (GBHI), University of California, San Francisco, CA, USA
| | - Neelum T Aggarwal
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Michelle M Mielke
- Department of Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
43
|
De Luca R, Bonanno M, Morini E, Marra A, Arcadi FA, Quartarone A, Calabrò RS. Sexual Dysfunctions in Females with Parkinson's Disease: A Cross-Sectional Study with a Psycho-Endocrinological Perspective. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59050845. [PMID: 37241076 DOI: 10.3390/medicina59050845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023]
Abstract
Background and Objectives: Normal human sexual functioning is a complex integration of an intact neuroanatomic substrate, vascular supply, a balanced hormonal profile, and a predominance of excitatory over inhibitory psychological mechanisms. However, sexual functioning in Parkinson's disease (PD) is often overlooked in clinical practice, especially in female patients. Materials and Methods: In this cross-sectional study, we have investigated the frequency of sexual dysfunction and the possible correlation with psycho-endocrinological factors in a sample of women with idiopathic PD. Patients were assessed using a semi-structured sexual interview, in addition to psychometric tools, including the Hamilton Rating Scale for Anxiety and for Depression and the Coping Orientation to the Problems Experiences-New Italian Version. Specific blood tests, including testosterone, follicle-stimulating hormone (FSH), luteinizing hormone (LH), estrogen E2, prolactin (PRL), and vitamin D3 were also evaluated. Results: Our results reported a statistical difference in sexual intercourse frequency before and after the onset of PD (p < 0.001). The percentage of women who complained about reduced sexual desire increased after diagnosis (52.7%) compared to the period before the onset of the illness (36.8%). The endocrinological profile in females with PD revealed statistically significant differences regarding testosterone (p < 0.0006), estradiol (p < 0.00), vitamin D3 (p < 0.006), and calcium (0.002). Depression (44% characterized by perceived feelings of anger and frustration during sexual intercourse) and anxiety symptoms (29.5% reported feelings of fear and anxiety for not satisfying the partner) with abnormal coping strategies (48.14% experienced feelings of anger and intolerance) were also found to be statistically significant. This study showed a high frequency of sexual dysfunction in female patients with PD, which correlated with sexual hormone abnormalities, mood/anxiety, and coping strategies alterations. This supports the idea that there is a need to better investigate the sexual function of female patients with PD to provide them with an adequate therapeutic approach and potentially improve quality of life.
Collapse
Affiliation(s)
- Rosaria De Luca
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Palermo, SS 113, C. da Casazza, 98123 Messina, Italy
| | - Mirjam Bonanno
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Palermo, SS 113, C. da Casazza, 98123 Messina, Italy
| | - Elisabetta Morini
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Palermo, SS 113, C. da Casazza, 98123 Messina, Italy
| | - Angela Marra
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Palermo, SS 113, C. da Casazza, 98123 Messina, Italy
| | - Francesca Antonia Arcadi
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Palermo, SS 113, C. da Casazza, 98123 Messina, Italy
| | - Angelo Quartarone
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Palermo, SS 113, C. da Casazza, 98123 Messina, Italy
| | - Rocco Salvatore Calabrò
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Palermo, SS 113, C. da Casazza, 98123 Messina, Italy
| |
Collapse
|
44
|
Gamage HKAH, Robinson KJ, Luu L, Paulsen IT, Laird AS. Machado Joseph disease severity is linked with gut microbiota alterations in transgenic mice. Neurobiol Dis 2023; 179:106051. [PMID: 36822548 DOI: 10.1016/j.nbd.2023.106051] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/06/2023] [Accepted: 02/18/2023] [Indexed: 02/23/2023] Open
Abstract
Emerging evidence suggests the presence of bidirectional interactions between the central nervous system and gut microbiota that may contribute to the pathogenesis of neurodegenerative diseases. However, the potential role of gut microbes in forms of spinocerebellar ataxia, such as the fatal neurodegenerative disease Machado Joseph disease (MJD), remains unexplored. Here, we examined whether gut microbiota alterations may be an early disease phenotype of MJD. We profiled the gut microbiota of male and female transgenic MJD mice (CMVMJD135) expressing human ATXN3 with expanded CAG repeats (133-143 CAG) at pre-symptomatic, symptomatic and well-established stages of the disease (7, 11 and 15 weeks of age, respectively). We compared these profiles with the gut microbiota of male and female wild-type (WT) littermate control mice at same ages. Correlation network analyses were employed to explore the relevance of microbiota changes to disease progression. The results demontrated distinct sex-dependent effects in disease development whereby male MJD mice displayed earlier motor impairments than female MJD mice. The gut microbiota community structure and composition also demonstrated sex-specific differences between MJD and WT mice. In both male and female MJD mice, the shifts in the microbiota were present by 7 weeks, before the onset of any symptoms. These pre-symptomatic microbial changes correlated with the severity of neurological impairments present at later stages of the disease. Previous efforts towards developing treatments for MJD have failed to yield meaningful outcomes. Our study reports a novel relationship between the gut microbiota and MJD development and severity. Elucidating how gut microbes are involved in MJD pathogenesis may offer new and efficacious treatment strategies for this currently untreatable disease.
Collapse
Affiliation(s)
- Hasinika K A H Gamage
- School of Natural Sciences, Macquarie University, NSW 2109, Australia; ARC Training Centre for Facilitated Advancement of Australia's Bioactives, Macquarie University, NSW 2109, Australia
| | - Katherine J Robinson
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW 2109, Australia
| | - Luan Luu
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW 2109, Australia
| | - Ian T Paulsen
- School of Natural Sciences, Macquarie University, NSW 2109, Australia; ARC Training Centre for Facilitated Advancement of Australia's Bioactives, Macquarie University, NSW 2109, Australia; ARC Centre of Excellence in Synthetic Biology, Macquarie University, NSW 2109, Australia
| | - Angela S Laird
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW 2109, Australia.
| |
Collapse
|
45
|
Bianco A, Antonacci Y, Liguori M. Sex and Gender Differences in Neurodegenerative Diseases: Challenges for Therapeutic Opportunities. Int J Mol Sci 2023; 24:6354. [PMID: 37047320 PMCID: PMC10093984 DOI: 10.3390/ijms24076354] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
The term "neurodegenerative diseases" (NDs) identifies a group of heterogeneous diseases characterized by progressive loss of selectively vulnerable populations of neurons, which progressively deteriorates over time, leading to neuronal dysfunction. Protein aggregation and neuronal loss have been considered the most characteristic hallmarks of NDs, but growing evidence confirms that significant dysregulation of innate immune pathways plays a crucial role as well. NDs vary from multiple sclerosis, in which the autoimmune inflammatory component is predominant, to more "classical" NDs, such as Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, and spinal muscular atrophy. Of interest, many of the clinical differences reported in NDs seem to be closely linked to sex, which may be justified by the significant changes in immune mechanisms between affected females and males. In this review, we examined some of the most studied NDs by looking at their pathogenic and phenotypical features to highlight sex-related discrepancies, if any, with particular interest in the individuals' responses to treatment. We believe that pointing out these differences in clinical practice may help achieve more successful precision and personalized care.
Collapse
Affiliation(s)
| | | | - Maria Liguori
- National Research Council (CNR), Institute of Biomedical Technologies, Bari Unit, 70125 Bari, Italy
| |
Collapse
|
46
|
Arazi H, Birak Olia RB, Eghbali E. Are the digit ratio (2D:4D) and hand grip strength related to Parkinson disease in elderly males? BMC Sports Sci Med Rehabil 2023; 15:34. [PMID: 36941653 PMCID: PMC10026433 DOI: 10.1186/s13102-023-00642-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 03/09/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND Sex hormones affect the pathogenesis of Parkinson patients and it has been suggested that gender is the most important factor in the development and progression of Parkinson's disease. Studies have shown that the second to fourth digit ratio (2D:4D) is affected by the prenatal testosterone and estrogen levels and can predict predisposition to disease. In addition, decreased muscle strength in people with Parkinson's has been repeatedly reported. Hand grip strength (HGS) is a suitable measure to evaluate the musculoskeletal system among the elderly and it is considered as an indicator of the overall strength of the body. This study aimed at investigating the relationship between Parkinson's disease and HGS and 2D:4D ratio. METHODS In this study 117 elderly men with Parkinson disease (mean age of 61.66 ± 11.28 years) and 156 healthy control subjects (mean age of 61.86 ± 6.29 years) participated. After determining the level of disability of Parkinson patients by a neurologist (level of disability in the range of 1-4), anthropometric indices (height, weight, length of the second and fourth fingers) and maximum HGS were measured. RESULTS Although 2D:4D ratios (right and left hand) of male patients with Parkinson's disease were higher than those of healthy males, this difference was not statistically significant (P = 0.12, P = 0.40; respectively). Conversely, HGS for the right and left hands of Parkinson patients were significantly lower than those of healthy males (P = 0.02, P = 0.03; respectively). The results showed a significant negative relationship between Parkinson disease and the right and left HGS (R = -0.16, P = 0.005; R = -0.17, P = 0.003; respectively). Parkinson disease had no significant relationship with 2D:4D of the right hand, left hand, mean finger ratio and DR-L 2D:4D (P > 0.05). The regression results showed that the right and left HGS were not able to predict Parkinson disease (P = 0.25, P = 0.16; respectively). CONCLUSION We concluded that HGS was negatively associated with the Parkinson disease, but conversely, 2D:4D may not be a valuable biomarker of elevated risk of Parkinson in elderly males.
Collapse
Affiliation(s)
- Hamid Arazi
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, P.O. Box: 41635-1438, Rasht, Iran.
| | - Roghayeh Bavafa Birak Olia
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, P.O. Box: 41635-1438, Rasht, Iran
| | - Ehsan Eghbali
- Department of Exercise Physiology, Faculty of Sport Sciences, University of Guilan, P.O. Box: 41635-1438, Rasht, Iran
| |
Collapse
|
47
|
Cartas-Cejudo P, Lachén-Montes M, Ferrer I, Fernández-Irigoyen J, Santamaría E. Sex-divergent effects on the NAD+-dependent deacetylase sirtuin signaling across the olfactory-entorhinal-amygdaloid axis in Alzheimer's and Parkinson's diseases. Biol Sex Differ 2023; 14:5. [PMID: 36755296 PMCID: PMC9906849 DOI: 10.1186/s13293-023-00487-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/16/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Smell impairment is one of the earliest features in Alzheimer's (AD) and Parkinson's diseases (PD). Due to sex differences exist in terms of smell and olfactory structures as well as in the prevalence and manifestation of both neurological syndromes, we have applied olfactory proteomics to favor the discovery of novel sex-biased physio-pathological mechanisms and potential therapeutic targets associated with olfactory dysfunction. METHODS SWATH-MS (sequential window acquisition of all theoretical fragment ion spectra mass spectrometry) and bioinformatic workflows were applied in 57 post-mortem olfactory tracts (OT) derived from controls with no known neurological history (n = 6F/11M), AD (n = 4F/13M) and PD (n = 7F/16M) subjects. Complementary molecular analyses by Western-blotting were performed in the olfactory bulb (OB), entorhinal cortex (EC) and amygdala areas. RESULTS 327 and 151 OT differentially expressed proteins (DEPs) were observed in AD women and AD men, respectively (35 DEPs in common). With respect to PD, 198 DEPs were identified in PD women, whereas 95 DEPs were detected in PD men (20 DEPs in common). This proteome dyshomeostasis induced a disruption in OT protein interaction networks and widespread sex-dependent pathway perturbations in a disease-specific manner, among them Sirtuin (SIRT) signaling. SIRT1, SIRT2, SIRT3 and SIRT5 protein levels unveiled a tangled expression profile across the olfactory-entorhinal-amygdaloid axis, evidencing disease-, sex- and brain structure-dependent changes in olfactory protein acetylation. CONCLUSIONS Alteration in the OT proteostasis was more severe in AD than in PD. Moreover, protein expression changes were more abundant in women than men independent of the neurological syndrome. Mechanistically, the tangled SIRT profile observed across the olfactory pathway-associated brain regions in AD and PD indicates differential NAD (+)-dependent deacetylase mechanisms between women and men. All these data shed new light on differential olfactory mechanisms across AD and PD, pointing out that the evaluation of the feasibility of emerging sirtuin-based therapies against neurodegenerative diseases should be considered with caution, including further sex dimension analyses in vivo and in clinical studies.
Collapse
Affiliation(s)
- Paz Cartas-Cejudo
- grid.410476.00000 0001 2174 6440Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospitalario Universitario de Navarra (HUN), IdiSNA, Navarra Institute for Health Research, Universidad Pública de Navarra (UPNA), Irunlarrea 3, 31008 Pamplona, Spain
| | - Mercedes Lachén-Montes
- grid.410476.00000 0001 2174 6440Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospitalario Universitario de Navarra (HUN), IdiSNA, Navarra Institute for Health Research, Universidad Pública de Navarra (UPNA), Irunlarrea 3, 31008 Pamplona, Spain
| | - Isidro Ferrer
- grid.5841.80000 0004 1937 0247Department of Pathology and Experimental Therapeutics, CIBERNED (Network Centre of Biomedical Research of Neurodegenerative Diseases), Bellvitge University Hospital/Bellvitge Biomedical Research Institute (IDIBELL), Institute of Health Carlos III, University of Barcelona, Hospitalet de Llobregat, Barcelona, Spain
| | - Joaquín Fernández-Irigoyen
- grid.410476.00000 0001 2174 6440Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospitalario Universitario de Navarra (HUN), IdiSNA, Navarra Institute for Health Research, Universidad Pública de Navarra (UPNA), Irunlarrea 3, 31008 Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Unit, Proteomics Platform, Navarrabiomed, Hospitalario Universitario de Navarra (HUN), IdiSNA, Navarra Institute for Health Research, Universidad Pública de Navarra (UPNA), Irunlarrea 3, 31008, Pamplona, Spain.
| |
Collapse
|
48
|
Chiu SY, Wyman-Chick KA, Ferman TJ, Bayram E, Holden SK, Choudhury P, Armstrong MJ. Sex differences in dementia with Lewy bodies: Focused review of available evidence and future directions. Parkinsonism Relat Disord 2023; 107:105285. [PMID: 36682958 PMCID: PMC10024862 DOI: 10.1016/j.parkreldis.2023.105285] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/07/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023]
Abstract
In this review, we summarize the current knowledge on sex differences in dementia with Lewy bodies (DLB) relating to epidemiology, clinical features, neuropathology, biomarkers, disease progression, and caregiving. While many studies show a higher DLB prevalence in men, this finding is inconsistent and varies by study approach. Visual hallucinations may be more common and occur earlier in women with DLB, whereas REM sleep behavior disorder may be more common and occur earlier in men. Several studies report a higher frequency of parkinsonism in men with DLB, while the frequency of fluctuations appears similar between sexes. Women tend to be older, have greater cognitive impairment at their initial visit, and are delayed in meeting DLB criteria compared to men. Women are also more likely to have Lewy body disease with co-existing AD-related pathology than so-called "pure" Lewy body disease, while men may present with either. Research is mixed regarding the impact of sex on DLB progression. Biomarker and treatment research assessing for sex differences is lacking. Women provide the majority of caregiving in DLB but how this affects the caregiving experience is uncertain. Gaining a better understanding of sex differences will be instrumental in aiding future development of sex-specific strategies in DLB for early diagnosis, care, and drug development.
Collapse
Affiliation(s)
- Shannon Y Chiu
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL, USA; Norman Fixel Institute for Neurologic Diseases, University of Florida, Gainesville, FL, USA.
| | - Kathryn A Wyman-Chick
- Center for Memory and Aging, Department of Neurology, HealthPartners, Saint Paul, MN, USA
| | - Tanis J Ferman
- Department of Psychiatry & Psychology, Mayo Clinic, Jacksonville, FL, USA
| | - Ece Bayram
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Samantha K Holden
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Parichita Choudhury
- Cleo Roberts Center, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Melissa J Armstrong
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL, USA; Norman Fixel Institute for Neurologic Diseases, University of Florida, Gainesville, FL, USA
| |
Collapse
|
49
|
Gudkov SV, Burmistrov DE, Kondakova EV, Sarimov RM, Yarkov RS, Franceschi C, Vedunova MV. An emerging role of astrocytes in aging/neuroinflammation and gut-brain axis with consequences on sleep and sleep disorders. Ageing Res Rev 2023; 83:101775. [PMID: 36334910 DOI: 10.1016/j.arr.2022.101775] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 10/05/2022] [Accepted: 10/30/2022] [Indexed: 11/18/2022]
Abstract
Understanding the role of astrocytes in the central nervous system has changed dramatically over the last decade. The accumulating findings indicate that glial cells are involved not only in the maintenance of metabolic and ionic homeostasis and in the implementation of trophic functions but also in cognitive functions and information processing in the brain. Currently, there are some controversies regarding the role of astrocytes in complex processes such as aging of the nervous system and the pathogenesis of age-related neurodegenerative diseases. Many findings confirm the important functional role of astrocytes in age-related brain changes, including sleep disturbance and the development of neurodegenerative diseases and particularly Alzheimer's disease. Until recent years, neurobiological research has focused mainly on neuron-glial interactions, in which individual astrocytes locally modulate neuronal activity and communication between neurons. The review considers the role of astrocytes in the physiology of sleep and as an important "player" in the development of neurodegenerative diseases. In addition, the features of the astrocytic network reorganization during aging are discussed.
Collapse
Affiliation(s)
- Sergey V Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia; Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Dmitriy E Burmistrov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Elena V Kondakova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Ruslan M Sarimov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilov str., 119991 Moscow, Russia.
| | - Roman S Yarkov
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Claudio Franceschi
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| | - Maria V Vedunova
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin ave., 603022 Nizhny Novgorod, Russia.
| |
Collapse
|
50
|
Ponsoni A, Sardeli AV, Costa FP, Mourão LF. Prevalence of sarcopenia in Parkinson's disease: A systematic review and meta-analysis. Geriatr Nurs 2023; 49:44-49. [PMID: 36413812 DOI: 10.1016/j.gerinurse.2022.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/21/2022]
Abstract
We meta-analysed the sarcopenia prevalence among patients with Parkinson´s disease (PD) in comparison to a control group and tested the effects of age, sex, sarcopenia assessments, and PD progression in the sarcopenia prevalence. The literature search was performed using five databases in March 2022. The prevalence of sarcopenia in patients with PD was 3 times higher than in the control group (OR 3.98). Subgroup analyses showed that among individuals aged ≥ 70 years the higher prevalence of sarcopenia in PD compared to controls (OR 5.32, P=0.08) tended to be higher (P=0.08) than the group < 70 years. Regarding PD progression, the prevalence of sarcopenia was not different between individuals scoring < 2.5 and ≥ 2.5 in the Hoehn and Yahr scale. Patients with PD have a higher probability of developing sarcopenia when compared with the control group and older PD patients trended to have even higher chance of sarcopenia than their older controls.
Collapse
Affiliation(s)
- Adriana Ponsoni
- Gerontology program, Faculty of Medical Sciences, University of Campinas - UNICAMP, Campinas, Brazil.
| | - Amanda Veiga Sardeli
- Gerontology program, Faculty of Medical Sciences, University of Campinas - UNICAMP, Campinas, Brazil; Laboratory of Exercise Physiology - FISEX, University of Campinas - UNICAMP, Campinas, Brazil; Institute of Inflammageing and Ageing, University of Birmingham, Birmingham, UK
| | - Flavia Pereira Costa
- Gerontology program, Faculty of Medical Sciences, University of Campinas - UNICAMP, Campinas, Brazil
| | - Lucia Figueiredo Mourão
- Gerontology program, Faculty of Medical Sciences, University of Campinas - UNICAMP, Campinas, Brazil
| |
Collapse
|