1
|
Pang S, Ren Z, Ding H, Chan P. Short-chain fatty acids mediate enteric and central nervous system homeostasis in Parkinson's disease: Innovative therapies and their translation. Neural Regen Res 2026; 21:938-956. [PMID: 40313087 DOI: 10.4103/nrr.nrr-d-24-01265] [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: 10/18/2024] [Accepted: 01/16/2025] [Indexed: 05/03/2025] Open
Abstract
Short-chain fatty acids, metabolites produced by the fermentation of dietary fiber by gut microbiota, have garnered significant attention due to their correlation with neurodegenerative diseases, particularly Parkinson's disease. In this review, we summarize the changes in short-chain fatty acid levels and the abundance of short-chain fatty acid-producing bacteria in various samples from patients with Parkinson's disease, highlighting the critical role of gut homeostasis imbalance in the pathogenesis and progression of the disease. Focusing on the nervous system, we discuss the molecular mechanisms by which short-chain fatty acids influence the homeostasis of both the enteric nervous system and the central nervous system. We identify key processes, including the activation of G protein-coupled receptors and the inhibition of histone deacetylases by short-chain fatty acids. Importantly, structural or functional disruptions in the enteric nervous system mediated by these fatty acids may lead to abnormal α-synuclein expression and gastrointestinal dysmotility, which could serve as an initiating event in Parkinson's disease. Furthermore, we propose that short-chain fatty acids help establish communication between the enteric nervous system and the central nervous system via the vagal nerve, immune circulation, and endocrine signaling. This communication may shed light on their potential role in the transmission of α-synuclein from the gut to the brain. Finally, we elucidate novel treatment strategies for Parkinson's disease that target short-chain fatty acids and examine the challenges associated with translating short-chain fatty acid-based therapies into clinical practice. In conclusion, this review emphasizes the pivotal role of short-chain fatty acids in regulating gut-brain axis integrity and their significance in the pathogenesis of Parkinson's disease from the perspective of the nervous system. Moreover, it highlights the potential value of short-chain fatty acids in early intervention for Parkinson's disease. Future research into the molecular mechanisms of short-chain fatty acids and their synergistic interactions with other gut metabolites is likely to advance the clinical translation of innovative short-chain fatty acid-based therapies for Parkinson's disease.
Collapse
Affiliation(s)
- Shimin Pang
- Department of Neurobiology and Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zhili Ren
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Hui Ding
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Piu Chan
- Department of Neurobiology and Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, China
- Clinical and Research Center for Parkinson's Disease, Xuanwu Hospital of Capital Medical University, Beijing, China
| |
Collapse
|
2
|
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
|
3
|
Bellini G, D'Antongiovanni V, Palermo G, Antonioli L, Fornai M, Ceravolo R, Bernardini N, Derkinderen P, Pellegrini C. α-Synuclein in Parkinson's Disease: From Bench to Bedside. Med Res Rev 2025; 45:909-946. [PMID: 39704040 PMCID: PMC11976381 DOI: 10.1002/med.22091] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/24/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024]
Abstract
α-Synuclein (α-syn), a pathological hallmark of PD, is emerging as a bridging element at the crossroads between neuro/immune-inflammatory responses and neurodegeneration in PD. Several evidence show that pathological α-syn accumulates in neuronal and non-neuronal cells (i.e., neurons, microglia, macrophages, skin cells, and intestinal cells) in central and peripheral tissues since the prodromal phase of the disease, contributing to brain pathology. Indeed, pathological α-syn deposition can promote neurogenic/immune-inflammatory responses that contribute to systemic and central neuroinflammation associated with PD. After providing an overview of the structure and functions of physiological α-syn as well as its pathological forms, we review current studies about the role of neuronal and non-neuronal α-syn at the crossroads between neuroinflammation and neurodegeneration in PD. In addition, we provide an overview of the correlation between the accumulation of α-syn in central and peripheral tissues and PD, related symptoms, and neuroinflammation. Special attention was paid to discussing whether targeting α-syn can represent a suitable therapeutical approach for PD.
Collapse
Affiliation(s)
- Gabriele Bellini
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
- Department of NeurologyThe Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, NYU Langone HealthNew York CityNew YorkUSA
| | - Vanessa D'Antongiovanni
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Giovanni Palermo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Roberto Ceravolo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Nunzia Bernardini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| | - Pascal Derkinderen
- Department of NeurologyNantes Université, CHU Nantes, INSERMNantesFrance
| | - Carolina Pellegrini
- Unit of Histology and Embryology, Department of Clinical and Experimental MedicineUniversity of PisaPisaItaly
| |
Collapse
|
4
|
Agarwal C, Koerner IP. Crosstalk between brain and the gastrointestinal system. BRAIN AND ORGAN COMMUNICATION 2025:195-213. [DOI: 10.1016/b978-0-443-22268-9.00011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
5
|
Mu L, Chen J, Li J, Nyirenda T, Hegland KW, Beach TG. Mechanisms of Swallowing, Speech and Voice Disorders in Parkinson's Disease: Literature Review with Our First Evidence for the Periperal Nervous System Involvement. Dysphagia 2024; 39:1001-1012. [PMID: 38498201 DOI: 10.1007/s00455-024-10693-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 02/29/2024] [Indexed: 03/20/2024]
Abstract
The majority of patients with Parkinson's disease (PD) develop swallowing, speech, and voice (SSV) disorders. Importantly, swallowing difficulty or dysphagia and related aspiration are life-threatening conditions for PD patients. Although PD treatments have significant therapeutic effects on limb motor function, their effects on SSV disorders are less impressive. A large gap in our knowledge is that the mechanisms of SSV disorders in PD are poorly understood. PD was long considered to be a central nervous system disorder caused by the death of dopaminergic neurons in the basal ganglia. Aggregates of phosphorylated α-synuclein (PAS) underlie PD pathology. SSV disorders were thought to be caused by the same dopaminergic problem as those causing impaired limb movement; however, there is little evidence to support this. The pharynx, larynx, and tongue play a critical role in performing upper airway (UA) motor tasks and their dysfunction results in disordered SSV. This review aims to provide an overview on the neuromuscular organization patterns, functions of the UA structures, clinical features of SSV disorders, and gaps in knowledge regarding the pathophysiology underlying SSV disorders in PD, and evidence supporting the hypothesis that SSV disorders in PD could be associated, at least in part, with PAS damage to the peripheral nervous system controlling the UA structures. Determining the presence and distribution of PAS lesions in the pharynx, larynx, and tongue will facilitate the identification of peripheral therapeutic targets and set a foundation for the development of new therapies to treat SSV disorders in PD.
Collapse
Affiliation(s)
- Liancai Mu
- Upper Airway Reserch Laboratory, Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ, 07110, USA.
- Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ, 07110, USA.
| | - Jingming Chen
- Upper Airway Reserch Laboratory, Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ, 07110, USA
| | - Jing Li
- Upper Airway Reserch Laboratory, Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ, 07110, USA
| | - Themba Nyirenda
- Upper Airway Reserch Laboratory, Center for Discovery and Innovation, Hackensack Meridian Health, 111 Ideation Way, Nutley, NJ, 07110, USA
| | - Karen Wheeler Hegland
- Upper Airway Dysfunction Laboratory, M.A. Program in Communication Sciences & Disorders, Department of Speech, Language and Hearing Sciences, College of Public Health and Health Professions, University of Florida, 1225 Center Dr., Gainesville, FL, 32611, USA
| | - Thomas G Beach
- Director of Neuroscience, Director of Brain and Body Donation Program, Banner Sun Health Research Institute, 10515 West Santa Fe Dr, Sun City, AZ, 85351, USA
| |
Collapse
|
6
|
Xiang J, Tang J, Kang F, Ye J, Cui Y, Zhang Z, Wang J, Wu S, Ye K. Gut-induced alpha-Synuclein and Tau propagation initiate Parkinson's and Alzheimer's disease co-pathology and behavior impairments. Neuron 2024; 112:3585-3601.e5. [PMID: 39241780 DOI: 10.1016/j.neuron.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 05/30/2024] [Accepted: 08/07/2024] [Indexed: 09/09/2024]
Abstract
Tau interacts with α-Synuclein (α-Syn) and co-localizes with it in the Lewy bodies, influencing α-Syn pathology in Parkinson's disease (PD). However, whether these biochemical events regulate α-Syn pathology spreading from the gut into the brain remains incompletely understood. Here, we show that α-Syn and Tau co-pathology is spread into the brain in gut-inducible SYN103+/- and/or TAU368+/- transgenic mouse models, eliciting behavioral defects. Gut pathology was initially observed, and α-Syn or Tau pathology was subsequently propagated into the DMV or NTS and then to other brain regions. Remarkably, more extensive spreading and widespread neuronal loss were found in double transgenic mice (Both) than in single transgenic mice. Truncal vagotomy and α-Syn deficiency significantly inhibited synucleinopathy or tauopathy spreading. The α-Syn PET tracer [18F]-F0502B detected α-Syn aggregates in the gut and brain. Thus, α-Syn and Tau co-pathology can propagate from the gut to the brain, triggering behavioral disorders.
Collapse
Affiliation(s)
- Jie Xiang
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| | - Jingrong Tang
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China
| | - Fei Kang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jiajun Ye
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Yueying Cui
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Shengxi Wu
- Department of Neurobiology, Fourth Military Medical University, Xi'an 710032, China.
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology (SUAT), Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
7
|
Shin C, Kim SI, Park SH, Kim JM, Lee JY, Chung SJ, Kim JW, Ahn TB, Park KW, Shin JH, Lee CY, Lee HJ, Kong SH, Suh YS, Kim HJ, Yang HK, Jeon B. Diagnostic accuracy and predictors of alpha-synuclein accumulation in the gastrointestinal tract of Parkinson's disease. NPJ Parkinsons Dis 2024; 10:155. [PMID: 39147801 PMCID: PMC11327357 DOI: 10.1038/s41531-024-00766-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/29/2024] [Indexed: 08/17/2024] Open
Abstract
The only characteristic of alpha-synuclein (AS) accumulation in the gastrointestinal (GI) tract of Parkinson's disease (PD) found in pathological studies is the "rostrocaudal gradient," which describes the more frequent presence of AS accumulation in the upper GI tract than in the lower GI tract. This study aimed to determine the diagnostic accuracy and identify predictors of AS accumulation in the GI tract of PD patients. The frequency of AS accumulation in the GI tract was compared between PD patients (N = 97) who underwent radical GI surgery for cancer and individually matched controls (N = 94). We evaluated AS accumulation in the neural structures using phosphorylated AS immunohistochemistry. A multivariable logistic regression analysis was conducted to determine the predictors of AS accumulation in the GI tract of PD patients. The frequency of AS accumulation was significantly higher in PD patients (75.3%) than in controls (8.5%, p-value < 0.001). The sensitivity and specificity of the full-layer evaluation were 75.3% and 91.5%, respectively. When the evaluation was confined to the mucosal/submucosal layer, the sensitivity and specificity were 46.9% and 94.7%, respectively. The rostrocaudal gradient of AS accumulation was found in PD patients. The duration from symptom onset to surgery was significantly longer in PD patients with AS accumulation (4.9 ± 4.9 years) than in PD patients without AS accumulation (1.8 ± 4.1 years, p-value = 0.005). Both disease duration and rostrocaudal gradient independently predicted the presence of AS accumulation in the GI tract of PD patients. Our study suggests PD-related AS accumulation in the GI tract follows a temporally increasing but spatially static progression pattern.
Collapse
Affiliation(s)
- Chaewon Shin
- Department of Neurology, Chungnam National University Sejong Hospital, Chungnam National University College of Medicine, 20, Bodeum 7-ro, Sejong-si, Republic of Korea
| | - Seong-Ik Kim
- Department of Pathology, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Sung-Hye Park
- Department of Pathology, College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Jong-Min Kim
- Department of Neurology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, 173 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jee-Young Lee
- Department of Neurology, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-gu, 07061, Seoul, Republic of Korea
| | - Sun Ju Chung
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea
| | - Jae Woo Kim
- Department of Neurology, Dong-A University Hospital, 26 Daesingongwon-ro, Seo-gu, Busan, Republic of Korea
| | - Tae-Beom Ahn
- Department of Neurology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, 23 Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea
| | - Kye Won Park
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea
- Pacific Parkinson Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Jung Hwan Shin
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Chan Young Lee
- Department of Neurology, Ewha Womans University Mokdong Hospital, 1071 Annyangcheon-ro, Yangcheon-gu, Seoul, Republic of Korea
| | - Hyuk-Joon Lee
- Department of Surgery, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Seong-Ho Kong
- Department of Surgery, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University Bundang Hospital, 82 Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Han-Joon Kim
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Han-Kwang Yang
- Department of Surgery, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, Republic of Korea
| | - Beomseok Jeon
- Department of Neurology, Chung-ang University Health Care System Hyundae Hospital, Namyangju-si, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
8
|
Seo MH, Kim SH, Yeo S. Serping1 associated with α-synuclein increase in colonic smooth muscles of MPTP-induced Parkinson's disease mice. Sci Rep 2024; 14:1140. [PMID: 38212417 PMCID: PMC10784473 DOI: 10.1038/s41598-024-51770-9] [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: 09/25/2023] [Accepted: 01/09/2024] [Indexed: 01/13/2024] Open
Abstract
Patients with Parkinson's disease (PD) have gastrointestinal motility disorders, which are common non-motor symptoms. However, the reasons for these motility disorders remain unclear. Increased alpha-synuclein (α-syn) is considered an important factor in peristalsis dysfunction in colonic smooth muscles in patients with PD. In this study, the morphological changes and association between serping1 and α-syn were investigated in the colon of the 1-methyl 4-phenyl 1,2,3,6-tetrahydropyridine-induced chronic PD model. Increased serping1 and α-syn were noted in the colon of the PD model, and decreased serping1 also induced a decrease in α-syn in C2C12 cells. Serping1 is a major regulator of physiological processes in the kallikrein-kinin system, controlling processes including inflammation and vasodilation. The kinin system also comprises bradykinin and bradykinin receptor 1. The factors related to the kallikrein-kinin system, bradykinin, and bradykinin receptor 1 were regulated by serping1 in C2C12 cells. The expression levels of bradykinin and bradykinin receptor 1, modulated by serping1 also increased in the colon of the PD model. These results suggest that the regulation of increased serping1 could alleviate Lewy-type α-synucleinopathy, a characteristic of PD. Furthermore, this study could have a positive effect on the early stages of PD progression because of the perception that α-syn in colonic tissues is present prior to the development of PD motor symptoms.
Collapse
Affiliation(s)
- Min Hyung Seo
- Department of Meridian and Acupoint, College of Korean Medicine, Sang Ji University, Wonju, 26339, Republic of Korea
- Division of Biological Science and Technology, Yonsei University, Yonseidae 1 Gil, Wonju, 26493, Republic of Korea
| | - Soo-Hwan Kim
- Division of Biological Science and Technology, Yonsei University, Yonseidae 1 Gil, Wonju, 26493, Republic of Korea.
| | - Sujung Yeo
- Research Institute of Korean Medicine, College of Korean Medicine, Sangji University, Wonju, 26339, Republic of Korea.
| |
Collapse
|
9
|
Yang Y, Stewart T, Zhang C, Wang P, Xu Z, Jin J, Huang Y, Liu Z, Lan G, Liang X, Sheng L, Shi M, Cai Z, Zhang J. Erythrocytic α-Synuclein and the Gut Microbiome: Kindling of the Gut-Brain Axis in Parkinson's Disease. Mov Disord 2024; 39:40-52. [PMID: 37798868 DOI: 10.1002/mds.29620] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/29/2023] [Accepted: 08/31/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND Progressive spreading of α-synuclein via gut-brain axis has been hypothesized in the pathogenesis of Parkinson's disease (PD). However, the source of seeding-capable α-synuclein in the gastrointestinal tract (GIT) has not been fully investigated. Additionally, the mechanism by which the GIT microbiome contributes to PD pathogenesis remains to be characterized. OBJECTIVES We aimed to investigate whether blood-derived α-synuclein might contribute to PD pathology via a gut-driven pathway and involve GIT microbiota. METHODS The GIT expression of α-synuclein and the transmission of extracellular vesicles (EVs) derived from erythrocytes/red blood cells (RBCs), with their cargo α-synuclein, to the GIT were explored with various methods, including radioactive labeling of RBC-EVs and direct analysis of the transfer of α-synuclein protein. The potential role of microbiota on the EVs transmission was further investigated by administering butyrate, the short-chain fatty acids produced by gut microbiota and studying mice with different α-synuclein genotypes. RESULTS This study demonstrated that RBC-EVs can effectively transport α-synuclein to the GIT in a region-dependent manner, along with variations closely associated with regional differences in the expression of gut-vascular barrier markers. The investigation further revealed that the infiltration of α-synuclein into the GIT was influenced significantly by butyrate and α-synuclein genotypes, which may also affect the GIT microbiome directly. CONCLUSION By demonstrating the transportation of α-synuclein through RBC-EVs to the GIT, and its potential association with gut-vascular barrier markers and gut microbiome, this work highlights a potential mechanism by which RBC α-synuclein may impact PD initiation and/or progression. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Ying Yang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Nanhu Brain-computer Interface Institute, Hangzhou, Zhejiang, China
- Department of Pathology, Peking University Health Science Center, Beijing, China
| | - Tessandra Stewart
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Can Zhang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pan Wang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Lingang Laboratory, Shanghai, China
- National Human Brain Bank for Health and Disease, Zhejiang University, Hangzhou, China
| | - Zhi Xu
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinghua Jin
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yang Huang
- Department of Pathology, Peking University Health Science Center, Beijing, China
| | - Zongran Liu
- Department of Pathology, Peking University Health Science Center, Beijing, China
| | - Guoyu Lan
- Department of Pathology, Peking University Health Science Center, Beijing, China
| | - Xingguang Liang
- Central Laboratory, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lifu Sheng
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Min Shi
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Zhijian Cai
- School of Basic Medicine, Zhejiang University, Hangzhou, China
| | - Jing Zhang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Nanhu Brain-computer Interface Institute, Hangzhou, Zhejiang, China
- Lingang Laboratory, Shanghai, China
- National Human Brain Bank for Health and Disease, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Seo MH, Kwon D, Kim SH, Yeo S. Association between Decreased SGK1 and Increased Intestinal α-Synuclein in an MPTP Mouse Model of Parkinson's Disease. Int J Mol Sci 2023; 24:16408. [PMID: 38003598 PMCID: PMC10671719 DOI: 10.3390/ijms242216408] [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: 09/27/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Parkinson's disease (PD) is a globally common progressive neurodegenerative disease resulting from the loss of dopaminergic neurons in the brain. Increased α-synuclein (α-syn) is associated with the degeneration of dopaminergic neurons and non-motor symptoms like gastrointestinal disorders. In this study, we investigated the association between serum/glucocorticoid-related kinase 1 (SGK1) and α-syn in the colon of a PD mouse model. SGK1 and α-syn expression patterns were opposite in the surrounding colon tissue, with decreased SGK1 expression and increased α-syn expression in the PD group. Immunofluorescence analyses revealed the colocation of SGK1 and α-syn; the PD group demonstrated weaker SGK1 expression and stronger α-syn expression than the control group. Immunoblotting analysis showed that Na+/K+ pump ATPase α1 expression levels were significantly increased in the PD group. In SW480 cells with SGK1 knockdown using SGK1 siRNA, decreasing SGK1 levels corresponded with significant increases in the expression levels of α-syn and ATPase α1. These results suggest that SGK1 significantly regulates Na+/K+ pump ATPase, influencing the relationship between electrolyte balance and fecal formation in the PD mouse model. Gastrointestinal disorders are some of the major prodromal symptoms of PD. Therefore, modulating SGK1 expression could be an important strategy for controlling PD.
Collapse
Affiliation(s)
- Min Hyung Seo
- Department of Meridian and Acupoint, College of Korean Medicine, Sang Ji University, Wonju 26339, Republic of Korea; (M.H.S.); (D.K.)
| | - Dasom Kwon
- Department of Meridian and Acupoint, College of Korean Medicine, Sang Ji University, Wonju 26339, Republic of Korea; (M.H.S.); (D.K.)
| | - Soo-Hwan Kim
- Division of Biological Science and Technology, Yonsei University, 1 Yonseidae-gil, Wonju 26493, Republic of Korea
| | - Sujung Yeo
- Research Institute of Korean Medicine, Sangji University, Wonju 26339, Republic of Korea
| |
Collapse
|
11
|
Cabezudo D, Tsafaras G, Van Acker E, Van den Haute C, Baekelandt V. Mutant LRRK2 exacerbates immune response and neurodegeneration in a chronic model of experimental colitis. Acta Neuropathol 2023; 146:245-261. [PMID: 37289222 PMCID: PMC10328902 DOI: 10.1007/s00401-023-02595-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/12/2023] [Accepted: 05/25/2023] [Indexed: 06/09/2023]
Abstract
The link between the gut and the brain in Parkinson's disease (PD) pathogenesis is currently a subject of intense research. Indeed, gastrointestinal dysfunction is known as an early symptom in PD and inflammatory bowel disease (IBD) has recently been recognised as a risk factor for PD. The leucine-rich repeat kinase 2 (LRRK2) is a PD- and IBD-related protein with highest expression in immune cells. In this study, we provide evidence for a central role of LRRK2 in gut inflammation and PD. The presence of the gain-of-function G2019S mutation significantly increases the disease phenotype and inflammatory response in a mouse model of experimental colitis based on chronic dextran sulphate sodium (DSS) administration. Bone marrow transplantation of wild-type cells into G2019S knock-in mice fully rescued this exacerbated response, proving the key role of mutant LRRK2 in immune cells in this experimental colitis model. Furthermore, partial pharmacological inhibition of LRRK2 kinase activity also reduced the colitis phenotype and inflammation. Moreover, chronic experimental colitis also induced neuroinflammation and infiltration of peripheral immune cells into the brain of G2019S knock-in mice. Finally, combination of experimental colitis with overexpression of α-synuclein in the substantia nigra aggravated motor deficits and dopaminergic neurodegeneration in G2019S knock-in mice. Taken together, our results link LRRK2 with the immune response in colitis and provide evidence that gut inflammation can impact brain homeostasis and contribute to neurodegeneration in PD.
Collapse
Affiliation(s)
- Diego Cabezudo
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49, box 1023, 3000, Leuven, Belgium
| | - George Tsafaras
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49, box 1023, 3000, Leuven, Belgium
| | - Eva Van Acker
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49, box 1023, 3000, Leuven, Belgium
| | - Chris Van den Haute
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49, box 1023, 3000, Leuven, Belgium
- Leuven Viral Vector Core, Herestraat 49, box 1023, 3000, Leuven, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Herestraat 49, box 1023, 3000, Leuven, Belgium.
| |
Collapse
|
12
|
Seo MH, Yeo S. The Effects of Serping1 siRNA in α-Synuclein Regulation in MPTP-Induced Parkinson's Disease. Biomedicines 2023; 11:1952. [PMID: 37509591 PMCID: PMC10377285 DOI: 10.3390/biomedicines11071952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Our understanding of the gastrointestinal system in the pathophysiology of Parkinson's disease (PD) has grown considerably over the last two decades. Patients with PD experience notable gastrointestinal symptoms, including constipation. In this study, the effects of knocked-down serping1, associated with the contraction and relaxation of smooth muscle and inflammation responses, by applying the serping1 siRNA were investigated in 1-methyl 4-phenyl 1,2,3,6-tetrahydropyridine-induced PD mice in an α-syn change aspect. In the result, serping1 expression was knocked down by the treatment of serping1 siRNA, and decreased serping1 induced the decrease α-syn in the colon. Furthermore, the changes in α-syn aggregation were also examined in the brain, and alleviated α-syn aggregation was also observed in an serping1 siRNA treatment group. The results indicated that serping1 siRNA could ease synucleinopathy related to the gastrointestinal system in PD. This study also raises the possibility that serping1 siRNA could alleviate α-syn aggregation in striatum and substantia nigra regions of the brain.
Collapse
Affiliation(s)
- Min Hyung Seo
- Department of Meridian and Acupoint, College of Korean Medicine, Sang Ji University, Wonju 26339, Republic of Korea
| | - Sujung Yeo
- Research Institute of Korean Medicine, Sang Ji University, #83 Sangjidae-Gil, Wonju 26339, Republic of Korea
| |
Collapse
|
13
|
Abstract
Several advances in fluid and tissue-based biomarkers for use in Parkinson's disease (PD) and other synucleinopathies have been made in the last several years. While work continues on species of alpha-synuclein (aSyn) and other proteins which can be measured from spinal fluid and plasma samples, immunohistochemistry and immunofluorescence from peripheral tissue biopsies and alpha-synuclein seeding amplification assays (aSyn-SAA: including real-time quaking induced conversion (RT-QuIC) and protein misfolding cyclic amplification (PMCA)) now offer a crucial advancement in their ability to identify aSyn species in PD patients in a categorical fashion (i.e., of aSyn + vs aSyn -); to augment clinical diagnosis however, aSyn-specific assays that have quantitative relevance to pathological burden remain an unmet need. Alzheimer's disease (AD) co-pathology is commonly found postmortem in PD, especially in those who develop dementia, and dementia with Lewy bodies (DLB). Biofluid biomarkers for tau and amyloid beta species can detect AD co-pathology in PD and DLB, which does have relevance for prognosis, but further work is needed to understand the interplay of aSyn tau, amyloid beta, and other pathological changes to generate comprehensive biomarker profiles for patients in a manner translatable to clinical trial design and individualized therapies.
Collapse
Affiliation(s)
- David G Coughlin
- Department of Neurosciences, University of California San Diego, 9444 Medical Center Drive, ECOB 03-021, MCC 0886, La Jolla, CA, 92037, USA.
| | - David J Irwin
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
14
|
Kang SY, Yun JY, Kang YK, Moon BS, Yoon HJ, Yoo MY, Kim BS. Salivary Gland Uptake on 18F-FP-CIT PET as a New Biomarker in Patients With Parkinsonism. Korean J Radiol 2023; 24:690-697. [PMID: 37404111 DOI: 10.3348/kjr.2023.0066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/31/2023] [Accepted: 04/27/2023] [Indexed: 07/06/2023] Open
Abstract
OBJECTIVE 18F-FP-CIT positron emission tomography (PET) is known for its high sensitivity and specificity for evaluating striatal dopamine transporter (DAT) binding. Recently, for the early diagnose of Parkinson's disease, many researchers focused on the diagnosis of synucleinopathy in organs involved in non-motor symptoms of Parkinson's disease. We investigated the feasibility of salivary gland uptake on 18F-FP-CIT PET as a new biomarker in patients with parkinsonism. MATERIALS AND METHODS A total of 219 participants with confirmed or presumed parkinsonism, including 54 clinically diagnosed idiopathic Parkinson's disease (IPD), 59 suspected and yet undiagnosed, and 106 with secondary parkinsonism, were enrolled. The standardized uptake value ratio (SUVR) of the salivary glands was measured on both early and delayed 18F-FP-CIT PET scans using the cerebellum as the reference region. Additionally, the delayed-to-early ratio (DE_ratio) of salivary gland was obtained. The results were compared between patients with different PET patterns. RESULTS The SUVR in early 18F-FP-CIT PET scan was significantly higher in patients with IPD pattern compared that in the non-dopaminergic degradation group (0.5 ± 0.19 vs. 0.6 ± 0.21, P < 0.001). Compared with the non-dopaminergic degradation group, the DE_ratio was significantly lower in patients with IPD (5.05 ± 1.7 vs. 4.0 ± 1.31, P < 0.001) or atypical parkinsonism patterns (5.05 ± 1.7 vs. 3.76 ± 0.96, P < 0.05). The DE_ratio was moderately and positively correlated with striatal DAT availability in both the whole striatum (r = 0.37, P < 0.001) and posterior putamen (r = 0.36, P < 0.001). CONCLUSION Parkinsonism patients with an IPD pattern exhibited a significant increase in uptake on early 18F-FP-CIT PET and a decrease in the DE_ratio in the salivary gland. Our findings suggest that salivary gland uptake of dual-phase 18F-FP-CIT PET can provide diagnostic information on DAT availability in patients with Parkinson's disease.
Collapse
Affiliation(s)
- Seo Young Kang
- Department of Nuclear Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Ji Young Yun
- Department of Neurology, Ewha Womans University College of Medicine, Seoul, Korea
| | - Yeon-Koo Kang
- Department of Nuclear Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Byung Seok Moon
- Department of Nuclear Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Hai-Jeon Yoon
- Department of Nuclear Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Min Young Yoo
- Department of Nuclear Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Bom Sahn Kim
- Department of Nuclear Medicine, Ewha Womans University College of Medicine, Seoul, Korea.
| |
Collapse
|
15
|
Simons E, Fleming SM. Role of rodent models in advancing precision medicine for Parkinson's disease. HANDBOOK OF CLINICAL NEUROLOGY 2023; 193:3-16. [PMID: 36803818 DOI: 10.1016/b978-0-323-85555-6.00002-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
With a current lack of disease-modifying treatments, an initiative toward implementing a precision medicine approach for treating Parkinson's disease (PD) has emerged. However, challenges remain in how to define and apply precision medicine in PD. To accomplish the goal of optimally targeted and timed treatment for each patient, preclinical research in a diverse population of rodent models will continue to be an essential part of the translational path to identify novel biomarkers for patient diagnosis and subgrouping, understand PD disease mechanisms, identify new therapeutic targets, and screen therapeutics prior to clinical testing. This review highlights the most common rodent models of PD and discusses how these models can contribute to defining and implementing precision medicine for the treatment of PD.
Collapse
Affiliation(s)
- Emily Simons
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| | - Sheila M Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States.
| |
Collapse
|
16
|
Videlock EJ, Hatami A, Zhu C, Kawaguchi R, Chen H, Khan T, Yehya AHS, Stiles L, Joshi S, Hoffman JM, Law KM, Rankin CR, Chang L, Maidment NT, John V, Geschwind DH, Pothoulakis C. Distinct Patterns of Gene Expression Changes in the Colon and Striatum of Young Mice Overexpressing Alpha-Synuclein Support Parkinson's Disease as a Multi-System Process. JOURNAL OF PARKINSON'S DISEASE 2023; 13:1127-1147. [PMID: 37638450 PMCID: PMC10657720 DOI: 10.3233/jpd-223568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/04/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Evidence supports a role for the gut-brain axis in Parkinson's disease (PD). Mice overexpressing human wild type α- synuclein (Thy1-haSyn) exhibit slow colonic transit prior to motor deficits, mirroring prodromal constipation in PD. Identifying molecular changes in the gut could provide both biomarkers for early diagnosis and gut-targeted therapies to prevent progression. OBJECTIVE To identify early molecular changes in the gut-brain axis in Thy1-haSyn mice through gene expression profiling. METHODS Gene expression profiling was performed on gut (colon) and brain (striatal) tissue from Thy1-haSyn and wild-type (WT) mice aged 1 and 3 months using 3' RNA sequencing. Analysis included differential expression, gene set enrichment and weighted gene co-expression network analysis (WGCNA). RESULTS At one month, differential expression (Thy1-haSyn vs. WT) of mitochondrial genes and pathways related to PD was discordant between gut and brain, with negative enrichment in brain (enriched in WT) but positive enrichment in gut. Linear regression of WGCNA modules showed partial independence of gut and brain gene expression changes. Thy1-haSyn-associated WGCNA modules in the gut were enriched for PD risk genes and PD-relevant pathways including inflammation, autophagy, and oxidative stress. Changes in gene expression were modest at 3 months. CONCLUSIONS Overexpression of haSyn acutely disrupts gene expression in the colon. While changes in colon gene expression are highly related to known PD-relevant mechanisms, they are distinct from brain changes, and in some cases, opposite in direction. These findings are in line with the emerging view of PD as a multi-system disease.
Collapse
Affiliation(s)
- Elizabeth J. Videlock
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Asa Hatami
- The Drug Discovery Lab, Mary S. Easton Center for Alzheimer’s Disease Research, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Chunni Zhu
- The Drug Discovery Lab, Mary S. Easton Center for Alzheimer’s Disease Research, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Riki Kawaguchi
- The Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
| | - Han Chen
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Tasnin Khan
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Ashwaq Hamid Salem Yehya
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Linsey Stiles
- Department of Medicine, Division of Endocrinology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Swapna Joshi
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jill M. Hoffman
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Ka Man Law
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Carl Robert Rankin
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Lin Chang
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Nigel T. Maidment
- Hatos Center for Neuropharmacology, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Varghese John
- The Drug Discovery Lab, Mary S. Easton Center for Alzheimer’s Disease Research, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Daniel H. Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Center for Autism Research and Treatment, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, CA, USA
- Institute for Precision Health, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Charalabos Pothoulakis
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
17
|
Coughlin DG, Hiniker A, Peterson C, Kim Y, Arezoumandan S, Giannini L, Pizzo D, Weintraub D, Siderowf A, Litvan I, Rissman RA, Galasko D, Hansen L, Trojanowski JQ, Lee E, Grossman M, Irwin D. Digital Histological Study of Neocortical Grey and White Matter Tau Burden Across Tauopathies. J Neuropathol Exp Neurol 2022; 81:953-964. [PMID: 36269086 PMCID: PMC9677241 DOI: 10.1093/jnen/nlac094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
3R/4R-tau species are found in Alzheimer disease (AD) and ∼50% of Lewy body dementias at autopsy (LBD+tau); 4R-tau accumulations are found in progressive supranuclear palsy (PSP) and corticobasal degeneration (CBD). Digital image analysis techniques can elucidate patterns of tau pathology more precisely than traditional methods but repeatability across centers is unclear. We calculated regional percentage areas occupied by tau pathological inclusions from the middle frontal cortex (MFC), superior temporal cortex (STC), and angular gyrus (ANG) from cases from the University of Pennsylvania and the University of California San Diego with AD, LBD+tau, PSP, or CBD (n = 150) using QuPath. In both cohorts, AD and LBD+tau had the highest grey and white matter tau burden in the STC (p ≤ 0.04). White matter tau burden was relatively higher in 4R-tauopathies than 3R/4R-tauopathies (p < 0.003). Grey and white matter tau were correlated in all diseases (R2=0.43-0.79, p < 0.04) with the greatest increase of white matter per unit grey matter tau observed in PSP (p < 0.02 both cohorts). Grey matter tau negatively correlated with MMSE in AD and LBD+tau (r = -4.4 to -5.4, p ≤ 0.02). These data demonstrate the feasibility of cross-institutional digital histology studies that generate finely grained measurements of pathology which can be used to support biomarker development and models of disease progression.
Collapse
Affiliation(s)
- David G Coughlin
- From the Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Annie Hiniker
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Claire Peterson
- Digital Neuropathology Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yongya Kim
- From the Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Sanaz Arezoumandan
- Digital Neuropathology Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lucia Giannini
- Digital Neuropathology Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neurology, Erasmus University Medical Center, Alzheimer Center, Rotterdam, The Netherlands
| | - Donald Pizzo
- Center for Advanced Laboratory Medicine, University of California San Diego, La Jolla, California, USA
| | - Daniel Weintraub
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrew Siderowf
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Irene Litvan
- From the Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Robert A Rissman
- From the Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Douglas Galasko
- From the Department of Neurosciences, University of California San Diego, La Jolla, California, USA
| | - Lawrence Hansen
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edward Lee
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Murray Grossman
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - David Irwin
- Digital Neuropathology Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
Wais T, Hasan M, Rai V, Agrawal DK. Gut-brain communication in COVID-19: molecular mechanisms, mediators, biomarkers, and therapeutics. Expert Rev Clin Immunol 2022; 18:947-960. [PMID: 35868344 PMCID: PMC9388545 DOI: 10.1080/1744666x.2022.2105697] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 07/21/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Infection with COVID-19 results in acute respiratory symptoms followed by long COVID multi-organ effects presenting with neurological, cardiovascular, musculoskeletal, and gastrointestinal (GI) manifestations. Temporal relationship between gastrointestinal and neurological symptoms is unclear but warranted for exploring better clinical care for COVID-19 patients. AREAS COVERED We critically reviewed the temporal relationship between gut-brain axis after SARS-CoV-2 infection and the molecular mechanisms involved in neuroinvasion following GI infection. Mediators are identified that could serve as biomarkers and therapeutic targets in SARS-CoV-2. We discussed the potential therapeutic approaches to mitigate the effects of GI infection with SARS-CoV-2. EXPERT OPINION Altered gut microbiota cause increased expression of various mediators, including zonulin causing disruption of tight junction. This stimulates enteric nervous system and signals to CNS precipitating neurological sequalae. Published reports suggest potential role of cytokines, immune cells, B(0)AT1 (SLC6A19), ACE2, TMRSS2, TMPRSS4, IFN-γ, IL-17A, zonulin, and altered gut microbiome in gut-brain axis and associated neurological sequalae. Targeting these mediators and gut microbiome to improve immunity will be of therapeutic significance. In-depth research and well-designed large-scale population-based clinical trials with multidisciplinary and collaborative approaches are warranted. Investigating the temporal relationship between organs involved in long-term sequalae is critical due to evolving variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Tameena Wais
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences. Pomona, CA 91766
| | - Mehde Hasan
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences. Pomona, CA 91766
| | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences. Pomona, CA 91766
| | - Devendra K. Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences. Pomona, CA 91766
| |
Collapse
|
19
|
Chen AD, Cao JX, Chen HC, Du HL, Xi XX, Sun J, Yin J, Jing YH, Gao LP. Rotenone aggravates PD-like pathology in A53T mutant human α-synuclein transgenic mice in an age-dependent manner. Front Aging Neurosci 2022; 14:842380. [PMID: 36004003 PMCID: PMC9393581 DOI: 10.3389/fnagi.2022.842380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/12/2022] [Indexed: 11/26/2022] Open
Abstract
Multiple factors such as genes, environment, and age are involved in developing Parkinson’s disease (PD) pathology. However, how various factors interact to cause PD remains unclear. Here, 3-month and 9-month-old hα-syn+/− mice were treated with low-dose rotenone for 2 months to explore the mechanisms that underline the environment–gene–age interaction in the occurrence of PD. We have examined the behavior of mice and the PD-like pathologies of the brain and gut. The present results showed that impairments of the motor function and olfactory function were more serious in old hα-syn+/– mice with rotenone than that in young mice. The dopaminergic neuron loss in the SNc is more in old hα-syn+/– mice with rotenone than in young mice. Expression of hα-syn+/– is increased in the SNc of hα-syn+/– mice following rotenone treatment for 2 months. Furthermore, the number of activated microglia cells increased in SNc and accompanied the high expression of inflammatory cytokines, namely, TNF-α and IL-18 in the midbrain of old hα-syn+/– mice treated with rotenone. Meanwhile, we found that after treatment with rotenone, hα-syn positive particles deposited in the intestinal wall, intestinal microflora, and T lymphocyte subtypes of Peyer’s patches changed, and intestinal mucosal permeability increased. Moreover, these phenomena were age-dependent. These findings suggested that rotenone aggravated the PD-like pathologies and affected the brain and gut of human α-syn+/– transgenic mice in an age-dependent manner.
Collapse
Affiliation(s)
- An-Di Chen
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jia-Xin Cao
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Hai-Chao Chen
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Hong-Li Du
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiao-Xia Xi
- Center of Experimental Animal, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jing Sun
- Center of Experimental Animal, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jie Yin
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yu-Hong Jing
- Institute of Anatomy and Histology & Embryology, Neuroscience, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- *Correspondence: Yu-Hong Jing,
| | - Li-Ping Gao
- Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Li-Ping Gao,
| |
Collapse
|
20
|
Shin C, Kim SI, Park SH, Shin JH, Lee CY, Yang HK, Lee HJ, Kong SH, Suh YS, Kim HJ, Jeon B. Sensitivity of Detecting Alpha-Synuclein Accumulation in the Gastrointestinal Tract and Tissue Volume Examined. J Mov Disord 2022; 15:264-268. [PMID: 35880377 PMCID: PMC9536904 DOI: 10.14802/jmd.22042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/31/2022] [Indexed: 11/25/2022] Open
Abstract
Objective This study aimed to evaluate whether a larger tissue volume increases the sensitivity of detecting alpha-synuclein (AS) pathology in the gastrointestinal (GI) tract. Methods Nine patients with Parkinson’s disease (PD) or idiopathic rapid eye movement sleep disorder (iRBD) who underwent GI operation and had full-depth intestinal blocks were included. All patients were selected from our previous study population. A total of 10 slides (5 serial sections from the proximal and distal blocks) per patient were analyzed. Results In previous studies, pathologic evaluation revealed phosphorylated AS (+) in 5/9 patients (55.6%) and in 1/5 controls (20.0%); in this extensive examination, this increased to 8/9 patients (88.9%) but remained the same in controls (20.0%). The severity and distribution of positive findings were similar between patients with iRBD and PD. Conclusion Examining a large tissue volume increased the sensitivity of detecting AS accumulation in the GI tract.
Collapse
Affiliation(s)
- Chaewon Shin
- Department of Neurology, Chungnam National University Sejong Hospital, Chungnam National University College of Medicine, Sejong, Korea
| | - Seong-Ik Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Hwan Shin
- Department of Neurology, MRC and Movement Disorder Center, Seoul National University Hospital, Parkinson Study Group, Seoul National University College of Medicine, Seoul, Korea
| | - Chan Young Lee
- Department of Neurology, Ehwa Womans University Mokdong Hospital, Seoul, Korea
| | - Han-Kwang Yang
- Department of Surgery, Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hyuk-Joon Lee
- Department of Surgery, Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Seong-Ho Kong
- Department of Surgery, Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Han-Joon Kim
- Department of Neurology, MRC and Movement Disorder Center, Seoul National University Hospital, Parkinson Study Group, Seoul National University College of Medicine, Seoul, Korea
| | - Beomseok Jeon
- Department of Neurology, MRC and Movement Disorder Center, Seoul National University Hospital, Parkinson Study Group, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Jotanovic J, Milin-Lazovic J, Alafuzoff I. Gastrointestinal Biopsy Obtained During Cancer Screening, a Biological Marker for α-Synucleinopathy? J Neuropathol Exp Neurol 2022; 81:356-362. [PMID: 35388426 PMCID: PMC9041339 DOI: 10.1093/jnen/nlac023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The hallmark alteration in α-synucleinopathies, α-synuclein, is observed not only in the brain but also in the peripheral tissues, particularly in the intestine. This suggests that endoscopic biopsies performed for colon cancer screening could facilitate the assessment of α-synuclein in the gastrointestinal (GI) tract. Using immunohistochemistry for α-synuclein, we assessed whether GI biopsies could be used to confirm an ongoing α-synucleinopathy. Seventy-four subjects with cerebral α-synucleinopathy in various Braak stages with concomitant GI biopsies were available for study. In 81% of the subjects, α-synuclein was seen in the mucosal/submucosal GI biopsies. Two subjects with severe cerebral α-synucleinopathy and a long delay between biopsy and death displayed no α-synuclein pathology in the gut, and 11 subjects with sparse cerebral α-synucleinopathy displayed GI α-synuclein up to 36 years prior to death. The finding that there was no GI α-synuclein in 19% of the subjects with cerebral α-synucleinopathy, and α-synuclein was observed in the gut of 11 subjects (15%) with sparse cerebral α-synucleinopathy even many years prior to death is unexpected and jeopardizes the use of assessment of α-synuclein in the peripheral tissue for confirmation of an ongoing cerebral α-synucleinopathy.
Collapse
Affiliation(s)
- Jelena Jotanovic
- Department of Pathology, Uppsala University Hospital, Uppsala, Sweden
| | - Jelena Milin-Lazovic
- Institute for Medical Statistics and Informatics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Irina Alafuzoff
- Department of Pathology, Uppsala University Hospital, Uppsala, Sweden.,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
22
|
Chalazonitis A, Rao M, Sulzer D. Similarities and differences between nigral and enteric dopaminergic neurons unravel distinctive involvement in Parkinson's disease. NPJ Parkinsons Dis 2022; 8:50. [PMID: 35459867 PMCID: PMC9033791 DOI: 10.1038/s41531-022-00308-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 03/14/2022] [Indexed: 11/09/2022] Open
Abstract
In addition to the well-known degeneration of midbrain dopaminergic neurons, enteric neurons can also be affected in neurodegenerative disorders such as Parkinson's disease (PD). Dopaminergic neurons have recently been identified in the enteric nervous system (ENS). While ENS dopaminergic neurons have been shown to degenerate in genetic mouse models of PD, analyses of their survival in enteric biopsies of PD patients have provided inconsistent results to date. In this context, this review seeks to highlight the distinctive and shared factors and properties that control the evolution of these two sets of dopaminergic neurons from neuronal precursors to aging neurons. Although their cellular sources and developmental times of origin differ, midbrain and ENS dopaminergic neurons express many transcription factors in common and their respective environments express similar neurotrophic molecules. For example, Foxa2 and Sox6 are expressed by both populations to promote the specification, differentiation, and long-term maintenance of the dopaminergic phenotype. Both populations exhibit sustained patterns of excitability that drive intrinsic vulnerability over time. In disorders such as PD, colon biopsies have revealed aggregation of alpha-synuclein in the submucosal plexus where dopaminergic neurons reside and lack blood barrier protection. Thus, these enteric neurons may be more susceptible to neurotoxic insults and aggregation of α-synuclein that spreads from gut to midbrain. Under sustained stress, inefficient autophagy leads to neurodegeneration, GI motility dysfunction, and PD symptoms. Recent findings suggest that novel neurotrophic factors such as CDNF have the potential to be used as neuroprotective agents to prevent and treat ENS symptoms of PD.
Collapse
Affiliation(s)
- Alcmène Chalazonitis
- Department of Pathology & Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA.
| | - Meenakshi Rao
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - David Sulzer
- Departments of Psychiatry, Neurology, and Pharmacology, Division of Molecular Therapeutics, New York State Psychiatry Institute, Columbia University, New York, NY, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| |
Collapse
|
23
|
Neuropathological substrates of cognition in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2022; 269:177-193. [PMID: 35248194 DOI: 10.1016/bs.pbr.2022.01.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Autopsy validation is still required for a definitive diagnosis of Parkinson's disease (Postuma et al., 2015), where the presence of Lewy bodies and Lewy neurites, composed primarily of alpha-synuclein, are observed in stereotyped patterns throughout regions of the brainstem, limbic, and neocortical regions of the brain (Braak et al., 2003). In spite of these relatively reliable observed patterns of alpha-synuclein pathology, there is a large degree of heterogeneity in the timing and features of neuropsychiatric and cognitive dysfunction in Parkinson's disease (Fereshtehnejad et al., 2015; Selikhova et al., 2009; Williams-Gray et al., 2013). Detailed studies of their neuropathological substrates of cognitive dysfunction and their associations with a variety of in vivo biomarkers have begun to disentangle this complex relationship, but ongoing multicentered, longitudinal studies of well-characterized and autopsy validated cases are still required.
Collapse
|
24
|
Scott GD, Arnold MR, Beach TG, Gibbons CH, Kanthasamy AG, Lebovitz RM, Lemstra AW, Shaw LM, Teunissen CE, Zetterberg H, Taylor AS, Graham TC, Boeve BF, Gomperts SN, Graff-Radford NR, Moussa C, Poston KL, Rosenthal LS, Sabbagh MN, Walsh RR, Weber MT, Armstrong MJ, Bang JA, Bozoki AC, Domoto-Reilly K, Duda JE, Fleisher JE, Galasko DR, Galvin JE, Goldman JG, Holden SK, Honig LS, Huddleston DE, Leverenz JB, Litvan I, Manning CA, Marder KS, Pantelyat AY, Pelak VS, Scharre DW, Sha SJ, Shill HA, Mari Z, Quinn JF, Irwin DJ. Fluid and Tissue Biomarkers of Lewy Body Dementia: Report of an LBDA Symposium. Front Neurol 2022; 12:805135. [PMID: 35173668 PMCID: PMC8841880 DOI: 10.3389/fneur.2021.805135] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
The Lewy Body Dementia Association (LBDA) held a virtual event, the LBDA Biofluid/Tissue Biomarker Symposium, on January 25, 2021, to present advances in biomarkers for Lewy body dementia (LBD), which includes dementia with Lewy bodies (DLBs) and Parkinson's disease dementia (PDD). The meeting featured eight internationally known scientists from Europe and the United States and attracted over 200 scientists and physicians from academic centers, the National Institutes of Health, and the pharmaceutical industry. Methods for confirming and quantifying the presence of Lewy body and Alzheimer's pathology and novel biomarkers were discussed.
Collapse
Affiliation(s)
- Gregory D. Scott
- Department of Pathology, Oregon Health and Science University, Portland, OR, United States
- Department of Pathology and Laboratory Services, VA Portland Medical Center, Portland, OR, United States
| | - Moriah R. Arnold
- Graduate Program in Biomedical Sciences, School of Medicine M.D./Ph.D. Program, Oregon Health and Science University, Portland, OR, United States
| | - Thomas G. Beach
- Civin Laboratory for Neuropathology and Brain and Body Donation Program, Banner Sun Health Research Institute, Sun City, AZ, United States
| | - Christopher H. Gibbons
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Anumantha G. Kanthasamy
- Department of Physiology and Pharmacology, Center for Brain Sciences and Neurodegenerative Diseases, University of Georgia, Athens, GA, United States
| | | | - Afina W. Lemstra
- Department of Neurology, Amsterdam University Medical Center (UMC), Alzheimer Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leslie M. Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Charlotte E. Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, University College London (UCL) Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at University College London, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | | | - Todd C. Graham
- Lewy Body Dementia Association, Lilburn, GA, United States
| | - Bradley F. Boeve
- Department of Neurology and Center for Sleep Medicine, Mayo Clinic, Rochester, MN, United States
| | - Stephen N. Gomperts
- Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | | | - Charbel Moussa
- Department of Neurology, Georgetown University Medical Center, Washington DC, CA, United States
| | - Kathleen L. Poston
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Liana S. Rosenthal
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Marwan N. Sabbagh
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Ryan R. Walsh
- Barrow Neurological Institute and Muhammed Ali Parkinson Center, Phoenix, AZ, United States
| | - Miriam T. Weber
- Department of Neurology, University of Rochester, Rochester, NY, United States
| | - Melissa J. Armstrong
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Jee A. Bang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Andrea C. Bozoki
- Department of Neurology, University of North Carolina, Chapel Hill, NC, United States
| | | | - John E. Duda
- Parkinson's Disease Research, Education and Clinical Center, Michael J. Crescenz VA Medical Center, Philadelphia, PA, United States
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jori E. Fleisher
- Department of Neurological Sciences, Rush Medical College, Chicago, IL, United States
| | - Douglas R. Galasko
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - James E. Galvin
- Department of Neurology, Comprehensive Center for Brain Health, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Jennifer G. Goldman
- Shirley Ryan Abilitylab and Department of Physical Medicine and Rehabilitation and Neurology, Parkinson's Disease and Movement Disorders, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Samantha K. Holden
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Lawrence S. Honig
- Columbia University Irving Medical Center, New York, NY, United States
| | - Daniel E. Huddleston
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| | - James B. Leverenz
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, OH, United States
| | - Irene Litvan
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Carol A. Manning
- Department of Neurology, University of Virginia, Charlottesville, VA, United States
| | - Karen S. Marder
- Columbia University Irving Medical Center, New York, NY, United States
| | - Alexander Y. Pantelyat
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Victoria S. Pelak
- Departments of Neurology and Ophthalmology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Douglas W. Scharre
- Department of Neurology, Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Sharon J. Sha
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, United States
| | - Holly A. Shill
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Zoltan Mari
- Lou Ruvo Center for Brain Health, Cleveland Clinic Lerner College of Medicine, Las Vegas, NV, United States
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
- Department of Neurology, VA Portland Medical Center, Portland, OR, United States
| | - David J. Irwin
- Department of Neurology, University of Pennsylvania Health System, Philadelphia, PA, United States
- Digital Neuropathology Laboratory, Philadelphia, PA, United States
- Lewy Body Disease Research Center of Excellence, Philadelphia, PA, United States
- Frontotemporal Degeneration Center, Philadelphia, PA, United States
| |
Collapse
|
25
|
Kluge A, Bunk J, Schaeffer E, Drobny A, Xiang W, Knacke H, Bub S, Lückstädt W, Arnold P, Lucius R, Berg D, Zunke F. OUP accepted manuscript. Brain 2022; 145:3058-3071. [PMID: 35722765 DOI: 10.1093/brain/awac115] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/12/2022] [Accepted: 03/13/2022] [Indexed: 11/13/2022] Open
Abstract
To date, no reliable clinically applicable biomarker has been established for Parkinson's disease. Our results indicate that a long anticipated blood test for Parkinson's disease may be realized. Following the isolation of neuron-derived extracellular vesicles of Parkinson's disease patients and non-Parkinson's disease individuals, immunoblot analyses were performed to detect extracellular vesicle-derived α-synuclein. Pathological α-synuclein forms derived from neuronal extracellular vesicles could be detected under native conditions and were significantly increased in all individuals with Parkinson's disease and clearly distinguished disease from the non-disease state. By performing an α-synuclein seeding assay these soluble conformers could be amplified and seeding of pathological protein folding was demonstrated. Amplified α-synuclein conformers exhibited β-sheet-rich structures and a fibrillary appearance. Our study demonstrates that the detection of pathological α-synuclein conformers from neuron-derived extracellular vesicles from blood plasma samples has the potential to evolve into a blood-biomarker of Parkinson's disease that is still lacking so far. Moreover, the distribution of seeding-competent α-synuclein within blood exosomes sheds a new light of pathological disease mechanisms in neurodegenerative disorders.
Collapse
Affiliation(s)
- Annika Kluge
- Department of Neurology, University Hospital Kiel, 24105 Kiel, Germany
| | - Josina Bunk
- Institute of Biochemistry, Christian-Albrecht-University Kiel, 24118 Kiel, Germany
| | - Eva Schaeffer
- Department of Neurology, University Hospital Kiel, 24105 Kiel, Germany
| | - Alice Drobny
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Henrike Knacke
- Department of Neurology, University Hospital Kiel, 24105 Kiel, Germany
| | - Simon Bub
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Wiebke Lückstädt
- Institute of Anatomy, Christian-Albrecht-University Kiel, 24118 Kiel, Germany
| | - Philipp Arnold
- Institute of Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Ralph Lucius
- Institute of Anatomy, Christian-Albrecht-University Kiel, 24118 Kiel, Germany
| | - Daniela Berg
- Department of Neurology, University Hospital Kiel, 24105 Kiel, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
26
|
Bindas AJ, Kulkarni S, Koppes RA, Koppes AN. Parkinson's disease and the gut: Models of an emerging relationship. Acta Biomater 2021; 132:325-344. [PMID: 33857691 DOI: 10.1016/j.actbio.2021.03.071] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/17/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized by a progressive loss of fine motor function that impacts 1-2 out of 1,000 people. PD occurs predominately late in life and lacks a definitive biomarker for early detection. Recent cross-disciplinary progress has implicated the gut as a potential origin of PD pathogenesis. The gut-origin hypothesis has motivated research on gut PD pathology and transmission to the brain, especially during the prodromal stage (10-20 years before motor symptom onset). Early findings have revealed several possible triggers for Lewy pathology - the pathological hallmark of PD - in the gut, suggesting that microbiome and epithelial interactions may play a greater than appreciated role. But the mechanisms driving Lewy pathology and gut-brain transmission in PD remain unknown. Development of artificial α-Synuclein aggregates (α-Syn preformed fibrils) and animal disease models have recapitulated features of PD progression, enabling for the first time, controlled investigation of the gut-origin hypothesis. However, the role of specific cells in PD transmission, such as neurons, remains limited and requires in vitro models for controlled evaluation and perturbation. Human cell populations, three-dimensional organoids, and microfluidics as discovery platforms inch us closer to improving existing treatment for patients by providing platforms for discovery and screening. This review includes a discussion of PD pathology, conventional treatments, in vivo and in vitro models, and future directions. STATEMENT OF SIGNIFICANCE: Parkinson's Disease remains a common neurodegenerative disease with palliative versus causal treatments. Recently, the gut-origin hypothesis, where Parkinson's disease is thought to originate and spread from the gut to the brain, has gained traction as a field of investigation. However, despite the wealth of studies and innovative approaches to accelerate the field, there remains a need for in vitro tools to enable fundamental biological understanding of disease progression, and compound screening and efficacy. In this review, we present a historical perspective of Parkinson's Disease pathogenesis, detection, and conventional therapy, animal and human models investigating the gut-origin hypothesis, in vitro models to enable controlled discovery, and future outlooks for this blossoming field.
Collapse
Affiliation(s)
- Adam J Bindas
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115, USA.
| | - Subhash Kulkarni
- Division of Gastroenterology and Hepatology, Johns Hopkins University, 720 Rutland Avenue., Baltimore, MD 21205, USA.
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115, USA.
| | - Abigail N Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115, USA; Department of Biology, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Beach TG, Adler CH, Sue LI, Shill HA, Driver-Dunckley E, Mehta SH, Intorcia AJ, Glass MJ, Walker JE, Arce R, Nelson CM, Serrano GE. Vagus Nerve and Stomach Synucleinopathy in Parkinson's Disease, Incidental Lewy Body Disease, and Normal Elderly Subjects: Evidence Against the "Body-First" Hypothesis. JOURNAL OF PARKINSONS DISEASE 2021; 11:1833-1843. [PMID: 34151862 PMCID: PMC10082635 DOI: 10.3233/jpd-212733] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Braak and others have proposed that Lewy-type α-synucleinopathy in Parkinson's disease (PD) may arise from an exogenous pathogen that passes across the gastric mucosa and then is retrogradely transported up the vagus nerve to the medulla. OBJECTIVE We tested this hypothesis by immunohistochemically staining, with a method specific for p-serine 129 α-synuclein (pSyn), stomach and vagus nerve tissue from an autopsy series of 111 normal elderly subjects, 33 with incidental Lewy body disease (ILBD) and 53 with PD. METHODS Vagus nerve samples were taken adjacent to the carotid artery in the neck. Stomach samples were taken from the gastric body, midway along the greater curvature. Formalin-fixed paraffin-embedded sections were immunohistochemically stained for pSyn, shown to be highly specific and sensitive for α-synuclein pathology. RESULTS Median disease duration for the PD group was 13 years. In the vagus nerve none of the 111 normal subjects had pSyn in the vagus, while 12/26 ILBD (46%) and 32/36 PD (89%) subjects were pSyn-positive. In the stomach none of the 102 normal subjects had pSyn while 5/30 (17%) ILBD and 42/52 (81%) of PD subjects were pSyn-positive. CONCLUSION As there was no pSyn in the vagus nerve or stomach of subjects without brain pSyn, these results support initiation of pSyn in the brain. The presence of pSyn in the vagus nerve and stomach of a subset of ILBD cases indicates that synucleinopathy within the peripheral nervous system may occur, within a subset of individuals, at preclinical stages of Lewy body disease.
Collapse
Affiliation(s)
| | - Charles H Adler
- Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Lucia I Sue
- Banner Sun Health Research Institute, Sun City, AZ, USA
| | | | | | - Shyamal H Mehta
- Mayo Clinic College of Medicine, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | | | | | | | - Richard Arce
- Banner Sun Health Research Institute, Sun City, AZ, USA
| | | | | |
Collapse
|
28
|
Schmidt C, Planchette AL, Nguyen D, Giardina G, Neuenschwander Y, Franco MD, Mylonas A, Descloux AC, Pomarico E, Radenovic A, Extermann J. High resolution optical projection tomography platform for multispectral imaging of the mouse gut. BIOMEDICAL OPTICS EXPRESS 2021; 12:3619-3629. [PMID: 34221683 PMCID: PMC8221953 DOI: 10.1364/boe.423284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/06/2021] [Accepted: 05/18/2021] [Indexed: 06/13/2023]
Abstract
Optical projection tomography (OPT) is a powerful tool for three-dimensional imaging of mesoscopic biological samples with great use for biomedical phenotyping studies. We present a fluorescent OPT platform that enables direct visualization of biological specimens and processes at a centimeter scale with high spatial resolution, as well as fast data throughput and reconstruction. We demonstrate nearly isotropic sub-28 µm resolution over more than 60 mm3 after reconstruction of a single acquisition. Our setup is optimized for imaging the mouse gut at multiple wavelengths. Thanks to a new sample preparation protocol specifically developed for gut specimens, we can observe the spatial arrangement of the intestinal villi and the vasculature network of a 3-cm long healthy mouse gut. Besides the blood vessel network surrounding the gastrointestinal tract, we observe traces of vasculature at the villi ends close to the lumen. The combination of rapid acquisition and a large field of view with high spatial resolution in 3D mesoscopic imaging holds an invaluable potential for gastrointestinal pathology research.
Collapse
Affiliation(s)
- Cédric Schmidt
- HEPIA/HES-SO, University of Applied Sciences of Western Switzerland, Rue de la Prairie 4, 1202 Geneva, Switzerland
| | - Arielle L. Planchette
- Laboratoire de Biologie à l’Échelle Nanométrique, School of Engineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - David Nguyen
- Zlatic Lab, Neurobiology, MRC-Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Gabriel Giardina
- HEPIA/HES-SO, University of Applied Sciences of Western Switzerland, Rue de la Prairie 4, 1202 Geneva, Switzerland
| | - Yoan Neuenschwander
- HEPIA/HES-SO, University of Applied Sciences of Western Switzerland, Rue de la Prairie 4, 1202 Geneva, Switzerland
| | - Mathieu Di Franco
- HEPIA/HES-SO, University of Applied Sciences of Western Switzerland, Rue de la Prairie 4, 1202 Geneva, Switzerland
| | - Alessio Mylonas
- Laboratoire de Biologie à l’Échelle Nanométrique, School of Engineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Adrien C. Descloux
- Laboratoire de Biologie à l’Échelle Nanométrique, School of Engineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Enrico Pomarico
- HEPIA/HES-SO, University of Applied Sciences of Western Switzerland, Rue de la Prairie 4, 1202 Geneva, Switzerland
| | - Aleksandra Radenovic
- Laboratoire de Biologie à l’Échelle Nanométrique, School of Engineering, École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Jérôme Extermann
- HEPIA/HES-SO, University of Applied Sciences of Western Switzerland, Rue de la Prairie 4, 1202 Geneva, Switzerland
| |
Collapse
|
29
|
Beach T, Chahine LM, Adler CH, Mollenhauer B. Author Response: In Vivo Distribution of α-Synuclein in Multiple Tissues and Biofluids in Parkinson Disease. Neurology 2021; 96:965-967. [PMID: 34001544 DOI: 10.1212/wnl.0000000000011938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
30
|
Preclinical Detection of Alpha-Synuclein Seeding Activity in the Colon of a Transgenic Mouse Model of Synucleinopathy by RT-QuIC. Viruses 2021; 13:v13050759. [PMID: 33926043 PMCID: PMC8145297 DOI: 10.3390/v13050759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/15/2021] [Accepted: 04/21/2021] [Indexed: 12/29/2022] Open
Abstract
In synucleinopathies such as Parkinson’s disease (PD) and dementia with Lewy body (DLB), pathological alpha-synuclein (α-syn) aggregates are found in the gastrointestinal (GI) tract as well as in the brain. In this study, using real-time quaking-induced conversion (RT-QuIC), we investigated the presence of α-syn seeding activity in the brain and colon tissue of G2-3 transgenic mice expressing human A53T α-syn. Here we show that pathological α-syn aggregates with seeding activity were present in the colon of G2-3 mice as early as 3 months old, which is in the presymptomatic stage prior to the observation of any neurological abnormalities. In contrast, α-syn seeding activity was not detectable in 3 month-old mouse brains and only identified at 6 months of age in one of three mice. In the symptomatic stage of 12 months of age, RT-QuIC seeding activity was consistently detectable in both the brain and colon of G2-3 mice. Our results indicate that the RT-QuIC assay can presymptomatically detect pathological α-syn aggregates in the colon of G2-3 mice several months prior to their detection in brain tissue.
Collapse
|
31
|
Jung YJ, Tweedie D, Scerba MT, Kim DS, Palmas MF, Pisanu A, Carta AR, Greig NH. Repurposing Immunomodulatory Imide Drugs (IMiDs) in Neuropsychiatric and Neurodegenerative Disorders. Front Neurosci 2021; 15:656921. [PMID: 33854417 PMCID: PMC8039148 DOI: 10.3389/fnins.2021.656921] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation represents a common trait in the pathology and progression of the major psychiatric and neurodegenerative disorders. Neuropsychiatric disorders have emerged as a global crisis, affecting 1 in 4 people, while neurological disorders are the second leading cause of death in the elderly population worldwide (WHO, 2001; GBD 2016 Neurology Collaborators, 2019). However, there remains an immense deficit in availability of effective drug treatments for most neurological disorders. In fact, for disorders such as depression, placebos and behavioral therapies have equal effectiveness as antidepressants. For neurodegenerative diseases such as Parkinson's disease and Alzheimer's disease, drugs that can prevent, slow, or cure the disease have yet to be found. Several non-traditional avenues of drug target identification have emerged with ongoing neurological disease research to meet the need for novel and efficacious treatments. Of these novel avenues is that of neuroinflammation, which has been found to be involved in the progression and pathology of many of the leading neurological disorders. Neuroinflammation is characterized by glial inflammatory factors in certain stages of neurological disorders. Although the meta-analyses have provided evidence of genetic/proteomic upregulation of inflammatory factors in certain stages of neurological disorders. Although the mechanisms underpinning the connections between neuroinflammation and neurological disorders are unclear, and meta-analysis results have shown high sensitivity to factors such as disorder severity and sample type, there is significant evidence of neuroinflammation associations across neurological disorders. In this review, we summarize the role of neuroinflammation in psychiatric disorders such as major depressive disorder, generalized anxiety disorder, post-traumatic stress disorder, and bipolar disorder, as well as in neurodegenerative disorders, such as Parkinson's disease and Alzheimer's disease, and introduce current research on the potential of immunomodulatory imide drugs (IMiDs) as a new treatment strategy for these disorders.
Collapse
Affiliation(s)
- Yoo Jin Jung
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
- Stanford Neurosciences Interdepartmental Program, Stanford University School of Medicine, Stanford, CA, United States
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Michael T Scerba
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Dong Seok Kim
- AevisBio, Inc., Gaithersburg, MD, United States
- Aevis Bio, Inc., Daejeon, South Korea
| | | | - Augusta Pisanu
- National Research Council, Institute of Neuroscience, Cagliari, Italy
| | - Anna R Carta
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
32
|
Gastrointestinal Dysfunction in Parkinson's Disease. J Clin Med 2021; 10:jcm10030493. [PMID: 33572547 PMCID: PMC7866791 DOI: 10.3390/jcm10030493] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 12/19/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease. Patients show deposits of pathological, aggregated α-synuclein not only in the brain but throughout almost the entire length of the digestive tract. This gives rise to non-motor symptoms particularly within the gastrointestinal tract and patients experience a wide range of frequent and burdensome symptoms such as dysphagia, bloating, and constipation. Recent evidence suggests that progressive accumulation of gastrointestinal pathology is underway several years before a clinical diagnosis of PD. Notably, constipation has been shown to increase the risk of developing PD and in contrast, truncal vagotomy seems to decrease the risk of PD. Animal models have demonstrated gut-to-brain spreading of pathological α-synuclein and it is currently being intensely studied whether PD begins in the gut of some patients. Gastrointestinal symptoms in PD have been investigated by the use of several different questionnaires. However, there is limited correspondence between subjective gastrointestinal symptoms and objective dysfunction along the gastrointestinal tract, and often the magnitude of dysfunction is underestimated by the use of questionnaires. Therefore, objective measures are important tools to clarify the degree of dysfunction in future studies of PD. Here, we summarize the types and prevalence of subjective gastrointestinal symptoms and objective dysfunction in PD. The potential importance of the gastrointestinal tract in the etiopathogenesis of PD is briefly discussed.
Collapse
|
33
|
Tanei ZI, Saito Y, Ito S, Matsubara T, Motoda A, Yamazaki M, Sakashita Y, Kawakami I, Ikemura M, Tanaka S, Sengoku R, Arai T, Murayama S. Lewy pathology of the esophagus correlates with the progression of Lewy body disease: a Japanese cohort study of autopsy cases. Acta Neuropathol 2021; 141:25-37. [PMID: 33150517 PMCID: PMC7785549 DOI: 10.1007/s00401-020-02233-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 01/05/2023]
Abstract
Lewy body disease (LBD) is a spectrum of progressive neurodegenerative disorders characterized by the wide distribution of Lewy bodies and neurites in the central and peripheral nervous system (CNS, PNS). Clinical diagnoses include Parkinson's disease (PD), dementia with Lewy bodies, or pure autonomic failure. All types of LBD are accompanied by non-motor symptoms (NMSs) including gastrointestinal dysfunctions such as constipation. Its relationship to Lewy body-related α-synucleinopathy (Lewy pathology) of the enteric nervous system (ENS) is attracting attention because it can precede the motor symptoms. To clarify the role of ENS Lewy pathology in disease progression, we performed a clinicopathological study using the Brain Bank for Aging Research in Japan. Five-hundred and eighteen cases were enrolled in the study. Lewy pathology of the CNS and PNS, including the lower esophagus as a representative of the ENS, was examined via autopsy findings. Results showed that one-third of older people (178 cases, 34%) exhibited Lewy pathology, of which 78 cases (43.8%) exhibited the pathology in the esophagus. In the esophageal wall, Auerbach's plexus (41.6%) was most susceptible to the pathology, followed by the adventitia (33.1%) and Meissner's plexus (14.6%). Lewy pathology of the esophagus was significantly associated with autonomic failures such as constipation (p < 0.0001) and among PNS regions, correlated the most with LBD progression (r = 0.95, p < 0.05). These findings suggest that the propagation of esophageal Lewy pathology is a predictive factor of LBD.
Collapse
Affiliation(s)
- Zen-Ichi Tanei
- Department of Neurology and Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuko Saito
- Department of Neurology and Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Shinji Ito
- Department of Pathology, Toranomon Hospital, Tokyo, Japan
| | - Tomoyasu Matsubara
- Department of Neurology and Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Atsuko Motoda
- Department of Neurology and Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Mikihiro Yamazaki
- Department of Neurology and Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- Department of Neurology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasuhiro Sakashita
- Department of Neurology and Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- Department of Neurology and Neurobiology of Aging, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Ito Kawakami
- Department of Neurology and Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- Dementia Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masako Ikemura
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo, Japan
| | - Renpei Sengoku
- Department of Neurology and Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- Department of Neurology, The Jikei University School of Medicine, Tokyo, Japan
| | - Tomio Arai
- Department of Pathology, Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, Tokyo, Japan
| | - Shigeo Murayama
- Department of Neurology and Neuropathology (the Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology, 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan.
- Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Osaka, Japan.
| |
Collapse
|
34
|
Shin C, Park SH, Shin JH, Yun JY, Yang HK, Lee HJ, Kong SH, Suh YS, Ahn TB, Jung KY, Kim JM, Kim HJ, Jeon B. Gastric synucleinopathy as prodromal pathological biomarker in idiopathic REM sleep behaviour disorder. J Neurol Neurosurg Psychiatry 2020; 92:jnnp-2020-324743. [PMID: 33188133 DOI: 10.1136/jnnp-2020-324743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 11/04/2022]
Affiliation(s)
- Chaewon Shin
- Department of Neurology, Chungnam National University, Daejeon, South Korea
- Department of Neurology, Chungnam National University Sejong Hospital, Sejong, South Korea
| | - Sung-Hye Park
- Department of Pathology, College of Medicine, Seoul National University, Seoul, South Korea
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea
| | - Jung Hwan Shin
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | - Ji Young Yun
- Department of Neurology, Ewha Womans University Seoul Hospital, Seoul, South Korea
| | - Han-Kwang Yang
- Department of Surgery, College of Medicine, Seoul National University, Seoul, South Korea
- Department of Surgery, Seoul National University Hospital, Seoul, South Korea
| | - Hyuk-Joon Lee
- Department of Surgery, College of Medicine, Seoul National University, Seoul, South Korea
- Department of Surgery, Seoul National University Hospital, Seoul, South Korea
| | - Seong-Ho Kong
- Department of Surgery, College of Medicine, Seoul National University, Seoul, South Korea
- Department of Surgery, Seoul National University Hospital, Seoul, South Korea
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Tae-Beom Ahn
- Department of Neurology, Kyung Hee University Hospital, Seoul, South Korea
| | - Ki-Young Jung
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
- Department of Neurology, College of Medicine, Seoul National University, Seoul, South Korea
| | - Jong-Min Kim
- Department of Neurology, College of Medicine, Seoul National University, Seoul, South Korea
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Han-Joon Kim
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
- Department of Neurology, College of Medicine, Seoul National University, Seoul, South Korea
| | - Beomseok Jeon
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
- Department of Neurology, College of Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
35
|
Rouaud T, Corbillé AG, Leclair-Visonneau L, de Guilhem de Lataillade A, Lionnet A, Preterre C, Damier P, Derkinderen P. Pathophysiology of Parkinson's disease: Mitochondria, alpha-synuclein and much more…. Rev Neurol (Paris) 2020; 177:260-271. [PMID: 33032797 DOI: 10.1016/j.neurol.2020.07.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/21/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a complex, age-related, neurodegenerative disease whose pathogenesis remains incompletely understood. Here, we give an overview of the progress that has been made over the past four decades in our understanding of this disorder. We review the role of mitochondria, environmental toxicants, alpha-synuclein and neuroinflammation in the development of PD. We also discuss more recent data from genetics, which strongly support the endosomal-lysosomal pathways and mitophagy as being central to PD. Finally, we discuss the emerging role of the gut-brain axis as a modulator of PD progression. This article is intended to provide a comprehensive, general and practical review of PD pathogenesis for the general neurologist.
Collapse
Affiliation(s)
- T Rouaud
- CHU de Nantes, Centre expert Parkinson, Department of Neurology, 44093 Nantes, France.
| | - A-G Corbillé
- CHU de Nantes, Centre expert Parkinson, Department of Neurology, 44093 Nantes, France.
| | | | | | - A Lionnet
- CHU de Nantes, Centre expert Parkinson, Department of Neurology, 44093 Nantes, France.
| | - C Preterre
- CHU de Nantes, Centre expert Parkinson, Department of Neurology, 44093 Nantes, France.
| | - P Damier
- CHU de Nantes, Centre expert Parkinson, Department of Neurology, 44093 Nantes, France.
| | - P Derkinderen
- CHU de Nantes, Centre expert Parkinson, Department of Neurology, 44093 Nantes, France.
| |
Collapse
|
36
|
Schaeffer E, Kluge A, Böttner M, Zunke F, Cossais F, Berg D, Arnold P. Alpha Synuclein Connects the Gut-Brain Axis in Parkinson's Disease Patients - A View on Clinical Aspects, Cellular Pathology and Analytical Methodology. Front Cell Dev Biol 2020; 8:573696. [PMID: 33015066 PMCID: PMC7509446 DOI: 10.3389/fcell.2020.573696] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/18/2020] [Indexed: 12/15/2022] Open
Abstract
Parkinson’s disease (PD) is marked by different kinds of pathological features, one hallmark is the aggregation of α-synuclein (aSyn). The development of aSyn pathology in the substantia nigra is associated to the manifestation of motor deficits at the time of diagnosis. However, most of the patients suffer additionally from non-motor symptoms, which may occur already in the prodromal phase of the disease years before PD is diagnosed. Many of these symptoms manifest in the gastrointestinal system (GIT) and some data suggest a potential link to the occurrence of pathological aSyn forms within the GIT. These clinical and pathological findings lead to the idea of a gut-brain route of aSyn pathology in PD. The identification of pathological aSyn in the intestinal system, e.g., by GIT biopsies, is therefore of highest interest for early diagnosis and early intervention in the phase of formation and propagation of aSyn. However, reliable methods to discriminate between physiological and pathological forms of enteral aSyn on the cellular and biochemical level are still missing. Moreover, a better understanding of the physiological function of aSyn within the GIT as well as its structure and pathological aggregation pathways are crucial to understand its role within the enteric nervous system and its spreading from the gut to the brain. In this review, we summarize clinical manifestations of PD in the GIT, and discuss biochemical findings from enteral biopsies. The relevance of pathological aSyn forms, their connection to the gut-brain axis and new developments to identify pathologic forms of aSyn by structural features are critically reviewed.
Collapse
Affiliation(s)
- Eva Schaeffer
- Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Annika Kluge
- Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Martina Böttner
- Institute of Anatomy, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Friederike Zunke
- Biochemical Institute, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Francois Cossais
- Institute of Anatomy, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Daniela Berg
- Department of Neurology, Christian-Albrechts-University of Kiel, Kiel, Germany.,Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Philipp Arnold
- Institute of Anatomy, Christian-Albrechts-University of Kiel, Kiel, Germany.,MSH Medical School Hamburg, Hamburg, Germany
| |
Collapse
|
37
|
Harapan BN, Frydrychowicz C, Classen J, Wittekind C, Gradistanac T, Rumpf JJ, Mueller W. No enhanced (p-) α-synuclein deposition in gastrointestinal tissue of Parkinson's disease patients. Parkinsonism Relat Disord 2020; 80:82-88. [PMID: 32971383 DOI: 10.1016/j.parkreldis.2020.08.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/15/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Neuronal alpha-synuclein (α-Syn) aggregation in the brain is believed to be a central component of the pathogenesis of Parkinson's disease (PD). α-Syn aggregates in the gastrointestinal tract have been suggested as a potential biomarker of PD that may even signal an early event of the Parkinsonian molecular pathology. However, studies further investigating this hypothesis have produced mixed results. OBJECTIVE To determine whether the prevalence of α-Syn- and serine 129-phosphorylated α-Syn (Ser129p-α-Syn) depositions detected in intestine from PD patients differed from that of non-Parkinsonian controls. METHODS In this retrospective study, we examined post-mortem small and large intestine samples of 25 PD patients and 20 age- and sex-matched controls without PD. Specimens were taken from archived paraffin-embedded tissue blocks. Immunohistochemical techniques were applied to detect α-Syn and Ser129p-α-Syn aggregates in situ. Immunoreactivity was quantified by a new approach that employed the detailed assessment of α-Syn- and Ser129p-α-Syn-positive morphological structures of the enteric nervous system (i.e., nerve fibers, myenteric and submucous plexus as well as ganglion cells). RESULTS α-Syn immunoreactivity was a common finding in intestinal tissues from PD patients and controls. Importantly, α-Syn and Ser129p-α-Syn immunoreactivity were significantly reduced in PD patients compared to controls in each of the morphological structures examined. CONCLUSIONS Immunohistochemical detection of intestinal α-Syn and Ser129p-α-Syn seems to be a frequent and potentially normal finding. Neither α-Syn nor Ser129p-α-Syn immunoreactivity may, therefore, be regarded as a molecular intestinal biomarker of PD pathology. Reduced intestinal α-Syn and Ser129p-α-Syn immunoreactivity in PD patients rather reflect PD-related neuronal degeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wolf Mueller
- Department of Neuropathology, University of Leipzig, Germany
| |
Collapse
|
38
|
Chahine LM, Beach TG, Brumm MC, Adler CH, Coffey CS, Mosovsky S, Caspell-Garcia C, Serrano GE, Munoz DG, White CL, Crary JF, Jennings D, Taylor P, Foroud T, Arnedo V, Kopil CM, Riley L, Dave KD, Mollenhauer B. In vivo distribution of α-synuclein in multiple tissues and biofluids in Parkinson disease. Neurology 2020; 95:e1267-e1284. [PMID: 32747521 PMCID: PMC7538226 DOI: 10.1212/wnl.0000000000010404] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 02/18/2020] [Indexed: 11/16/2022] Open
Abstract
Objective The Systemic Synuclein Sampling Study (S4) measured α-synuclein in multiple tissues and biofluids within the same patients with Parkinson disease (PD) vs healthy controls (HCs). Methods S4 was a 6-site cross-sectional observational study of participants with early, moderate, or advanced PD and HCs. Motor and nonmotor measures and dopamine transporter SPECT were obtained. Biopsies of skin, colon, submandibular gland (SMG), CSF, saliva, and blood were collected. Tissue biopsy sections were stained with 5C12 monoclonal antibody against pathologic α-synuclein; digital images were interpreted by neuropathologists blinded to diagnosis. Biofluid total α-synuclein was quantified using ELISA. Results The final cohort included 59 patients with PD and 21 HCs. CSF α-synuclein was lower in patients with PD vs HCs; sensitivity/specificity of CSF α-synuclein for PD diagnosis was 87.0%/63.2%, respectively. Sensitivity of α-synuclein immunoreactivity for PD diagnosis was 56.1% for SMG and 24.1% for skin; specificity was 92.9% and 100%, respectively. There were no significant relationships between different measures of α-synuclein within participants. Conclusions S4 confirms lower total α-synuclein levels in CSF in patients with PD compared to HCs, but specificity is low. In contrast, α-synuclein immunoreactivity in skin and SMG is specific for PD but sensitivity is low. Relationships within participants across different tissues and biofluids could not be demonstrated. Measures of pathologic forms of α-synuclein with higher accuracy are critically needed. Classification of evidence This study provides Class III evidence that total CSF α-synuclein does not accurately distinguish patients with PD from HCs, and that monoclonal antibody staining for SMG and skin total α-synuclein is specific but not sensitive for PD diagnosis.
Collapse
Affiliation(s)
- Lana M Chahine
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany.
| | - Thomas G Beach
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Michael C Brumm
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Charles H Adler
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Christopher S Coffey
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Sherri Mosovsky
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Chelsea Caspell-Garcia
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Geidy E Serrano
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - David G Munoz
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Charles L White
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - John F Crary
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Danna Jennings
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Peggy Taylor
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Tatiana Foroud
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Vanessa Arnedo
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Catherine M Kopil
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Lindsey Riley
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Kuldip D Dave
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | - Brit Mollenhauer
- From the Department of Neurology (L.M.C., S.M.), University of Pittsburgh, PA; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; University of Iowa (M.C.B., C.S.C., C.C.-G.), Iowa City; Department of Neurology (C.H.A.), Mayo Clinic College of Medicine, Scottsdale, AZ; St. Michael's Hospital (D.G.M.), Toronto, Canada; University of Texas Southwestern Medical School (C.L.W.), Dallas; Icahn School of Medicine at Mount Sinai (J.F.C.), New York, NY; Institute for Neurodegenerative Disorders (D.J.), New Haven, CT; BioLegend Inc. (P.T.), Dedham, MA; Indiana University (T.F.), Indianapolis; The Michael J. Fox Foundation for Parkinson's Research (V.A., C.M.K., L.R., K.D.D.), New York, NY; and Department of Neurology (B.M.), Center of Parkinsonism and Movement Disorders Paracelsus-Elena Klinik Kassel and University Medical Center Göttingen, Germany
| | | |
Collapse
|
39
|
Chalazonitis A, Li Z, Pham TD, Chen J, Rao M, Lindholm P, Saarma M, Lindahl M, Gershon MD. Cerebral dopamine neurotrophic factor is essential for enteric neuronal development, maintenance, and regulation of gastrointestinal transit. J Comp Neurol 2020; 528:2420-2444. [PMID: 32154930 DOI: 10.1002/cne.24901] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/19/2020] [Accepted: 03/02/2020] [Indexed: 12/25/2022]
Abstract
Cerebral dopamine neurotrophic factor (CDNF) is expressed in the brain and is neuroprotective. We have previously shown that CDNF is also expressed in the bowel and that its absence leads to degeneration and autophagy in the enteric nervous system (ENS), particularly in the submucosal plexus. We now demonstrate that enteric CDNF immunoreactivity is restricted to neurons (submucosal > myenteric) and is not seen in glia, interstitial cells of Cajal, or smooth muscle. Expression of CDNF, moreover, is essential for the normal development and survival of enteric dopaminergic neurons; thus, expression of the dopaminergic neuronal markers, dopamine, tyrosine hydroxylase, and dopamine transporter are deficient in the ileum of Cdnf -/- mice. The normal age-related decline in proportions of submucosal dopaminergic neurons is exacerbated in Cdnf -/- animals. The defect in Cdnf -/- animals is not dopamine-restricted; proportions of other submucosal neurons (NOS-, GABA-, and CGRP-expressing), are also deficient. The deficits in submucosal neurons are reflected functionally in delayed gastric emptying, slowed colonic motility, and prolonged total gastrointestinal transit. CDNF is expressed selectively in isolated enteric neural crest-derived cells (ENCDC), which also express the dopamine-related transcription factor Foxa2. Addition of CDNF to ENCDC promotes development of dopaminergic neurons; moreover, survival of these neurons becomes CDNF-dependent after exposure to bone morphogenetic protein 4. The effects of neither glial cell-derived neurotrophic factor (GDNF) nor serotonin are additive with CDNF. We suggest that CDNF plays a critical role in development and long-term maintenance of dopaminergic and other sets of submucosal neurons.
Collapse
Affiliation(s)
- Alcmène Chalazonitis
- Department of Pathology and Cell Biology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York
| | - ZhiShan Li
- Department of Pathology and Cell Biology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York
| | - Tuan D Pham
- Department of Pathology and Cell Biology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York
| | - Jason Chen
- Department of Pathology and Cell Biology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York
| | - Meenakshi Rao
- Department of Pediatrics, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York
| | - Päivi Lindholm
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Maria Lindahl
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Michael D Gershon
- Department of Pathology and Cell Biology, Columbia University, Vagelos College of Physicians and Surgeons, New York, New York
| |
Collapse
|
40
|
de Guilhem de Lataillade A, Lebouvier T, Noble W, Leclair-Visonneau L, Derkinderen P. Enteric synucleinopathy: from trendy concept to real entity. FREE NEUROPATHOLOGY 2020; 1:26. [PMID: 37283671 PMCID: PMC10209962 DOI: 10.17879/freeneuropathology-2020-2920] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 08/23/2020] [Indexed: 06/08/2023]
Abstract
An accumulating body of literature has emerged in the past 25 years to show that Parkinson's disease (PD) is not only a disorder of the brain but also of the gastrointestinal tract and more generally of the gut-brain axis. Gastrointestinal symptoms occur in almost every PD patient at some point and in nearly every case examined pathologically autopsy studies find alpha-synuclein deposits, the pathological hallmarks of PD, in the enteric nervous system. This concept of 'enteric synucleinopathy' led to the hypothesis that the enteric nervous system might play a pivotal role in the initiation and spreading of PD. Although this hypothesis opens up interesting perspectives on the pathogenesis of neurodegenerative disorders, some important questions are still pending. The present opinion paper describes and compares the physiological and pathophysiological properties of alpha-synuclein in the brain and the enteric nervous system. We conclude that the existing data supports the existence of pathological alpha-synuclein species in the gut in PD. We also discuss if gut-brain interactions are important in other neurodegenerative disorders.
Collapse
Affiliation(s)
- Adrien de Guilhem de Lataillade
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
- CHU Nantes, Department of Neurology, Nantes, F-44093, France
| | - Thibaud Lebouvier
- Univ. Lille, Inserm URM_S1172, CHU Lille, DistAlz, Licend, F-59000 Lille, France
| | - Wendy Noble
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, 5 Cutcombe Road, Camberwell, London. SE5 9RX, UK
| | - Laurène Leclair-Visonneau
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Pascal Derkinderen
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
- CHU Nantes, Department of Neurology, Nantes, F-44093, France
| |
Collapse
|
41
|
Coughlin DG, Hurtig H, Irwin DJ. Pathological Influences on Clinical Heterogeneity in Lewy Body Diseases. Mov Disord 2020; 35:5-19. [PMID: 31660655 PMCID: PMC7233798 DOI: 10.1002/mds.27867] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/06/2019] [Accepted: 09/03/2019] [Indexed: 12/11/2022] Open
Abstract
PD, PD with dementia, and dementia with Lewy bodies are clinical syndromes characterized by the neuropathological accumulation of alpha-synuclein in the CNS that represent a clinicopathological spectrum known as Lewy body disorders. These clinical entities have marked heterogeneity of motor and nonmotor symptoms with highly variable disease progression. The biological basis for this clinical heterogeneity remains poorly understood. Previous attempts to subtype patients within the spectrum of Lewy body disorders have centered on clinical features, but converging evidence from studies of neuropathology and ante mortem biomarkers, including CSF, neuroimaging, and genetic studies, suggest that Alzheimer's disease beta-amyloid and tau copathology strongly influence clinical heterogeneity and prognosis in Lewy body disorders. Here, we review previous clinical biomarker and autopsy studies of Lewy body disorders and propose that Alzheimer's disease copathology is one of several likely pathological contributors to clinical heterogeneity of Lewy body disorders, and that such pathology can be assessed in vivo. Future work integrating harmonized assessments and genetics in PD, PD with dementia, and dementia with Lewy bodies patients followed to autopsy will be critical to further refine the classification of Lewy body disorders into biologically distinct endophenotypes. This approach will help facilitate clinical trial design for both symptomatic and disease-modifying therapies to target more homogenous subsets of Lewy body disorders patients with similar prognosis and underlying biology. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- David G Coughlin
- University of Pennsylvania Health System, Department of Neurology
- Digital Neuropathology Laboratory
- Lewy Body Disease Research Center of Excellence
| | - Howard Hurtig
- University of Pennsylvania Health System, Department of Neurology
| | - David J Irwin
- University of Pennsylvania Health System, Department of Neurology
- Digital Neuropathology Laboratory
- Lewy Body Disease Research Center of Excellence
- Frontotemporal Degeneration Center, Philadelphia PA, USA 19104
| |
Collapse
|
42
|
Lindahl M, Chalazonitis A, Palm E, Pakarinen E, Danilova T, Pham TD, Setlik W, Rao M, Võikar V, Huotari J, Kopra J, Andressoo JO, Piepponen PT, Airavaara M, Panhelainen A, Gershon MD, Saarma M. Cerebral dopamine neurotrophic factor-deficiency leads to degeneration of enteric neurons and altered brain dopamine neuronal function in mice. Neurobiol Dis 2019; 134:104696. [PMID: 31783118 PMCID: PMC7000201 DOI: 10.1016/j.nbd.2019.104696] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/07/2019] [Accepted: 11/24/2019] [Indexed: 12/13/2022] Open
Abstract
Cerebral dopamine neurotrophic factor (CDNF) is neuroprotective for nigrostriatal dopamine neurons and restores dopaminergic function in animal models of Parkinson’s disease (PD). To understand the role of CDNF in mammals, we generated CDNF knockout mice (Cdnf−/−), which are viable, fertile, and have a normal life-span. Surprisingly, an age-dependent loss of enteric neurons occurs selectively in the submucosal but not in the myenteric plexus. This neuronal loss is a consequence not of increased apoptosis but of neurodegeneration and autophagy. Quantitatively, the neurodegeneration and autophagy found in the submucosal plexus in duodenum, ileum and colon of the Cdnf−/− mouse are much greater than in those of Cdnf+/+ mice. The selective vulnerability of submucosal neurons to the absence of CDNF is reminiscent of the tendency of pathological abnormalities to occur in the submucosal plexus in biopsies of patients with PD. In contrast, the number of substantia nigra dopamine neurons and dopamine and its metabolite concentrations in the striatum are unaltered in Cdnf−/− mice; however, there is an age-dependent deficit in the function of the dopamine system in Cdnf−/− male mice analyzed. This is observed as D-amphetamine-induced hyperactivity, aberrant dopamine transporter function, and as increased D-amphetamine-induced dopamine release demonstrating that dopaminergic axon terminal function in the striatum of the Cdnf−/− mouse brain is altered. The deficiencies of Cdnf−/− mice, therefore, are reminiscent of those seen in early stages of Parkinson’s disease.
Collapse
Affiliation(s)
- Maria Lindahl
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland.
| | | | - Erik Palm
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Emmi Pakarinen
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Tatiana Danilova
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Tuan D Pham
- Department of Pathology & Cell Biology, Columbia University, NY, New York, USA
| | - Wanda Setlik
- Department of Pathology & Cell Biology, Columbia University, NY, New York, USA
| | - Meenakshi Rao
- Department of Pathology & Cell Biology, Columbia University, NY, New York, USA
| | - Vootele Võikar
- Neuroscience Center/Laboratory Animal Center, Mustialankatu 1, FI-00014, University of Helsinki, Finland
| | - Jatta Huotari
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Jaakko Kopra
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, Viikinkaari 5E, FI-00014, University of Helsinki, Finland
| | - Jaan-Olle Andressoo
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Petteri T Piepponen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, Viikinkaari 5E, FI-00014, University of Helsinki, Finland
| | - Mikko Airavaara
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Anne Panhelainen
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| | - Michael D Gershon
- Department of Pathology & Cell Biology, Columbia University, NY, New York, USA
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE Unit, Viikinkaari 5D, FI-00014, University of Helsinki, Finland
| |
Collapse
|
43
|
Scheperjans F, Derkinderen P, Borghammer P. The Gut and Parkinson's Disease: Hype or Hope? JOURNAL OF PARKINSONS DISEASE 2019; 8:S31-S39. [PMID: 30584161 PMCID: PMC6311363 DOI: 10.3233/jpd-181477] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In the last two decades it has become clear that Parkinson’s disease (PD) is associated with a plethora of gastrointestinal symptoms originating from functional and structural changes in the gut and its associated neural structures. This is of particular interest not only because such symptoms have a major impact on the quality of life of PD patients, but also since accumulating evidence suggests that in at least a subgroup of patients, these disturbances precede the motor symptoms and diagnosis of PD by years and may thus give important insights into the origin and pathogenesis of the disease. In this mini-review we attempt to concisely summarize the current knowledge after two decades of research on the gut-brain axis in PD. We focus on alpha-synuclein pathology, biomarkers, and the gut microbiota and envision the development and impact of these research areas for the two decades to come.
Collapse
Affiliation(s)
- Filip Scheperjans
- Department of Neurology, Helsinki University Hospital and Department of Clinical Neurosciences (Neurology), University of Helsinki, Helsinki, Finland
| | | | - Per Borghammer
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
44
|
Killinger BA, Madaj Z, Sikora JW, Rey N, Haas AJ, Vepa Y, Lindqvist D, Chen H, Thomas PM, Brundin P, Brundin L, Labrie V. The vermiform appendix impacts the risk of developing Parkinson's disease. Sci Transl Med 2019; 10:10/465/eaar5280. [PMID: 30381408 DOI: 10.1126/scitranslmed.aar5280] [Citation(s) in RCA: 200] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/14/2018] [Accepted: 06/08/2018] [Indexed: 12/12/2022]
Abstract
The pathogenesis of Parkinson's disease (PD) involves the accumulation of aggregated α-synuclein, which has been suggested to begin in the gastrointestinal tract. Here, we determined the capacity of the appendix to modify PD risk and influence pathogenesis. In two independent epidemiological datasets, involving more than 1.6 million individuals and over 91 million person-years, we observed that removal of the appendix decades before PD onset was associated with a lower risk for PD, particularly for individuals living in rural areas, and delayed the age of PD onset. We also found that the healthy human appendix contained intraneuronal α-synuclein aggregates and an abundance of PD pathology-associated α-synuclein truncation products that are known to accumulate in Lewy bodies, the pathological hallmark of PD. Lysates of human appendix tissue induced the rapid cleavage and oligomerization of full-length recombinant α-synuclein. Together, we propose that the normal human appendix contains pathogenic forms of α-synuclein that affect the risk of developing PD.
Collapse
Affiliation(s)
- Bryan A Killinger
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Zachary Madaj
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Jacek W Sikora
- Proteomics Center of Excellence, Northwestern University, Evanston, IL, USA
| | - Nolwen Rey
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA.,Paris-Saclay Institute of Neuroscience, Centre National de la Recherche Scientifique, 91190 Gif-sur-Yvette, France
| | - Alec J Haas
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Yamini Vepa
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Daniel Lindqvist
- Department of Clinical Sciences, Psychiatry, Faculty of Medicine, Lund University, Lund, Sweden.,Psychiatric Clinic, Lund, Division of Psychiatry, Lund, Sweden
| | - Honglei Chen
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Paul M Thomas
- Proteomics Center of Excellence, Northwestern University, Evanston, IL, USA
| | - Patrik Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Lena Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Viviane Labrie
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA. .,Centre for Addiction and Mental Health, Toronto, ON, Canada
| |
Collapse
|
45
|
Shin C, Yang HK, Park S, Lee HJ, Kong SH, Suh YS, Huh YJ, Kim YJ, Park SY, Ahn TB, Lee SH, Kim HJ, Jeon B. Prospective cohort study of patients with early gastric cancer to detect prodromal Parkinson disease (EGC-PPD): A study protocol and baseline characteristics. J Clin Neurosci 2019; 66:26-32. [DOI: 10.1016/j.jocn.2019.05.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 05/21/2019] [Indexed: 01/27/2023]
|
46
|
Tsukita K, Sakamaki-Tsukita H, Tanaka K, Suenaga T, Takahashi R. Value of in vivo α-synuclein deposits in Parkinson's disease: A systematic review and meta-analysis. Mov Disord 2019; 34:1452-1463. [PMID: 31322768 DOI: 10.1002/mds.27794] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 01/06/2023] Open
Abstract
Previous studies that have investigated the potential of in vivo abnormal α-synuclein deposits as a pathological biomarker for PD included few participants and reported different diagnostic accuracies. Here, we aimed to confirm the diagnostic value of in vivo α-synuclein deposits in PD through a systematic review and meta-analysis, with special emphasis on determining the tissue most suitable for examination and assessing whether anti-native α-synuclein or anti-phosphorylated α-synuclein antibodies should be used. Databases were searched on December 30, 2018. We finally included 41 case-control studies that examined in vivo tissue samples using anti-native α-synuclein or anti-phosphorylated α-synuclein antibody in PD patients and controls. Using a univariate random-effects model, pooled sensitivity and specificity (95% confidence interval) of anti-native α-synuclein antibody were 0.54 (0.49-0.60) and 0.72 (0.68-0.76) for the gastrointestinal tract and 0.76 (0.60-0.89) and 0.60 (0.43-0.74) for the skin. Pooled sensitivity and specificity (95% confidence interval) of anti-phosphorylated α-synuclein antibody were 0.43 (0.37-0.48) and 0.82 (0.78-0.86) for the gastrointestinal tract, 0.76 (0.69-0.82) and 1.00 (0.98-1.00) for the skin, 0.42 (0.26-0.59) and 0.94 (0.84-0.99) for the minor salivary glands, and 0.66 (0.51-0.79) and 0.96 (0.86-1.00) for the submandibular glands. Although ubiquitous heterogeneity between the included studies should be noted when interpreting our results, our analyses demonstrated the following: (1) in vivo α-synuclein immunoreactivity has the potential as a pathological biomarker for PD; (2) anti-phosphorylated α-synuclein antibody consistently has higher specificity than anti-native α-synuclein antibody; and (3) skin biopsy examination using anti-phosphorylated α-synuclein antibody has the best diagnostic accuracy, although feasibility remains an important issue. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Kazuto Tsukita
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Neurology, Tenri Hospital, Tenri, Japan.,Laboratory of Biological Science, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Haruhi Sakamaki-Tsukita
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Neurology, Tenri Hospital, Tenri, Japan
| | - Kanta Tanaka
- Department of Neurology, Tenri Hospital, Tenri, Japan.,Division of Stroke Care Unit, National Cerebral and Cardiovascular Center, Suita, Japan
| | | | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
47
|
Chiang HL, Lin CH. Altered Gut Microbiome and Intestinal Pathology in Parkinson's Disease. J Mov Disord 2019; 12:67-83. [PMID: 31158941 PMCID: PMC6547039 DOI: 10.14802/jmd.18067] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 02/20/2019] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder arising from an interplay between genetic and environmental risk factors. Studies have suggested that the pathological hallmarks of intraneuronal α-synuclein aggregations may start from the olfactory bulb and the enteric nervous system of the gut and later propagate to the brain via the olfactory tract and the vagus nerve. This hypothesis correlates well with clinical symptoms, such as constipation, that may develop up to 20 years before the onset of PD motor symptoms. Recent interest in the gut-brain axis has led to vigorous research into the gastrointestinal pathology and gut microbiota changes in patients with PD. In this review, we provide current clinical and pathological evidence of gut involvement in PD by summarizing the changes in gut microbiota composition and gut inflammation associated with its pathogenesis.
Collapse
Affiliation(s)
- Han-Lin Chiang
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
48
|
Bu J, Liu J, Liu K, Wang Z. Diagnostic utility of gut α-synuclein in Parkinson's disease: A systematic review and meta-analysis. Behav Brain Res 2019; 364:340-347. [PMID: 30802532 DOI: 10.1016/j.bbr.2019.02.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/22/2019] [Accepted: 02/22/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The accumulation of misfolded α-synuclein in the brain is associated with Parkinson's disease. However, the association between gut α-synuclein and PD and diagnostic value of α-synuclein in the gut still remain controversial. METHODS A literature search from inception to June 2018 was conducted, yielding 21 studies eligible for a systematic review and meta-analysis. We included studies that reported data on gut α-synuclein or phosphorylated α-synuclein in patients with PD and controls. The odds ratio(OR) was pooled using random-effect models, and heterogeneity was reported as I2. Studies were assessed for quality using the Newcastle-Ottawa Scale. Sensitivity and specificity analyses were performed using the summary receiver operating characteristics curve approach. RESULTS The database search yielded 395 results, of which 21 studies were deemed relevant. The PD group had a pooled OR of 10.01 (95% CI: 3.15-31.82, I2 = 72%) for gut α-synuclein compared with the control group. Sensitivity and specificity for distinguishing PD from controls were 0.568 and 0.819, respectively, for the colon, and 0.579 and 0.822, respectively, for phosphorylated α-synuclein. CONCLUSION Heterogeneity was high in most of the studies. This systematic review and meta-analysis confirmed a high degree of association between gut α-synuclein and PD, which suggested that gut α-synuclein is a potential therapeutic intervention. The measurement of gut α-synuclein alone could lead to the underdiagnosis of PD. Future research combining gut α-synuclein with other biochemical markers could improve the standardization of current assays.
Collapse
Affiliation(s)
- Jin Bu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022,China
| | - Jinsong Liu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kun Liu
- Institution of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhaohui Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022,China.
| |
Collapse
|
49
|
Kim JY, Illigens BM, McCormick MP, Wang N, Gibbons CH. Alpha-Synuclein in Skin Nerve Fibers as a Biomarker for Alpha-Synucleinopathies. J Clin Neurol 2019; 15:135-142. [PMID: 30938106 PMCID: PMC6444158 DOI: 10.3988/jcn.2019.15.2.135] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/01/2018] [Accepted: 10/02/2018] [Indexed: 12/05/2022] Open
Abstract
The common pathological features of synucleinopathies are abnormal aggregates of the synaptic protein alpha-synuclein (αSN) in the cytoplasm of neurons or glia. These abnormal aggregates appear several years before the onset of clinical manifestations, and so the early detection of αSN in body fluids or peripheral tissues (e.g., cerebrospinal fluid, colonic mucosa, salivary glands, and skin) is considered a potential tool for identifying synucleinopathies. Performing a skin biopsy is a practical option because it is a relatively noninvasive, safe, and reliable method to measure αSN deposition in the peripheral nervous system. Moreover, there is growing research interest in the use of cutaneous synuclein deposition as a biomarker for synucleinopathies. The aim of this study was to interpret the current data on cutaneous αSN deposition and present the current perspectives and future prospects.
Collapse
Affiliation(s)
- Jee Young Kim
- Department of Neurology, Myongji Hospital, Hanyang University Medical Center, Goyang, Korea.
| | - Ben Mw Illigens
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Michael P McCormick
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ningshan Wang
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christopher H Gibbons
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
50
|
Killinger B, Labrie V. The Appendix in Parkinson's Disease: From Vestigial Remnant to Vital Organ? JOURNAL OF PARKINSON'S DISEASE 2019; 9:S345-S358. [PMID: 31609697 PMCID: PMC6839473 DOI: 10.3233/jpd-191703] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 09/13/2019] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) has long been considered a brain disease, but studies now point to the gastrointestinal (GI) tract as a potential starting point for PD. In particular, the human vermiform appendix has been implicated in PD. The appendix is a tissue rich in immune cells, serving as part of the gut-associated lymphoid tissue and as a storehouse for the gut microbiome. The functions of the appendix converge with recent evidence demonstrating that gut inflammation and shifts in the microbiome are linked to PD. Some epidemiological studies have linked removal of the appendix to lowered PD risk, though there is controversy among these associations. What is apparent is that there is an abundance of aggregated forms of α-synuclein in the appendix relevant to PD pathology. α-Synuclein pathology is thought to propagate from gut to brain via the vagus nerve, which innervates GI tract locations, including the appendix. Remarkably, α-synuclein aggregates in the appendix occur not only in PD patients, but are also present in healthy individuals. This has led to the proposal that in the appendix α-synuclein aggregates are not unique to PD. Moreover, the molecular events leading to PD and the mechanisms by which α-synuclein aggregates transfers from gut to brain may be identifiable in the human appendix. The influence of the appendix on GI inflammation, autoimmunity, microbiome storage, and the lymphatic system may be yet unexplored mechanisms by which the appendix contributes to PD. Overall, the appendix represents a promising tissue site to advance our understanding of PD pathobiology.
Collapse
Affiliation(s)
- Bryan Killinger
- Department of Neurological Sciences, Rush Medical College, Chicago, IL, USA
| | - Viviane Labrie
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
- Division of Psychiatry and Behavioral Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| |
Collapse
|