1
|
Feng M, Zou Z, Shou P, Peng W, Liu M, Li X. Gut microbiota and Parkinson's disease: potential links and the role of fecal microbiota transplantation. Front Aging Neurosci 2024; 16:1479343. [PMID: 39679259 PMCID: PMC11638248 DOI: 10.3389/fnagi.2024.1479343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide and seriously affects the quality of life of elderly patients. PD is characterized by the loss of dopaminergic neurons in the substantia nigra as well as abnormal accumulation of α-synuclein in neurons. Recent research has deepened our understanding of the gut microbiota, revealing that it participates in the pathological process of PD through the gut-brain axis, suggesting that the gut may be the source of PD. Therefore, studying the relationship between gut microbiota and PD is crucial for improving our understanding of the disease's prevention, diagnosis, and treatment. In this review, we first describe the bidirectional regulation of the gut-brain axis by the gut microbiota and the mechanisms underlying the involvement of gut microbiota and their metabolites in PD. We then summarize the different species of gut microbiota found in patients with PD and their correlations with clinical symptoms. Finally, we review the most comprehensive animal and human studies on treating PD through fecal microbiota transplantation (FMT), discussing the challenges and considerations associated with this treatment approach.
Collapse
Affiliation(s)
- Maosen Feng
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- Department of Gastroenterology, National Clinical Key Specialty, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- School of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Zhiyan Zou
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Pingping Shou
- School of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Wei Peng
- Department of Gastroenterology, National Clinical Key Specialty, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Mingxue Liu
- School of Life Sciences and Engineering, Southwest University of Science and Technology, Mianyang, China
| | - Xiaoan Li
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
- Department of Gastroenterology, National Clinical Key Specialty, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| |
Collapse
|
2
|
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
|
3
|
Guimarães RP, de Resende MCS, Tavares MM, Belardinelli de Azevedo C, Ruiz MCM, Mortari MR. Construct, Face, and Predictive Validity of Parkinson's Disease Rodent Models. Int J Mol Sci 2024; 25:8971. [PMID: 39201659 PMCID: PMC11354451 DOI: 10.3390/ijms25168971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease globally. Current drugs only alleviate symptoms without halting disease progression, making rodent models essential for researching new therapies and understanding the disease better. However, selecting the right model is challenging due to the numerous models and protocols available. Key factors in model selection include construct, face, and predictive validity. Construct validity ensures the model replicates pathological changes seen in human PD, focusing on dopaminergic neurodegeneration and a-synuclein aggregation. Face validity ensures the model's symptoms mirror those in humans, primarily reproducing motor and non-motor symptoms. Predictive validity assesses if treatment responses in animals will reflect those in humans, typically involving classical pharmacotherapies and surgical procedures. This review highlights the primary characteristics of PD and how these characteristics are validated experimentally according to the three criteria. Additionally, it serves as a valuable tool for researchers in selecting the most appropriate animal model based on established validation criteria.
Collapse
Affiliation(s)
- Rayanne Poletti Guimarães
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Maria Clara Souza de Resende
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Miguel Mesquita Tavares
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Caio Belardinelli de Azevedo
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| | - Miguel Cesar Merino Ruiz
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
- Neurological Rehabilitation Unit, Sarah Network of Rehabilitation Hospitals, Brasília 70335-901, Brazil
| | - Márcia Renata Mortari
- Neuropharma Lab, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília 70910-900, Brazil; (R.P.G.); (M.C.S.d.R.); (M.M.T.); (C.B.d.A.); (M.C.M.R.)
| |
Collapse
|
4
|
Zhang C, Chen T, Fan M, Tian J, Zhang S, Zhao Z, Liu X, Ma H, Yang L, Chen Y. Electroacupuncture improves gastrointestinal motility through a central-cholinergic pathway-mediated GDNF releasing from intestinal glial cells to protect intestinal neurons in Parkinson's disease rats. Neurotherapeutics 2024; 21:e00369. [PMID: 38744625 PMCID: PMC11305299 DOI: 10.1016/j.neurot.2024.e00369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/18/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024] Open
Abstract
Constipation symptoms of Parkinson's disease (PD) seriously reduce the quality of life of patients and aggravate the development of the disease, but current treatment options still cannot alleviate the progress of constipation. Electroacupuncture (EA) is a new method for the treatment of constipation, which can effectively treat the symptoms of constipation in PD patients. However, the specific regulatory mechanisms of EA in the treatment of constipation symptoms in PD remain unclear. The aim of this study is to investigate the therapeutic effect of EA on PD constipation rats and its regulatory mechanism. A rotenone (ROT)-induced gastrointestinal motility disorder model was used to simulate the pathological process of constipation in PD. The results showed that EA could effectively promote gastrointestinal peristalsis, reduce α-synuclein accumulation in substantia nigra and colon and colonic injury in rats after ROT administration. Mechanistically, EA activation of the central-cholinergic pathway increases acetylcholine release in the colon. At the same time, EA up-regulated the co-expression of enteric glial cells (EGCs) and α7 nicotinic acetylcholine receptor (α7nAChR). EA increased the expression of choline acetyltransferase (ChAT), neuronal nitric oxide synthase (nNOS), and tyrosine hydroxylase (TH) in the colon of PD rats. Further mechanistic studies showed that EA increased the expression of glial cell-derived neurotrophic factor (GDNF), GFRa1 and p-AKT in colon tissues. The present study confirmed that EA upregulates α7nAChR through a central-cholinergic mechanism to promote GDNF release from EGCs, thereby protecting intestinal neurons and thereby improving gastrointestinal motility.
Collapse
Affiliation(s)
- Can Zhang
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Tan Chen
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Mingwei Fan
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Jinlan Tian
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Shuhui Zhang
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Zijian Zhao
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xinru Liu
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Huaiyuan Ma
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lijuan Yang
- Center Research Institute, Binzhou Medical University Hospital, Binzhou, China
| | - Yan Chen
- Institute of Digestive Diseases, Binzhou Medical University Hospital, Binzhou, Shandong, China; Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China.
| |
Collapse
|
5
|
Yang Y, Zhang Z. α-Synuclein pathology from the body to the brain: so many seeds so close to the central soil. Neural Regen Res 2024; 19:1463-1472. [PMID: 38051888 PMCID: PMC10883481 DOI: 10.4103/1673-5374.387967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/24/2023] [Indexed: 12/07/2023] Open
Abstract
ABSTRACT α-Synuclein is a protein that mainly exists in the presynaptic terminals. Abnormal folding and accumulation of α-synuclein are found in several neurodegenerative diseases, including Parkinson's disease. Aggregated and highly phosphorylated α-synuclein constitutes the main component of Lewy bodies in the brain, the pathological hallmark of Parkinson's disease. For decades, much attention has been focused on the accumulation of α-synuclein in the brain parenchyma rather than considering Parkinson's disease as a systemic disease. Recent evidence demonstrates that, at least in some patients, the initial α-synuclein pathology originates in the peripheral organs and spreads to the brain. Injection of α-synuclein preformed fibrils into the gastrointestinal tract triggers the gut-to-brain propagation of α-synuclein pathology. However, whether α-synuclein pathology can occur spontaneously in peripheral organs independent of exogenous α-synuclein preformed fibrils or pathological α-synuclein leakage from the central nervous system remains under investigation. In this review, we aimed to summarize the role of peripheral α-synuclein pathology in the pathogenesis of Parkinson's disease. We also discuss the pathways by which α-synuclein pathology spreads from the body to the brain.
Collapse
Affiliation(s)
- Yunying Yang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
6
|
Naik B, Sasikumar J, Das SP. From Skin and Gut to the Brain: The Infectious Journey of the Human Commensal Fungus Malassezia and Its Neurological Consequences. Mol Neurobiol 2024:10.1007/s12035-024-04270-w. [PMID: 38871941 DOI: 10.1007/s12035-024-04270-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
The human mycobiome encompasses diverse communities of fungal organisms residing within the body and has emerged as a critical player in shaping health and disease. While extensive research has focused on the skin and gut mycobiome, recent investigations have pointed toward the potential role of fungal organisms in neurological disorders. Among those fungal organisms, the presence of the commensal fungus Malassezia in the brain has created curiosity because of its commensal nature and primary association with the human skin and gut. This budding yeast is responsible for several diseases, such as Seborrheic dermatitis, Atopic dermatitis, Pityriasis versicolor, Malassezia folliculitis, dandruff, and others. However recent findings surprisingly show the presence of Malassezia DNA in the brain and have been linked to diseases like Alzheimer's disease, Parkinson's disease, Multiple sclerosis, and Amyotrophic lateral sclerosis. The exact role of Malassezia in these disorders is unknown, but its ability to infect human cells, travel through the bloodstream, cross the blood-brain barrier, and reside along with the lipid-rich neuronal cells are potential mechanisms responsible for pathogenesis. This also includes the induction of pro-inflammatory cytokines, disruption of the blood-brain barrier, gut-microbe interaction, and accumulation of metabolic changes in the brain environment. In this review, we discuss these key findings from studies linking Malassezia to neurological disorders, emphasizing the complex and multifaceted nature of these cases. Furthermore, we discuss potential mechanisms through which Malassezia might contribute to the development of neurological conditions. Future investigations will open up new avenues for our understanding of the fungal gut-brain axis and how it influences human behavior. Collaborative research efforts among microbiologists, neuroscientists, immunologists, and clinicians hold promise for unraveling the enigmatic connections between human commensal Malassezia and neurological disorders.
Collapse
Affiliation(s)
- Bharati Naik
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Jayaprakash Sasikumar
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Shankar Prasad Das
- Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India.
| |
Collapse
|
7
|
Dautan D, Paslawski W, Montejo SG, Doyon DC, Marangiu R, Kaplitt MG, Chen R, Dawson VL, Zhang X, Dawson TM, Svenningsson P. Gut-Initiated Alpha Synuclein Fibrils Drive Parkinson's Disease Phenotypes: Temporal Mapping of non-Motor Symptoms and REM Sleep Behavior Disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.22.590542. [PMID: 38712208 PMCID: PMC11071367 DOI: 10.1101/2024.04.22.590542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Parkinson's disease (PD) is characterized by progressive motor as well as less recognized non-motor symptoms that arise often years before motor manifestation, including sleep and gastrointestinal disturbances. Despite the heavy burden on the patient's quality of life, these non-motor manifestations are poorly understood. To elucidate the temporal dynamics of the disease, we employed a mouse model involving injection of alpha-synuclein (αSyn) pre-formed fibrils (PFF) in the duodenum and antrum as a gut-brain model of Parkinsonism. Using anatomical mapping of αSyn-PFF propagation and behavioral and physiological characterizations, we unveil a correlation between post-injection time the temporal dynamics of αSyn propagation and non-motor/motor manifestations of the disease. We highlight the concurrent presence of αSyn aggregates in key brain regions, expressing acetylcholine or dopamine, involved in sleep duration, wakefulness, and particularly REM-associated atonia corresponding to REM behavioral disorder-like symptoms. This study presents a novel and in-depth exploration into the multifaceted nature of PD, unraveling the complex connections between α-synucleinopathies, gut-brain connectivity, and the emergence of non-motor phenotypes.
Collapse
Affiliation(s)
- Daniel Dautan
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Wojciech Paslawski
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Sergio G. Montejo
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Daniel C. Doyon
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Roberta Marangiu
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Division of Neurosurgery, Department of Neurosurgery, New-York Hospital-Cornell Medical College, New York, NY, USA
| | - Michael G. Kaplitt
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Division of Neurosurgery, Department of Neurosurgery, New-York Hospital-Cornell Medical College, New York, NY, USA
| | - Rong Chen
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Valina L. Dawson
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Xiaoaun Zhang
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Ted M. Dawson
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
8
|
Pellegrini C, Travagli RA. Gastrointestinal dysmotility in rodent models of Parkinson's disease. Am J Physiol Gastrointest Liver Physiol 2024; 326:G345-G359. [PMID: 38261717 PMCID: PMC11212145 DOI: 10.1152/ajpgi.00225.2023] [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: 10/09/2023] [Revised: 01/09/2024] [Accepted: 01/22/2024] [Indexed: 01/25/2024]
Abstract
Multiple studies describe prodromal, nonmotor dysfunctions that affect the quality of life of patients who subsequently develop Parkinson's disease (PD). These prodromal dysfunctions comprise a wide array of autonomic issues, including severe gastrointestinal (GI) motility disorders such as dysphagia, delayed gastric emptying, and chronic constipation. Indeed, strong evidence from studies in humans and animal models suggests that the GI tract and its neural, mainly vagal, connection to the central nervous system (CNS) could have a major role in the etiology of PD. In fact, misfolded α-synuclein aggregates that form Lewy bodies and neurites, i.e., the histological hallmarks of PD, are detected in the enteric nervous system (ENS) before clinical diagnosis of PD. The aim of the present review is to provide novel insights into the pathogenesis of GI dysmotility in PD, focusing our attention on functional, neurochemical, and molecular alterations in animal models.
Collapse
Affiliation(s)
- Carolina Pellegrini
- Unit of Histology and Medical Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | |
Collapse
|
9
|
Smith R, Hovren H, Bowser R, Bakkar N, Garruto R, Ludolph A, Ravits J, Gaertner L, Murphy D, Lebovitz R. Misfolded alpha-synuclein in amyotrophic lateral sclerosis: Implications for diagnosis and treatment. Eur J Neurol 2024; 31:e16206. [PMID: 38270442 PMCID: PMC11235862 DOI: 10.1111/ene.16206] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 11/30/2023] [Accepted: 12/28/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND Alpha-synuclein (α-Syn) oligomers and fibrils have been shown to augment the aggregation of TAR DNA-binding Protein 43 (TDP-43) monomers in vitro, supporting the idea that TDP-43 proteinopathies such as ALS may be modulated by the presence of toxic forms of α-Syn. Recently, parkinsonian features were reported in a study of European patients and Lewy bodies have been demonstrated pathologically in a similar series of patients. Based on these and other considerations, we sought to determine whether seed-competent α-Syn can be identified in spinal fluid of patients with ALS including familial, sporadic, and Guamanian forms of the disease. METHODS Based on the finding that α-Syn has been found to be a prion-like protein, we have utilized a validated α-Synuclein seed amplification assay to determine if seed-competent α-Syn could be detected in the spinal fluid of patients with ALS. RESULTS Toxic species of α-Syn were detected in CSF in 18 of 127 ALS patients, 5 of whom were from Guam. Two out of twenty six samples from patients with C9orf72 variant ALS had positive seed-amplification assays (SAAs). No positive tests were noted in superoxide dismutase type 1 ALS subjects (n = 14). The SAA was negative in 31 control subjects. CONCLUSIONS Our findings suggest that a sub-group of ALS occurs in which self-replicating α-Syn is detectable and likely contributes to its pathogenesis. This finding may have implications for the diagnosis and treatment of this disorder.
Collapse
Affiliation(s)
| | - Hanna Hovren
- Amprion Clinical LaboratorySan DiegoCaliforniaUSA
| | | | | | | | | | - John Ravits
- University of California, San DiegoLa JollaCaliforniaUSA
| | - Lia Gaertner
- Bay Area Lyme Disease FoundationPortola ValleyCaliforniaUSA
| | - Davan Murphy
- Center for Neurologic StudyLa JollaCaliforniaUSA
| | | |
Collapse
|
10
|
Hu XM, Song LZX, Zhang ZZ, Ruan X, Li HC, Yu Z, Huang L. Electroacupuncture at ST25 corrected gut microbial dysbiosis and SNpc lipid peroxidation in Parkinson's disease rats. Front Microbiol 2024; 15:1358525. [PMID: 38450172 PMCID: PMC10915097 DOI: 10.3389/fmicb.2024.1358525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/31/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction Parkinson's disease (PD) remains one kind of a complex, progressive neurodegenerative disease. Levodopa and dopamine agonists as widely utilized PD therapeutics have not shown significant positive long-term outcomes. Emerging evidences indicate that electroacupuncture (EA) have potential effects on the therapy of nervous system disorders, particularly PD, but its specific underlying mechanism(s) remains poorly understood, leading to the great challenge of clinical application and management. Previous study has shown that acupuncture ameliorates PD motor symptoms and dopaminergic neuron damage by modulating intestinal dysbiosis, but its intermediate pathway has not been sufficiently investigated. Methods A rat model of PD was induced using rotenone. The therapeutic effect of EA on PD was assessed using the pole and rotarod tests and immunohistostaining for tyrosine hydroxylase (TH) in the substantia nigra (SN) of brain. The role of gut microbiota was explored using 16S rRNA gene sequencing and metabonomic analysis. PICRUSt2 analysis, lipidomic analysis, LPS and inflammatory factor assays were used for subsequent exploration and validation. Correlation analysis was used to identify the key bacteria that EA regulates lipid metabolism to improve PD. Results The present study firstly reappeared the effects of EA on protecting motor function and dopaminergic neurons and modulation of gut microbial dysbiosis in rotenone-induced PD rat model. EA improved motor dysfunction (via the pole and rotarod tests) and protected TH+ neurons in PD rats. EA increased the abundance of beneficial bacteria such as Lactobacillus, Dubosiella and Bifidobacterium and decreased the abundance of Escherichia-Shigella and Morganella belonging to Pseudomonadota, suggesting that the modulation of gut microbiota by EA improving the symptoms of PD motility via alleviating LPS-induced inflammatory response and oxidative stress, which was also validated by various aspects such as microbial gene functional analysis, fecal metabolomics analysis, LPS and inflammatory factor assays and SNpc lipidomics analysis. Moreover, correlation analyses also verified strong correlations of Escherichia-Shigella and Morganella with motor symptoms and SNpc lipid peroxidation, explicating targets and intermediate pathways through which EA improve PD exercise symptom. Conclusion Our results indicate that the improvement of motor function in PD model by EA may be mediated in part by restoring the gut microbiota, which intermediate processes involve circulating endotoxins and inflammatory mediators, SNpc oxidative stress and lipid peroxidation. The gut-microbiome - brain axis may be a potential mechanism of EA treatment for the PD.
Collapse
Affiliation(s)
- Xuan-ming Hu
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li-zhe-xiong Song
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
- School of Acupuncture-Moxibustion, Tuina of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhi-zi Zhang
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
- School of Acupuncture-Moxibustion, Tuina of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xi Ruan
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hai-chang Li
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhi Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lin Huang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
11
|
Zou X, Chen X, Wen Y, Jing X, Luo M, Xin F, Tang Y, Hu M, Liu J, Xu F. Gastric-filling ultrasonography to evaluate gastric motility in patients with Parkinson's disease. Front Neurol 2024; 15:1294260. [PMID: 38410194 PMCID: PMC10895041 DOI: 10.3389/fneur.2024.1294260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/25/2024] [Indexed: 02/28/2024] Open
Abstract
Background Delayed gastric emptying is a common non-motor symptom of Parkinson's disease (PD). However, there is currently no objective evaluation and diagnostic method for this condition. Objectives The purpose of this study was to evaluate the feasibility of gastric-filling ultrasonography for gastric motility in patients with PD and the relationship between gastric dynamics and gastrointestinal symptoms and motor symptoms of PD. Design setting and patients We performed a case-control study with 38 patients with PD and 34 healthy controls. Methods All patients underwent a 120-min ultrasonography examination using a 500-ml semi-liquid test meal. We determined the antral contraction amplitude (ACA), the antrum contraction frequency (ACF), the motility index (MI), and the gastric antral cross-sectional area (CSA). We acquired the CSA at six time points: fasting for 12 h (T0), immediately after drinking the semi-liquid test meal (T1); and at 30 (T30), 60 (T60), 90 (T90), and 120 (T120) min. We calculated the gastric emptying rate (GER) at different time points by using the CSA. We compared the GER between the groups and evaluated the correlation between the GER and gastrointestinal symptoms and motor symptoms of PD. Results The MI and ACF were significantly lower in the PD group compared with the control group (P < 0.05). The GER at T30 and the ACA showed no significant difference between the groups (P > 0.05). At different time points, the GER was significantly different between the PD and control groups (P < 0.001). There was no significant association between the GER and gastrointestinal symptoms; none of them were risk factors for impaired gastric emptying (odds ratio > 1). The GER was negatively correlated with the severity of PD motor symptoms (P < 0.05). Conclusion Patients with PD had significantly delayed gastric emptying, which was negatively correlated with the severity of PD motor symptoms. Measuring gastric emptying by gastric-filling ultrasound had good diagnostic value in clinical screening for delayed gastric motility in patients with PD. Clinical Trial Registration https://www.chictr.org.cn/showproj.html?proj=126304.
Collapse
Affiliation(s)
- Xianwei Zou
- Department of Neurology, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiaqing Chen
- Department of Neurology, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Yanxia Wen
- Department of Neurology, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiaofeng Jing
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
| | - Man Luo
- Department of Neurology, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Fengyue Xin
- Department of Ultrasonography, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Yao Tang
- Department of Neurology, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Mengfei Hu
- Department of Neurology, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Jian Liu
- Department of Ultrasonography, First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Fan Xu
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Espinosa-Oliva AM, Ruiz R, Soto MS, Boza-Serrano A, Rodriguez-Perez AI, Roca-Ceballos MA, García-Revilla J, Santiago M, Serres S, Economopoulus V, Carvajal AE, Vázquez-Carretero MD, García-Miranda P, Klementieva O, Oliva-Martín MJ, Deierborg T, Rivas E, Sibson NR, Labandeira-García JL, Machado A, Peral MJ, Herrera AJ, Venero JL, de Pablos RM. Inflammatory bowel disease induces pathological α-synuclein aggregation in the human gut and brain. Neuropathol Appl Neurobiol 2024; 50:e12962. [PMID: 38343067 DOI: 10.1111/nan.12962] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 12/22/2023] [Accepted: 01/16/2024] [Indexed: 02/15/2024]
Abstract
AIMS According to Braak's hypothesis, it is plausible that Parkinson's disease (PD) originates in the enteric nervous system (ENS) and spreads to the brain through the vagus nerve. In this work, we studied whether inflammatory bowel diseases (IBDs) in humans can progress with the emergence of pathogenic α-synuclein (α-syn) in the gastrointestinal tract and midbrain dopaminergic neurons. METHODS We have analysed the gut and the ventral midbrain from subjects previously diagnosed with IBD and form a DSS-based rat model of gut inflammation in terms of α-syn pathology. RESULTS Our data support the existence of pathogenic α-syn in both the gut and the brain, thus reinforcing the potential role of the ENS as a contributing factor in PD aetiology. Additionally, we have analysed the effect of a DSS-based rat model of gut inflammation to demonstrate (i) the appearance of P-α-syn inclusions in both Auerbach's and Meissner's plexuses (gut), (ii) an increase in α-syn expression in the ventral mesencephalon (brain) and (iii) the degeneration of nigral dopaminergic neurons, which all are considered classical hallmarks in PD. CONCLUSION These results strongly support the plausibility of Braak's hypothesis and emphasise the significance of peripheral inflammation and the gut-brain axis in initiating α-syn aggregation and transport to the substantia nigra, resulting in neurodegeneration.
Collapse
Affiliation(s)
- Ana M Espinosa-Oliva
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Rocío Ruiz
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Manuel Sarmiento Soto
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Churchill Hospital, Oxford, UK
| | - Antonio Boza-Serrano
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Ana I Rodriguez-Perez
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Health Research Institute (IDIS), Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - María A Roca-Ceballos
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Juan García-Revilla
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Marti Santiago
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Sébastien Serres
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Churchill Hospital, Oxford, UK
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Vasiliki Economopoulus
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Churchill Hospital, Oxford, UK
| | - Ana E Carvajal
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| | | | - Pablo García-Miranda
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| | - Oxana Klementieva
- Dementia Research Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - María J Oliva-Martín
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Eloy Rivas
- Departamento de Anatomía Patológica, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Nicola R Sibson
- Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Churchill Hospital, Oxford, UK
| | - José L Labandeira-García
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Health Research Institute (IDIS), Santiago de Compostela, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Alberto Machado
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - María J Peral
- Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| | - Antonio J Herrera
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - José L Venero
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Rocío M de Pablos
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
13
|
Ratan Y, Rajput A, Pareek A, Pareek A, Jain V, Sonia S, Farooqui Z, Kaur R, Singh G. Advancements in Genetic and Biochemical Insights: Unraveling the Etiopathogenesis of Neurodegeneration in Parkinson's Disease. Biomolecules 2024; 14:73. [PMID: 38254673 PMCID: PMC10813470 DOI: 10.3390/biom14010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/15/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative movement disorder worldwide, which is primarily characterized by motor impairments. Even though multiple hypotheses have been proposed over the decades that explain the pathogenesis of PD, presently, there are no cures or promising preventive therapies for PD. This could be attributed to the intricate pathophysiology of PD and the poorly understood molecular mechanism. To address these challenges comprehensively, a thorough disease model is imperative for a nuanced understanding of PD's underlying pathogenic mechanisms. This review offers a detailed analysis of the current state of knowledge regarding the molecular mechanisms underlying the pathogenesis of PD, with a particular emphasis on the roles played by gene-based factors in the disease's development and progression. This study includes an extensive discussion of the proteins and mutations of primary genes that are linked to PD, including α-synuclein, GBA1, LRRK2, VPS35, PINK1, DJ-1, and Parkin. Further, this review explores plausible mechanisms for DAergic neural loss, non-motor and non-dopaminergic pathologies, and the risk factors associated with PD. The present study will encourage the related research fields to understand better and analyze the current status of the biochemical mechanisms of PD, which might contribute to the design and development of efficacious and safe treatment strategies for PD in future endeavors.
Collapse
Affiliation(s)
- Yashumati Ratan
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Aishwarya Rajput
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, Rajasthan, India; (A.R.); (A.P.); (A.P.)
| | - Vivek Jain
- Department of Pharmaceutical Sciences, Mohan Lal Sukhadia University, Udaipur 313001, Rajasthan, India;
| | - Sonia Sonia
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar 143005, Punjab, India;
| | - Zeba Farooqui
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA;
| | - Ranjeet Kaur
- Adesh Institute of Dental Sciences and Research, Bathinda 151101, Punjab, India;
| | - Gurjit Singh
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA;
| |
Collapse
|
14
|
Weber MA, Kerr G, Thangavel R, Conlon MM, Gumusoglu SB, Gupta K, Abdelmotilib HA, Halhouli O, Zhang Q, Geerling JC, Narayanan NS, Aldridge GM. Alpha-Synuclein Pre-Formed Fibrils Injected into Prefrontal Cortex Primarily Spread to Cortical and Subcortical Structures. JOURNAL OF PARKINSON'S DISEASE 2024; 14:81-94. [PMID: 38189765 PMCID: PMC10836574 DOI: 10.3233/jpd-230129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/13/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB) are characterized by diffuse spread of alpha-synuclein (α-syn) throughout the brain. Patients with PDD and DLB have a neuropsychological pattern of deficits that include executive dysfunction, such as abnormalities in planning, timing, working memory, and behavioral flexibility. The prefrontal cortex (PFC) plays a major role in normal executive function and often develops α-syn aggregates in DLB and PDD. OBJECTIVE To investigate the long-term behavioral and cognitive consequences of α-syn pathology in the cortex and characterize pathological spread of α-syn. METHODS We injected human α-syn pre-formed fibrils into the PFC of wild-type male mice. We then assessed the behavioral and cognitive effects between 12- and 21-months post-injection and characterized the spread of pathological α-syn in cortical, subcortical, and brainstem regions. RESULTS We report that PFC PFFs: 1) induced α-syn aggregation in multiple cortical and subcortical regions with sparse aggregation in midbrain and brainstem nuclei; 2) did not affect interval timing or spatial learning acquisition but did mildly alter behavioral flexibility as measured by intraday reversal learning; and 3) increased open field exploration. CONCLUSIONS This model of cortical-dominant pathology aids in our understanding of how local α-syn aggregation might impact some symptoms in PDD and DLB.
Collapse
Affiliation(s)
- Matthew A. Weber
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Gemma Kerr
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ramasamy Thangavel
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mackenzie M. Conlon
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Serena B. Gumusoglu
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Kalpana Gupta
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Hisham A. Abdelmotilib
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Oday Halhouli
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Qiang Zhang
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Joel C. Geerling
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Nandakumar S. Narayanan
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| | - Georgina M. Aldridge
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
15
|
Sampson T. Circulating Extracellular Vesicles as a Potential Mediator of Synuclein Pathology in the Gut. Mov Disord 2024; 39:3-5. [PMID: 38294044 PMCID: PMC10832292 DOI: 10.1002/mds.29673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 02/01/2024] Open
Abstract
The spread of pathological alpha-synuclein (aSyn) from the gastrointestinal tract to the brain, or the brain to the intestine, is of increasing interest given observations in experimental models. While there has been a focus on neuronal-mediated spread and propagation, non-neuronal reservoirs of aSyn are also present. In this issue, Yang et al describe the ability of extracellular vesicles, derived from red blood cells in circulation to mediate the deposition of aSyn into the GI tract. These vesicles may therefore represent a potential non-neuronal pathway by which aSyn may spread across body sites in the initiation and progression of Parkinson’s disease and other synucleinopathies.
Collapse
Affiliation(s)
- Timothy Sampson
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
16
|
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: 2] [Impact Index Per Article: 2.0] [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
|
17
|
Yamakado H, Takahashi R. Experimental Animal Models of Prodromal Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:S369-S379. [PMID: 38427504 PMCID: PMC11492006 DOI: 10.3233/jpd-230393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 03/03/2024]
Abstract
There is an estimated 35-45% loss of striatal dopamine at the time of diagnosis of Parkinson's disease (PD), and cases clinically diagnosed in the early stages may already be pathologically in advanced stages. Recent large-scale clinical trials of disease-modifying therapies (DMT) also suggest the necessity of targeting patients at earlier stages of the disease. From this perspective, the prodromal phase of PD is currently the focus of attention, emphasizing the need for a prodromal mouse model that accurately reflects the pathophysiology, along with early biomarkers. To establish prodromal animal model of PD with high face validity that reflects the disease state, the model must possess high construct validity that accurately incorporates clinical and pathological features in the prodromal phase. Furthermore, as a preclinical model of DMT, the model must possess high predictive validity to accurately evaluate the response to intervention. This review provides an overview of animal models which reflect the characteristics of prodromal PD, including alpha-synuclein (aS) accumulation and associated early non-motor symptoms, with a focus on the aS propagation model and genetic model. In addition, we discuss the challenges associated with these models. The genetic model often fails to induce motor symptoms, while aS propagation models skip the crucial step of initial aS aggregate formation, thereby not fully replicating the entire natural course of the disease. Identifying factors that induce the transition from prodromal to symptomatic phase is important as a preclinical model for DMT to prevent or delay the onset of the disease.
Collapse
Affiliation(s)
- Hodaka Yamakado
- Department of Therapeutics for Multiple System Atrophy, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
18
|
Song LZX, Xu N, Yu Z, Yang H, Xu CC, Qiu Z, Dai JW, Xu B, Hu XM. The effect of electroacupuncture at ST25 on Parkinson's disease constipation through regulation of autophagy in the enteric nervous system. Anat Rec (Hoboken) 2023; 306:3214-3228. [PMID: 36655864 DOI: 10.1002/ar.25148] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 11/22/2022] [Accepted: 11/29/2022] [Indexed: 01/20/2023]
Abstract
The effectiveness and safety of electroacupuncture (EA) for constipation have been confirmed by numerous clinical studies and experiments, and there are also studies on the efficacy of EA for Parkinson's disease (PD) motor symptoms. However, there are few researches on EA for PD constipation. Autophagy is thought to be involved in the mechanistic process of EA in the central nervous system (CNS) intervention in Parkinson's pathology. However, whether it has the same effect on the enteric nervous system (ENS) has not been elucidated. Therefore, we investigated whether EA at Tianshu (ST25) acupoint promotes the clearance of α-Syn and damaged mitochondria aggregated in the ENS in a model of rotenone-induced PD constipation. This study evaluated constipation symptoms by stool characteristics, excretion volume, and water content, and the expression levels of colonic ATG5, LC3II, and Parkin were detected by Western Blot (WB) and Real-Time Quantitative PCR (RT-qPCR). The relationship between the location of α-Syn and Parkin in the colonic ENS was observed by immunofluorescence (IF). The results showed that EA intervention significantly relieved the symptoms of rotenone-induced constipation in PD rats, reversed the rotenone-induced down-regulation of colonic ATG5, LC3II, and Parkin expression, and the positional relationship between colonic α-Syn and Parkin proved to be highly correlated. It is suggested that EA might be helpful in treating PD constipation by modulating Parkin-induced mitochondrial autophagy.
Collapse
Affiliation(s)
- Li-Zhe-Xiong Song
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Na Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhi Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hui Yang
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Cheng-Cheng Xu
- Nanjing Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, China
| | - Zi Qiu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing-Wen Dai
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bin Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuan-Ming Hu
- Nanjing Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
19
|
Kleine Bardenhorst S, Cereda E, Severgnini M, Barichella M, Pezzoli G, Keshavarzian A, Desideri A, Pietrucci D, Aho VTE, Scheperjans F, Hildebrand F, Weis S, Egert M, Karch A, Vital M, Rübsamen N. Gut microbiota dysbiosis in Parkinson disease: A systematic review and pooled analysis. Eur J Neurol 2023; 30:3581-3594. [PMID: 36593694 DOI: 10.1111/ene.15671] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 11/04/2022] [Accepted: 12/07/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND PURPOSE The role of the gut microbiome in the pathogenesis of Parkinson disease (PD) is under intense investigation, and the results presented are still very heterogeneous. These discrepancies arise not only from the highly heterogeneous pathology of PD, but also from widely varying methodologies at all stages of the workflow, from sampling to final statistical analysis. The aim of the present work is to harmonize the workflow across studies to reduce the methodological heterogeneity and to perform a pooled analysis to account for other sources of heterogeneity. METHODS We performed a systematic review to identify studies comparing the gut microbiota of PD patients to healthy controls. A workflow was designed to harmonize processing across all studies from bioinformatics processing to final statistical analysis using a Bayesian random-effects meta-analysis based on individual patient-level data. RESULTS The results show that harmonizing workflows minimizes differences between statistical methods and reveals only a small set of taxa being associated with the pathogenesis of PD. Increased shares of the genera Akkermansia and Bifidobacterium and decreased shares of the genera Roseburia and Faecalibacterium were most characteristic for PD-associated microbiota. CONCLUSIONS Our study summarizes evidence that reduced levels of butyrate-producing taxa in combination with possible degradation of the mucus layer by Akkermansia may promote intestinal inflammation and reduced permeability of the gut mucosal layer. This may allow potentially pathogenic metabolites to transit and enter the enteric nervous system.
Collapse
Affiliation(s)
| | - Emanuele Cereda
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marco Severgnini
- Institute of Biomedical Technologies, Italian National Research Council, Milan, Italy
| | | | - Gianni Pezzoli
- Parkinson Institute, ASST-Pini-CTO, Milan, Italy
- Fondazione Grigioni per il Morbo di Parkinson, Milan, Italy
| | - Ali Keshavarzian
- Rush Center for Integrated Microbiome & Chronobiology Research, Chicago, Illinois, USA
- Departments of Medicine, Physiology, Anatomy, and Cell Biology, Rush University, Chicago, Illinois, USA
| | | | - Daniele Pietrucci
- Department for Innovation in Biological, Agro-food, and Forest Systems, University of Tuscia, Viterbo, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, IBIOM, Italian National Research Council, Bari, Italy
| | - Velma T E Aho
- DNA Sequencing and Genomics Laboratory, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
- Department of Neurology, Helsinki University Hospital, and Clinicum, University of Helsinki, Helsinki, Finland
| | - Filip Scheperjans
- Department of Neurology, Helsinki University Hospital, and Clinicum, University of Helsinki, Helsinki, Finland
| | | | - Severin Weis
- Microbiology and Hygiene Group, Institute of Precision Medicine, Furtwangen University, Furtwangen, Germany
| | - Markus Egert
- Microbiology and Hygiene Group, Institute of Precision Medicine, Furtwangen University, Furtwangen, Germany
| | - André Karch
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Marius Vital
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Nicole Rübsamen
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| |
Collapse
|
20
|
Muhammad B, Li H, Gu Y, Xue S, Gao Y, Xu Z, Fang X, Ding H, Wu F, Geng D, Niu H. IL-1β/IL-1R1 signaling is involved in the propagation of α-synuclein pathology of the gastrointestinal tract to the brain. J Neurochem 2023; 166:830-846. [PMID: 37434423 DOI: 10.1111/jnc.15886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/03/2023] [Accepted: 06/07/2023] [Indexed: 07/13/2023]
Abstract
The pathological hallmark of Parkinson's disease (PD) is the intraneuronal accumulation of misfolded alpha-synuclein (termed Lewy bodies) in dopaminergic neurons of substantia nigra par compacta (SNc). It is assumed that the α-syn pathology is induced by gastrointestinal inflammation and then transfers to the brain by the gut-brain axis. Therefore, the relationship between gastrointestinal inflammation and α-syn pathology leading to PD remains to be investigated. In our study, rotenone (ROT) oral administration induces gastrointestinal tract (GIT) inflammation in mice. In addition, we used pseudorabies virus (PRV) for tracing studies and performed behavioral testing. We observed that ROT treatments enhance macrophage activation, inflammatory mediator expression, and α-syn pathology in the GIT 6-week post-treatment (P6). Moreover, pathological α-syn was localized with IL-1R1 positive neural cells in GIT. In line with these findings, we also find pS129-α-syn signals in the dorsal motor nucleus of the vagus (DMV) and tyrosine hydroxylase in the nigral-striatum dynamically change from 3-week post-treatment (P3) to P6. Following that, pS129-α-syn was dominant in the enteric neural cell, DMV, and SNc, accompanied by microglial activation, and these phenotypes were absent in IL-1R1r/r mice. These data suggest that IL-1β/IL-1R1-dependent inflammation of GIT can induce α-syn pathology, which then propagates to the DMV and SNc, resulting in PD.
Collapse
Affiliation(s)
- Bilal Muhammad
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Haiying Li
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Yunlu Gu
- Department of Neuroscience, Xuzhou Medical University, Xuzhou, China
| | - Senlin Xue
- Department of Post-Graduation, Xuzhou Medical University, Xuzhou, China
| | - Yao Gao
- Department of Post-Graduation, Xuzhou Medical University, Xuzhou, China
| | - Zhou Xu
- Department of Post-Graduation, Xuzhou Medical University, Xuzhou, China
| | - Xiaoli Fang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Haohan Ding
- Department of Neuroscience, Xuzhou Medical University, Xuzhou, China
| | - Fang Wu
- Department of Neuroscience, Xuzhou Medical University, Xuzhou, China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Haichen Niu
- Department of Genetics, Xuzhou Medical University, Xuzhou, China
- Public Experimental Research Center of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
21
|
Liu Z, Lemus J, Smirnova IV, Liu W. Rehabilitation for non-motor symptoms for patients with Parkinson's disease from an α-synuclein perspective: a narrative review. EXPLORATION OF NEUROPROTECTIVE THERAPY 2023; 3:235-257. [PMID: 37920444 PMCID: PMC10621781 DOI: 10.37349/ent.2023.00049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/22/2023] [Indexed: 11/04/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder affecting aged population around the world. PD is characterized by neuronal Lewy bodies present in the substantia nigra of the midbrain and the loss of dopaminergic neurons with various motor and non-motor symptoms associated with the disease. The protein α-synuclein has been extensively studied for its contribution to PD pathology, as α-synuclein aggregates form the major component of Lewy bodies, a hallmark of PD. In this narrative review, the authors first focus on a brief explanation of α-synuclein aggregation and circumstances under which aggregation can occur, then present a hypothesis for PD pathogenesis in the peripheral nervous system (PNS) and how PD can spread to the central nervous system from the PNS via the transport of α-synuclein aggregates. This article presents arguments both for and against this hypothesis. It also presents various non-pharmacological rehabilitation approaches and management techniques for both motor and non-motor symptoms of PD and the related pathology. This review seeks to examine a possible hypothesis of PD pathogenesis and points to a new research direction focus on rehabilitation therapy for patients with PD. As various non-motor symptoms of PD appear to occur earlier than motor symptoms, more focus on the treatment of non-motor symptoms as well as a better understanding of the biochemical mechanisms behind those non-motor symptoms may lead to better long-term outcomes for patients with PD.
Collapse
Affiliation(s)
- Zhaoyang Liu
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Orthopedic Surgery and Sports Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jessica Lemus
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Irina V. Smirnova
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Wen Liu
- Department of Physical Therapy, Rehabilitation Science, and Athletic Training, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
22
|
Schmitt V, Masanetz RK, Weidenfeller M, Ebbinghaus LS, Süß P, Rosshart SP, von Hörsten S, Zunke F, Winkler J, Xiang W. Gut-to-brain spreading of pathology in synucleinopathies: A focus on molecular signalling mediators. Behav Brain Res 2023; 452:114574. [PMID: 37423320 DOI: 10.1016/j.bbr.2023.114574] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
Synucleinopathies are a group of neurodegenerative disorders, classically characterized by the accumulation of aggregated alpha synuclein (aSyn) in the central nervous system. Parkinson's disease (PD) and multiple system atrophy (MSA) are the two prominent members of this family. Current treatment options mainly focus on the motor symptoms of these diseases. However, non-motor symptoms, including gastrointestinal (GI) symptoms, have recently gained particular attention, as they are frequently associated with synucleinopathies and often arise before motor symptoms. The gut-origin hypothesis has been proposed based on evidence of an ascending spreading pattern of aggregated aSyn from the gut to the brain, as well as the comorbidity of inflammatory bowel disease and synucleinopathies. Recent advances have shed light on the mechanisms underlying the progression of synucleinopathies along the gut-brain axis. Given the rapidly expanding pace of research in the field, this review presents a summary of the latest findings on the gut-to-brain spreading of pathology and potential pathology-reinforcing mediators in synucleinopathies. Here, we focus on 1) gut-to-brain communication pathways, including neuronal pathways and blood circulation, and 2) potential molecular signalling mediators, including bacterial amyloid proteins, microbiota dysbiosis-induced alterations in gut metabolites, as well as host-derived effectors, including gut-derived peptides and hormones. We highlight the clinical relevance and implications of these molecular mediators and their possible mechanisms in synucleinopathies. Moreover, we discuss their potential as diagnostic markers in distinguishing the subtypes of synucleinopathies and other neurodegenerative diseases, as well as for developing novel individualized therapeutic options for synucleinopathies.
Collapse
Affiliation(s)
- Verena Schmitt
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Rebecca Katharina Masanetz
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Martin Weidenfeller
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Lara Savannah Ebbinghaus
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Patrick Süß
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Stephan P Rosshart
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Stephan von Hörsten
- Department for Experimental Therapy, University Hospital Erlangen, Preclinical Experimental Center, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany
| | - Wei Xiang
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Germany.
| |
Collapse
|
23
|
Videlock EJ, Xing T, Yehya AHS, Travagli RA. Experimental models of gut-first Parkinson's disease: A systematic review. Neurogastroenterol Motil 2023; 35:e14604. [PMID: 37125607 PMCID: PMC10524037 DOI: 10.1111/nmo.14604] [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: 12/02/2022] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND There is strong support from studies in humans and in animal models that Parkinson's disease (PD) may begin in the gut. This brings about a unique opportunity for researchers in the field of neurogastroenterology to contribute to advancing the field and making contributions that could lead to the ability to diagnose and treat PD in the premotor stages. Lack of familiarity with some of the aspects of the experimental approaches used in these studies may present a barrier for neurogastroenterology researchers to enter the field. Much remains to be understood about intestinal-specific components of gut-first PD pathogenesis and the field would benefit from contributions of enteric and central nervous system neuroscientists. PURPOSE To address these issues, we have conducted a systematic review of the two most frequently used experimental models of gut-first PD: transneuronal propagation of α-synuclein preformed fibrils and oral exposure to environmental toxins. We have reviewed the details of these studies and present methodological considerations for the use of these models. Our aim is that this review will serve as a framework and useful reference for neuroscientists, gastroenterologists, and neurologists interested in applying their expertise to advancing our understanding of gut-first PD.
Collapse
Affiliation(s)
- Elizabeth J. Videlock
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Tiaosi Xing
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, USA
| | - Ashwaq Hamid Salem Yehya
- Center for Inflammatory Bowel Diseases, Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | | |
Collapse
|
24
|
Rocha E, Chamoli M, Chinta SJ, Andersen JK, Wallis R, Bezard E, Goldberg M, Greenamyre T, Hirst W, Kuan WL, Kirik D, Niedernhofer L, Rappley I, Padmanabhan S, Trudeau LE, Spillantini M, Scott S, Studer L, Bellantuono I, Mortiboys H. Aging, Parkinson's Disease, and Models: What Are the Challenges? AGING BIOLOGY 2023; 1:e20230010. [PMID: 38978807 PMCID: PMC11230631 DOI: 10.59368/agingbio.20230010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Parkinson's disease (PD) is a chronic, neurodegenerative condition characterized by motor symptoms such as bradykinesia, rigidity, and tremor, alongside multiple nonmotor symptoms. The appearance of motor symptoms is linked to progressive dopaminergic neuron loss within the substantia nigra. PD incidence increases sharply with age, suggesting a strong association between mechanisms driving biological aging and the development and progression of PD. However, the role of aging in the pathogenesis of PD remains understudied. Numerous models of PD, including cell models, toxin-induced models, and genetic models in rodents and nonhuman primates (NHPs), reproduce different aspects of PD, but preclinical studies of PD rarely incorporate age as a factor. Studies using patient neurons derived from stem cells via reprogramming methods retain some aging features, but their characterization, particularly of aging markers and reproducibility of neuron type, is suboptimal. Investigation of age-related changes in PD using animal models indicates an association, but this is likely in conjunction with other disease drivers. The biggest barrier to drawing firm conclusions is that each model lacks full characterization and appropriate time-course assessments. There is a need to systematically investigate whether aging increases the susceptibility of mouse, rat, and NHP models to develop PD and understand the role of cell models. We propose that a significant investment in time and resources, together with the coordination and sharing of resources, knowledge, and data, is required to accelerate progress in understanding the role of biological aging in PD development and improve the reliability of models to test interventions.
Collapse
Affiliation(s)
- Emily Rocha
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Shankar J Chinta
- Buck Institute for Research on Aging, Novato, CA, USA
- Touro University California, College of Pharmacy, Vallejo, CA, USA
| | | | - Ruby Wallis
- The Healthy Lifespan Institute, Sheffield, United Kingdom
| | | | | | - Tim Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases and Department of Neurology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - We-Li Kuan
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (BRAINS), Lund, Sweden
| | - Laura Niedernhofer
- Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Irit Rappley
- Recursion pharmaceuticals, Salt Lake City, UT, USA
| | | | - Louis-Eric Trudeau
- Department of pharmacology and physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Maria Spillantini
- Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | | | - Lorenz Studer
- The Center for Stem Cell Biology and Developmental Biology Program, Sloan-Kettering Institute for Cancer Research, New York, NY, USA
| | - Ilaria Bellantuono
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Oncology and Metabolism, The Medical School, Sheffield, United Kingdom
| | - Heather Mortiboys
- The Healthy Lifespan Institute, Sheffield, United Kingdom
- Department of Neuroscience, Sheffield Institute of Translational Neuroscience (SITraN), University of Sheffield, Sheffield, United Kindgom
| |
Collapse
|
25
|
Carceles-Cordon M, Weintraub D, Chen-Plotkin AS. Cognitive heterogeneity in Parkinson's disease: A mechanistic view. Neuron 2023; 111:1531-1546. [PMID: 37028431 PMCID: PMC10198897 DOI: 10.1016/j.neuron.2023.03.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/22/2022] [Accepted: 03/13/2023] [Indexed: 04/09/2023]
Abstract
Cognitive impairment occurs in most individuals with Parkinson's disease (PD), exacting a high toll on patients, their caregivers, and the healthcare system. In this review, we begin by summarizing the current clinical landscape surrounding cognition in PD. We then discuss how cognitive impairment and dementia may develop in PD based on the spread of the pathological protein alpha-synuclein (aSyn) from neurons in brainstem regions to those in the cortical regions of the brain responsible for higher cognitive functions, as first proposed in the Braak hypothesis. We appraise the Braak hypothesis from molecular (conformations of aSyn), cell biological (cell-to-cell spread of pathological aSyn), and organ-level (region-to-region spread of aSyn pathology at the whole brain level) viewpoints. Finally, we argue that individual host factors may be the most poorly understood aspect of this pathological process, accounting for substantial heterogeneity in the pattern and pace of cognitive decline in PD.
Collapse
Affiliation(s)
- Marc Carceles-Cordon
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dan Weintraub
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
26
|
Weber MA, Kerr G, Thangavel R, Conlon MM, Abdelmotilib HA, Halhouli O, Zhang Q, Geerling JC, Narayanan NS, Aldridge GM. Alpha-synuclein pre-formed fibrils injected into prefrontal cortex primarily spread to cortical and subcortical structures and lead to isolated behavioral symptoms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.526365. [PMID: 36778400 PMCID: PMC9915664 DOI: 10.1101/2023.01.31.526365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Parkinson's disease dementia (PDD) and dementia with Lewy bodies (DLB) are characterized by diffuse spread of alpha-synuclein (α-syn) throughout the brain. Patients with PDD and DLB have a neuropsychological pattern of deficits that include executive dysfunction, such as abnormalities in planning, timing, working memory, and behavioral flexibility. The prefrontal cortex (PFC) plays a major role in normal executive function and often develops α-syn aggregates in DLB and PDD. To investigate the consequences of α-syn pathology in the cortex, we injected human α-syn pre-formed fibrils into the PFC of wildtype mice. We report that PFC PFFs: 1) induced α-syn aggregation in multiple cortical and subcortical regions with sparse aggregation in midbrain and brainstem nuclei; 2) did not affect interval timing or spatial learning acquisition but did mildly alter behavioral flexibility as measured by intraday reversal learning; 3) increased open field exploration; and 4) did not affect susceptibility to an inflammatory challenge. This model of cortical-dominant pathology aids in our understanding of how local α-syn aggregation might impact some symptoms in PDD and DLB.
Collapse
Affiliation(s)
- Matthew A. Weber
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| | - Gemma Kerr
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| | - Ramasamy Thangavel
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| | - Mackenzie M. Conlon
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City
| | | | - Oday Halhouli
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| | - Qiang Zhang
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| | - Joel C. Geerling
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| | | | - Georgina M. Aldridge
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City
| |
Collapse
|
27
|
Chen M, Mor DE. Gut-to-Brain α-Synuclein Transmission in Parkinson's Disease: Evidence for Prion-like Mechanisms. Int J Mol Sci 2023; 24:ijms24087205. [PMID: 37108366 PMCID: PMC10139032 DOI: 10.3390/ijms24087205] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Parkinson's disease (PD) is a multifactorial disorder involving both motor and non-motor symptoms caused by the progressive death of distinct neuronal populations, including dopaminergic neurons in the substantia nigra. The deposition of aggregated α-synuclein protein into Lewy body inclusions is a hallmark of the disorder, and α-synuclein pathology has been found in the enteric nervous system (ENS) of PD patients up to two decades prior to diagnosis. In combination with the high occurrence of gastrointestinal dysfunction in early stages of PD, current evidence strongly suggests that some forms of PD may originate in the gut. In this review, we discuss human studies that support ENS Lewy pathology as a characteristic feature of PD, and present evidence from humans and animal model systems that α-synuclein aggregation may follow a prion-like spreading cascade from enteric neurons, through the vagal nerve, and into the brain. Given the accessibility of the human gut to pharmacologic and dietary interventions, therapeutic strategies aimed at reducing pathological α-synuclein in the gastrointestinal tract hold significant promise for PD treatment.
Collapse
Affiliation(s)
- Merry Chen
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Danielle E Mor
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
28
|
Bang Y, Kwon Y, Kim M, Moon SH, Jung K, Choi HJ. Ursolic acid enhances autophagic clearance and ameliorates motor and non-motor symptoms in Parkinson's disease mice model. Acta Pharmacol Sin 2023; 44:752-765. [PMID: 36138143 PMCID: PMC10042858 DOI: 10.1038/s41401-022-00988-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/22/2022] [Indexed: 11/08/2022] Open
Abstract
Protein aggregation and the abnormal accumulation of aggregates are considered as common mechanisms of neurodegeneration such as Parkinson's disease (PD). Ursolic acid (UA), a natural pentacyclic triterpenoid compound, has shown a protective activity in several experimental models of brain dysfunction through inhibiting oxidative stress and inflammatory responses and suppressing apoptotic signaling in the brain. In this study, we investigated whether UA promoted autophagic clearance of protein aggregates and attenuated the pathology and characteristic symptoms in PD mouse model. Mice were injected with rotenone (1 mg · kg-1 · d-1, i.p.) five times per week for 1 or 2 weeks. We showed that rotenone injection induced significant motor deficit and prodromal non-motor symptoms accompanied by a significant dopaminergic neuronal loss and the deposition of aggregated proteins such as p62 and ubiquitin in the substantia nigra and striatum. Co-injection of UA (10 mg · kg-1 · d-1, i.p.) ameliorated all the rotenone-induced pathological alterations. In differentiated human neuroblastoma SH-SY5Y cells, two-step treatment with a proteasome inhibitor MG132 (0.25, 2.5 μM) induced marked accumulation of ubiquitin and p62 with clear and larger aggresome formation, while UA (5 μM) significantly attenuated the MG132-induced protein accumulation. Furthermore, we demonstrated that UA (5 μM) significantly increased autophagic clearance by promoting autophagic flux in primary neuronal cells and SH-SY5Y cells; UA affected autophagy regulation by increasing the phosphorylation of JNK, which triggered the dissociation of Bcl-2 from Beclin 1. These results suggest that UA could be a promising therapeutic candidate for reducing PD progression from the prodromal stage by regulating abnormal protein accumulation in the brain.
Collapse
Affiliation(s)
- Yeojin Bang
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, South Korea
| | - Yoonjung Kwon
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, South Korea
| | - Mihyang Kim
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, South Korea
| | - Soung Hee Moon
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, South Korea
| | - Kiwon Jung
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, South Korea
| | - Hyun Jin Choi
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon, Gyeonggi-do, 11160, South Korea.
| |
Collapse
|
29
|
Forloni G. Alpha Synuclein: Neurodegeneration and Inflammation. Int J Mol Sci 2023; 24:ijms24065914. [PMID: 36982988 PMCID: PMC10059798 DOI: 10.3390/ijms24065914] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Alpha-Synuclein (α-Syn) is one of the most important molecules involved in the pathogenesis of Parkinson's disease and related disorders, synucleinopathies, but also in several other neurodegenerative disorders with a more elusive role. This review analyzes the activities of α-Syn, in different conformational states, monomeric, oligomeric and fibrils, in relation to neuronal dysfunction. The neuronal damage induced by α-Syn in various conformers will be analyzed in relation to its capacity to spread the intracellular aggregation seeds with a prion-like mechanism. In view of the prominent role of inflammation in virtually all neurodegenerative disorders, the activity of α-Syn will also be illustrated considering its influence on glial reactivity. We and others have described the interaction between general inflammation and cerebral dysfunctional activity of α-Syn. Differences in microglia and astrocyte activation have also been observed when in vivo the presence of α-Syn oligomers has been combined with a lasting peripheral inflammatory effect. The reactivity of microglia was amplified, while astrocytes were damaged by the double stimulus, opening new perspectives for the control of inflammation in synucleinopathies. Starting from our studies in experimental models, we extended the perspective to find useful pointers to orient future research and potential therapeutic strategies in neurodegenerative disorders.
Collapse
Affiliation(s)
- Gianluigi Forloni
- Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy
| |
Collapse
|
30
|
Abstract
Abnormalities in gut microbiota have been suggested to be involved in the pathophysiology and progression of Parkinson's disease (PD). Gastrointestinal nonmotor symptoms often precede the onset of motor features in PD, suggesting a role for gut dysbiosis in neuroinflammation and α-synuclein (α-syn) aggregation. In the first part of this chapter, we analyze critical features of healthy gut microbiota and factors (environmental and genetic) that modify its composition. In the second part, we focus on the mechanisms underlying the gut dysbiosis and how it alters anatomically and functionally the mucosal barrier, triggering neuroinflammation and subsequently α-syn aggregation. In the third part, we describe the most common alterations in the gut microbiota of PD patients, dividing the gastrointestinal system in higher and lower tract to examine the association between microbiota abnormalities and clinical features. In the final section, we report on current and future therapeutic approaches to gut dysbiosis aiming to either reduce the risk for PD, modify the disease course, or improve the pharmacokinetic profile of dopaminergic therapies. We also suggest that further studies will be needed to clarify the role of the microbiome in PD subtyping and of pharmacological and nonpharmacological interventions in modifying specific microbiota profiles in individualizing disease-modifying treatments in PD.
Collapse
Affiliation(s)
- Salvatore Bonvegna
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Milan, Italy
| | - Roberto Cilia
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Department of Clinical Neurosciences, Parkinson and Movement Disorders Unit, Milan, Italy.
| |
Collapse
|
31
|
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
|
32
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
33
|
Hong D, Zhang C, Wu W, Lu X, Zhang L. Modulation of the gut-brain axis via the gut microbiota: a new era in treatment of amyotrophic lateral sclerosis. Front Neurol 2023; 14:1133546. [PMID: 37153665 PMCID: PMC10157060 DOI: 10.3389/fneur.2023.1133546] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/17/2023] [Indexed: 05/10/2023] Open
Abstract
There are trillions of different microorganisms in the human digestive system. These gut microbes are involved in the digestion of food and its conversion into the nutrients required by the body. In addition, the gut microbiota communicates with other parts of the body to maintain overall health. The connection between the gut microbiota and the brain is known as the gut-brain axis (GBA), and involves connections via the central nervous system (CNS), the enteric nervous system (ENS), and endocrine and immune pathways. The gut microbiota regulates the central nervous system bottom-up through the GBA, which has prompted researchers to pay considerable attention to the potential pathways by which the gut microbiota might play a role in the prevention and treatment of amyotrophic lateral sclerosis (ALS). Studies with animal models of ALS have shown that dysregulation of the gut ecology leads to dysregulation of brain-gut signaling. This, in turn, induces changes in the intestinal barrier, endotoxemia, and systemic inflammation, which contribute to the development of ALS. Through the use of antibiotics, probiotic supplementation, phage therapy, and other methods of inducing changes in the intestinal microbiota that can inhibit inflammation and delay neuronal degeneration, the clinical symptoms of ALS can be alleviated, and the progression of the disease can be delayed. Therefore, the gut microbiota may be a key target for effective management and treatment of ALS.
Collapse
Affiliation(s)
- Du Hong
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Chi Zhang
- Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Wenshuo Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaohui Lu
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Liping Zhang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Liping Zhang
| |
Collapse
|
34
|
Gut-oriented disease modifying therapy for Parkinson's disease. J Formos Med Assoc 2023; 122:9-18. [PMID: 36182642 DOI: 10.1016/j.jfma.2022.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 01/10/2023]
Abstract
Neuropathology studies have shown that the pathognomonic feature of Parkinson's disease (PD), one of the most common neurodegenerative disorders, may start from the gut enteric nervous system and then spread to the central dopaminergic neurons through the gut-brain axis. With the advent of metagenomic sequencing and metabolomic analysis, a plethora of evidence has revealed different gut microbiomes and gut metabolites in patients with PD compared with unaffected controls. Currently, although dopaminergic treatments and deep brain stimulation can provide some symptomatic benefits for motor symptoms of the disease, their long-term use is problematic. A mechanism-targeted therapy to halt the neurodegeneration is lacking. The recently observed gut microenvironmental changes in the early stages of the disease play a vital role in the PD pathogenesis. Patients whose disease begins in the gut may benefit most from interventions that target the gut microenvironments. In this review, we will summarize the current studies demonstrating multifunctional roles of gut microbiota in the gut-brain axis of PD and the currently available evidence for targeting the gut microbiota as a novel approach to potential disease-modifying therapy in PD.
Collapse
|
35
|
Aggregation of alpha-synuclein in enteric neurons does not impact function in vitro. Sci Rep 2022; 12:22211. [PMID: 36564445 PMCID: PMC9789045 DOI: 10.1038/s41598-022-26543-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Recent evidence implicates a gut-first pathogenesis in the enteric nervous system (ENS) within a portion of PD patients, yet in vitro investigations have primarily focused on the central nervous system. Here, the preformed fibril (PFF) PD model is applied with co-administered groups of butyrate and lipopolysaccharide to model the effects of the local gut microbiome. Significant PFF uptake and retention occur in isolated rat enteric neurons compared to untreated controls resulting in increasing immunostained aggregate conformation-specific, alpha-synuclein (a-Syn) average intensity between 6 µg PFF and untreated controls. Cortical neurons significantly retain PFFs with an increase in aggregated a-Syn average intensity within all dosages. Differences in growth cone morphology but not dynamics in PFF-treated ENS cultures occur. Electrophysiological recordings via a microelectrode array (MEA) indicate no overall difference in spontaneous spike rate. However, only untreated controls respond to PD-relevant dopamine stimulus, while 1 µg PFF and control populations respond to stimulus with ENS-abundant acetylcholine. Finally, no differences in substance P levels-correlated with PD and neurodegeneration-are observed. Overall, these findings suggest the ENS retains PFF dosage absent acute loss in function, however, does experience changes in growth cone morphology and dopamine-stimulated activity.
Collapse
|
36
|
Salim S, Ahmad F, Banu A, Mohammad F. Gut microbiome and Parkinson's disease: Perspective on pathogenesis and treatment. J Adv Res 2022:S2090-1232(22)00242-9. [PMID: 36332796 PMCID: PMC10403695 DOI: 10.1016/j.jare.2022.10.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/26/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a disease of ⍺-synuclein aggregation-mediated dopaminergic neuronal loss in the substantia nigra pars compacta, which leads to motor and non-motor symptoms. Through the last two decades of research, there has been growing consensus that inflammation-mediated oxidative stress, mitochondrial dysfunction, and cytokine-induced toxicity are mainly involved in neuronal damage and loss associated with PD. However, it remains unclear how these mechanisms relate to sporadic PD, a more common form of PD. Both enteric and central nervous systems have been implicated in the pathogenesis of sporadic PD, thus highlighting the crosstalk between the gut and brain. AIM of Review: In this review, we summarize how alterations in the gut microbiome can affect PD pathogenesis. We highlight various mechanisms increasing/decreasing the risk of PD development. Based on the previous supporting evidence, we suggest how early interventions could protect against PD development and how controlling specific factors, including our diet, could modify our perspective on disease mechanisms and therapeutics. We explain the strong relationship between the gut microbiota and the brain in PD subjects, by delineating the multiple mechanisms involved inneuroinflammation and oxidative stress. We conclude that the neurodetrimental effects of western diet (WD) and the neuroprotective effects of Mediterranean diets should be further exploredin humans through clinical trials. Key Scientific Concepts of Review: Alterations in the gut microbiome and associated metabolites may contribute to pathogenesis in PD. In some studies, probiotics have been shown to exert anti-oxidative effects in PD via improved mitochondrial dynamics and homeostasis, thus reducing PD-related consequences. However, there is a significant unmet need for randomized clinical trials to investigate the effectiveness of microbial products, probiotic-based supplementation, and dietary intervention in reversing gut microbial dysbiosis in PD.
Collapse
|
37
|
Niederberger E, Wilken-Schmitz A, Manderscheid C, Schreiber Y, Gurke R, Tegeder I. Non-Reproducibility of Oral Rotenone as a Model for Parkinson's Disease in Mice. Int J Mol Sci 2022; 23:ijms232012658. [PMID: 36293513 PMCID: PMC9604506 DOI: 10.3390/ijms232012658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Oral rotenone has been proposed as a model for Parkinson’s disease (PD) in mice. To establish the model in our lab and study complex behavior we followed a published treatment regimen. C57BL/6 mice received 30 mg/kg body weight of rotenone once daily via oral administration for 4 and 8 weeks. Motor functions were assessed by RotaRod running. Immunofluorescence studies were used to analyze the morphology of dopaminergic neurons, the expression of alpha-Synuclein (α-Syn), and inflammatory gliosis or infiltration in the substantia nigra. Rotenone-treated mice did not gain body weight during treatment compared with about 4 g in vehicle-treated mice, which was however the only robust manifestation of drug treatment and suggested local gut damage. Rotenone-treated mice had no deficits in motor behavior, no loss or sign of degeneration of dopaminergic neurons, no α-Syn accumulation, and only mild microgliosis, the latter likely an indirect remote effect of rotenone-evoked gut dysbiosis. Searching for explanations for the model failure, we analyzed rotenone plasma concentrations via LC-MS/MS 2 h after administration of the last dose to assess bioavailability. Rotenone was not detectable in plasma at a lower limit of quantification of 2 ng/mL (5 nM), showing that oral rotenone had insufficient bioavailability to achieve sustained systemic drug levels in mice. Hence, oral rotenone caused local gastrointestinal toxicity evident as lack of weight gain but failed to evoke behavioral or biological correlates of PD within 8 weeks.
Collapse
Affiliation(s)
- Ellen Niederberger
- Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor Stern-Kai 7, 60590 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor Stern-Kai 7, 60596 Frankfurt, Germany
- Correspondence: ; Tel.: +49-69-6301-7616; Fax: +49-69-6301-7636
| | - Annett Wilken-Schmitz
- Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor Stern-Kai 7, 60590 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor Stern-Kai 7, 60596 Frankfurt, Germany
| | - Christine Manderscheid
- Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor Stern-Kai 7, 60590 Frankfurt, Germany
| | - Yannick Schreiber
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor Stern-Kai 7, 60596 Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern-Kai 7, 60596 Frankfurt, Germany
| | - Robert Gurke
- Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor Stern-Kai 7, 60590 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor Stern-Kai 7, 60596 Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Theodor Stern-Kai 7, 60596 Frankfurt, Germany
| | - Irmgard Tegeder
- Institute for Clinical Pharmacology, Goethe-University Frankfurt, Theodor Stern-Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
38
|
Li N, Gao X, Zheng L, Huang Q, Zeng F, Chen H, Farag MA, Zhao C. Advances in fucoxanthin chemistry and management of neurodegenerative diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 105:154352. [PMID: 35917771 DOI: 10.1016/j.phymed.2022.154352] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/24/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Neurodegenerative diseases are chronic, currently incurable, diseases of the elderly, which are characterized by protein misfolding and neuronal damage. Fucoxanthin, derived from marine brown algae, presents a promising candidate for the development of effective therapeutic strategies. HYPOTHESIS AND PURPOSE The relationship between neurodegenerative disease management and fucoxanthin has not yet been clarified. This study focuses on the fundamental mechanisms and targets of fucoxanthin in Alzheimer's and Parkinson's disease management, showing that communication between the brain and the gut contributes to neurodegenerative diseases and early diagnosis of ophthalmic diseases. This paper also presents, new insights for future therapeutic directions based on the integrated application of artificial intelligence. CONCLUSION Fucoxanthin primarily binds to amyloid fibrils with spreading properties such as Aβ, tau, and α-synuclein to reduce their accumulation levels, alleviate inflammatory factors, and restore mitochondrial membranes to prevent oxidative stress via Nrf2 and Akt signaling pathways, involving reduction of specific secretases. In addition, fucoxanthin may serve as a preventive diagnosis for neurodegenerative diseases through ophthalmic disorders. It can modulate gut microbes and has potential for the alleviation and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Na Li
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xiaoxiang Gao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Lingjun Zheng
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qihui Huang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Feng Zeng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Hongbin Chen
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou 362000, China.
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Cairo, Egypt.
| | - Chao Zhao
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
39
|
Manfready RA, Goetz CG, Keshavarzian A. Intestinal microbiota and neuroinflammation in Parkinson's disease: At the helm of the gut-brain axis. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 167:81-99. [PMID: 36427960 DOI: 10.1016/bs.irn.2022.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Emerging data suggest that disrupted intestinal microbiota, or dysbiosis, may be responsible for multiple features of Parkinson's disease (PD), from initiation, to progression, to therapeutic response. We have progressed greatly in our understanding of microbial signatures associated with PD, and have gained important insights into how dysbiosis and intestinal permeability promote neurodegeneration through neuroinflammation and Lewy body formation. These insights underscore the potential of microbiota-directed therapies, which include dietary, pharmacologic, and lifestyle interventions.
Collapse
Affiliation(s)
- Richard A Manfready
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, United States
| | - Christopher G Goetz
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, United States
| | - Ali Keshavarzian
- Department of Internal Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, IL, United States; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States.
| |
Collapse
|
40
|
Sawamura M, Onoe H, Tsukada H, Isa K, Yamakado H, Okuda S, Ikuno M, Hatanaka Y, Murayama S, Uemura N, Isa T, Takahashi R. Lewy Body Disease Primate Model with α-Synuclein Propagation from the Olfactory Bulb. Mov Disord 2022; 37:2033-2044. [PMID: 35989519 DOI: 10.1002/mds.29161] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/24/2022] [Accepted: 07/01/2022] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Lewy body diseases (LBDs), which are pathologically defined as the presence of intraneuronal α-synuclein (α-Syn) inclusions called Lewy bodies, encompass Parkinson's disease, Parkinson's disease with dementia, and dementia with Lewy bodies. Autopsy studies have shown that the olfactory bulb (OB) is one of the regions where Lewy pathology develops and initiates its spread in the brain. OBJECTIVE This study aims to clarify how Lewy pathology spreads from the OB and affects brain functions using nonhuman primates. METHODS We inoculated α-Syn preformed fibrils into the unilateral OBs of common marmosets (Callithrix jacchus) and performed pathological analyses, manganese-enhanced magnetic resonance imaging, and 18 F-fluoro-2-deoxy-d-glucose positron emission tomography up to 6 months postinoculation. RESULTS Severe α-Syn pathology was observed within the olfactory pathway and limbic system, while mild α-Syn pathology was seen in a wide range of brain regions, including the substantia nigra pars compacta, locus coeruleus, and even dorsal motor nucleus of the vagus nerve. The brain imaging analyses showed reduction in volume of the OB and progressive glucose hypometabolism in widespread brain regions, including the occipital lobe, and extended beyond the pathologically affected regions. CONCLUSIONS We generated a novel nonhuman primate LBD model with α-Syn propagation from the OB. This model suggests that α-Syn propagation from the OB is related to OB atrophy and cerebral glucose hypometabolism in LBDs. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Masanori Sawamura
- Department of Neurology Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hirotaka Onoe
- Human Brain Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hideo Tsukada
- Central Research Laboratory, Hamamatsu Photonics K.K, Shizuoka, Japan
| | - Kaoru Isa
- Department of Physiology and Neurobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hodaka Yamakado
- Department of Neurology Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinya Okuda
- Department of Neurology Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masashi Ikuno
- Department of Neurology Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yusuke Hatanaka
- Department of Neurology Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigeo Murayama
- Department of Neuropathology (Brain Bank for Aging Research), Tokyo Metropolitan Geriatric Hospital & Institute of Gerontology, Tokyo, Japan
- Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Osaka, Japan
| | - Norihito Uemura
- Department of Neurology Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tadashi Isa
- Human Brain Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Physiology and Neurobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
41
|
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
|
42
|
Chen SJ, Lin CH. Gut microenvironmental changes as a potential trigger in Parkinson's disease through the gut-brain axis. J Biomed Sci 2022; 29:54. [PMID: 35897024 PMCID: PMC9327249 DOI: 10.1186/s12929-022-00839-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/22/2022] [Indexed: 11/10/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease attributed to the synergistic effects of genetic risk and environmental stimuli. Although PD is characterized by motor dysfunction resulting from intraneuronal alpha-synuclein accumulations, termed Lewy bodies, and dopaminergic neuronal degeneration in the substantia nigra, multiple systems are involved in the disease process, resulting in heterogenous clinical presentation and progression. Genetic predisposition to PD regarding aberrant immune responses, abnormal protein aggregation, autophagolysosomal impairment, and mitochondrial dysfunction leads to vulnerable neurons that are sensitive to environmental triggers and, together, result in neuronal degeneration. Neuropathology studies have shown that, at least in some patients, Lewy bodies start from the enteric nervous system and then spread to the central dopaminergic neurons through the gut-brain axis, suggesting the contribution of an altered gut microenvironment in the pathogenesis of PD. A plethora of evidence has revealed different gut microbiomes and gut metabolites in patients with PD compared to unaffected controls. Chronic gut inflammation and impaired intestinal barrier integrity have been observed in human PD patients and mouse models of PD. These observations led to the hypothesis that an altered gut microenvironment is a potential trigger of the PD process in a genetically susceptible host. In this review, we will discuss the complex interplay between genetic factors and gut microenvironmental changes contributing to PD pathogenesis.
Collapse
Affiliation(s)
- Szu-Ju Chen
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan.,Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100, Taiwan. .,Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
43
|
Tsafaras G, Baekelandt V. The role of LRRK2 in the periphery: link with Parkinson's disease and inflammatory diseases. Neurobiol Dis 2022; 172:105806. [PMID: 35781002 DOI: 10.1016/j.nbd.2022.105806] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/30/2022] [Accepted: 06/22/2022] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is currently considered a multisystemic disorder rather than a pure brain disease, in line with the multiple hit hypothesis from Braak. However, despite increasing evidence that the pathology might originate in the periphery, multiple unknown aspects and contradictory data on the pathological processes taking place in the periphery jeopardize the interpretation and therapeutic targeting of PD. Mutations in the leucine-rich-repeat kinase 2 (LRRK2) gene have been widely linked with familial and sporadic PD cases. However, the actual role of LRRK2 in PD pathophysiology is far from understood. There is evidence that LRRK2 may be involved in alpha-synuclein (α-synuclein) pathology and immune cell regulation, but it has also been associated with inflammatory diseases such as inflammatory bowel disease, tuberculosis, leprosy, and several other bacterial infections. In this review, we focus on the different roles of LRRK2 in the periphery. More specifically, we discuss the involvement of LRRK2 in the propagation of α-synuclein pathology and its regulatory role in peripheral inflammation. A deeper understanding of the multidimensional functions of LRRK2 will pave the way for more accurate characterization of PD pathophysiology and its association with other inflammatory diseases.
Collapse
Affiliation(s)
- George Tsafaras
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| |
Collapse
|
44
|
Yang R, Gao G, Yang H. The Pathological Mechanism Between the Intestine and Brain in the Early Stage of Parkinson's Disease. Front Aging Neurosci 2022; 14:861035. [PMID: 35813958 PMCID: PMC9263383 DOI: 10.3389/fnagi.2022.861035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 06/02/2022] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is the second most common chronic progressive neurodegenerative disease. The main pathological features are progressive degeneration of neurons and abnormal accumulation of α-synuclein. At present, the pathogenesis of PD is not completely clear, and many changes in the intestinal tract may be the early pathogenic factors of PD. These changes affect the central nervous system (CNS) through both nervous and humoral pathways. α-Synuclein deposited in the intestinal nerve migrates upward along the vagus nerve to the brain. Inflammation and immune regulation mediated by intestinal immune cells may be involved, affecting the CNS through local blood circulation. In addition, microorganisms and their metabolites may also affect the progression of PD. Therefore, paying attention to the multiple changes in the intestinal tract may provide new insight for the early diagnosis and treatment of PD.
Collapse
|
45
|
Non-human primates in prion diseases. Cell Tissue Res 2022; 392:7-20. [PMID: 35661921 DOI: 10.1007/s00441-022-03644-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 05/21/2022] [Indexed: 11/02/2022]
Abstract
The fascinating history of prion diseases is intimately linked to the use of nonhuman primates as experimental models, which brought so fundamental and founding information about transmissibility, pathogenesis, and resistance of prions. These models are still of crucial need for risk assessment of human health and may contribute to pave a new way towards the moving field of prion-like entities which now includes the main human neurodegenerative diseases (especially Alzheimer's and Parkinson's diseases).
Collapse
|
46
|
Nishiwaki H, Ito M, Hamaguchi T, Maeda T, Kashihara K, Tsuboi Y, Ueyama J, Yoshida T, Hanada H, Takeuchi I, Katsuno M, Hirayama M, Ohno K. Short chain fatty acids-producing and mucin-degrading intestinal bacteria predict the progression of early Parkinson's disease. NPJ Parkinsons Dis 2022; 8:65. [PMID: 35650236 PMCID: PMC9160257 DOI: 10.1038/s41531-022-00328-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 05/05/2022] [Indexed: 01/07/2023] Open
Abstract
To elucidate the relevance of gut dysbiosis in Parkinson’s disease (PD) in disease progression, we made random forest models to predict the progression of PD in two years by gut microbiota in 165 PD patients. The area under the receiver operating characteristic curves (AUROCs) of gut microbiota-based models for Hoehn & Yahr (HY) stages 1 and 2 were 0.799 and 0.705, respectively. Similarly, gut microbiota predicted the progression of Movement Disorder Society-Unified Parkinson’s Disease Rating Scale (MDS-UPDRS) III scores in an early stage of PD with AUROC = 0.728. Decreases of short-chain fatty acid-producing genera, Fusicatenibacter, Faecalibacterium, and Blautia, as well as an increase of mucin-degrading genus Akkermansia, predicted accelerated disease progression. The four genera remained unchanged in two years in PD, indicating that the taxonomic changes were not the consequences of disease progression. PD patients with marked gut dysbiosis may thus be destined to progress faster than those without gut dysbiosis.
Collapse
Affiliation(s)
- Hiroshi Nishiwaki
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomonari Hamaguchi
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuya Maeda
- Division of Neurology and Gerontology, Department of Internal Medicine, School of Medicine, Iwate Medical University, Iwate, Japan
| | | | - Yoshio Tsuboi
- Department of Neurology, Fukuoka University, Fukuoka, Japan
| | - Jun Ueyama
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takumi Yoshida
- Department of Computer Science, Nagoya Institute of Technology, Nagoya, Japan
| | - Hiroyuki Hanada
- Center for Advanced Intelligence Project, RIKEN, Tokyo, Japan
| | - Ichiro Takeuchi
- Department of Computer Science, Nagoya Institute of Technology, Nagoya, Japan.,Center for Advanced Intelligence Project, RIKEN, Tokyo, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaaki Hirayama
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
47
|
Mawe GM, Browning KN, Manfredsson FP, Camilleri M, Hamilton FA, Hollander JA, Sieber BA, Greenwel P, Shea-Donohue T, Wiley JW. 2021 Workshop: Neurodegenerative Diseases in the Gut-Brain Axis-Parkinson's Disease. Gastroenterology 2022; 162:1574-1582. [PMID: 35149029 PMCID: PMC9038653 DOI: 10.1053/j.gastro.2022.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 12/02/2022]
Affiliation(s)
- Gary M Mawe
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont.
| | - Kirsteen N Browning
- Department of Neural and Behavioral Science, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Fredric P Manfredsson
- Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, Arizona
| | | | - Frank A Hamilton
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jonathan A Hollander
- National Institute of Environmental Health Sciences, National Institutes of Health, Bethesda, Maryland
| | - Beth-Anne Sieber
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Patricia Greenwel
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Terez Shea-Donohue
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - John W Wiley
- Department of Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
48
|
Yoshida S, Hasegawa T. Deciphering the prion-like behavior of pathogenic protein aggregates in neurodegenerative diseases. Neurochem Int 2022; 155:105307. [PMID: 35181393 DOI: 10.1016/j.neuint.2022.105307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/12/2022] [Accepted: 02/13/2022] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases are hitherto classified based on their core clinical features, the anatomical distribution of neurodegeneration, and the cell populations mainly affected. On the other hand, the wealth of neuropathological, genetic, molecular and biochemical studies have identified the existence of distinct insoluble protein aggregates in the affected brain regions. These findings have spread the use of a collective term, proteinopathy, for neurodegenerative disorders with particular type of structurally altered protein accumulation. Particularly, a recent breakthrough in this field came with the discovery that these protein aggregates can transfer from one cell to another, thereby converting normal proteins to potentially toxic, misfolded species in a prion-like manner. In this review, we focus specifically on the molecular and cellular basis that underlies the seeding activity and transcellular spreading phenomenon of neurodegeneration-related protein aggregates, and discuss how these events contribute to the disease progression.
Collapse
Affiliation(s)
- Shun Yoshida
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 9808574, Japan; Department of Neurology, National Hospital Organization Yonezawa Hospital, Yonezawa, Yamagata, 992-1202, Japan
| | - Takafumi Hasegawa
- Division of Neurology, Department of Neuroscience & Sensory Organs, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 9808574, Japan.
| |
Collapse
|
49
|
Anis E, Xie A, Brundin L, Brundin P. Digesting recent findings: gut alpha-synuclein, microbiome changes in Parkinson's disease. Trends Endocrinol Metab 2022; 33:147-157. [PMID: 34949514 DOI: 10.1016/j.tem.2021.11.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 11/24/2021] [Indexed: 01/22/2023]
Abstract
Two hallmarks of Parkinson's disease (PD) are the widespread deposition of misfolded alpha-synuclein (αSyn) protein in the nervous system and loss of substantia nigra dopamine neurons. Recent research has suggested that αSyn aggregates in the enteric nervous system (ENS) lead to prodromal gastrointestinal (GI) symptoms such as constipation in PD, then propagating to the brain stem and eventually triggering neurodegeneration and motor symptoms. Additionally, whether the microbiome changes in PD contribute to the primary pathogenesis or, alternatively, are consequential to either the disease process or medication is still unclear. In this review, we discuss the possible roles of αSyn and microbiome changes in the GI system in PD and consider if and how the changes interact and contribute to the disease process and symptoms.
Collapse
Affiliation(s)
- Ehraz Anis
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Aoji Xie
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Lena Brundin
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Patrik Brundin
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
50
|
Chen H, Wang K, Scheperjans F, Killinger B. Environmental triggers of Parkinson's disease - Implications of the Braak and dual-hit hypotheses. Neurobiol Dis 2022; 163:105601. [PMID: 34954321 PMCID: PMC9525101 DOI: 10.1016/j.nbd.2021.105601] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/05/2021] [Accepted: 12/22/2021] [Indexed: 11/22/2022] Open
Abstract
Idiopathic Parkinson's disease (PD) may take decades to develop, during which many risk or protective factors may come into play to initiate the pathogenesis or modify its progression to clinical PD. The lack of understanding of this prodromal phase of PD and the factors involved has been a major hurdle in the study of PD etiology and preventive strategies. Although still controversial, the Braak and dual-hit hypotheses that PD may start peripherally in the olfactory structures and/or the gut provides a theoretical platform to identify the triggers and modifiers of PD prodromal development and progression. This is particularly true for the search of environmental causes of PD as the olfactory structures and gut are the major human mucosal interfaces with the environment. In this review, we lay out our personal views about how the Braak and dual-hit hypotheses may help us search for the environmental triggers and modifiers for PD, summarize available experimental and epidemiological evidence, and discuss research gaps and strategies.
Collapse
Affiliation(s)
- Honglei Chen
- Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA.
| | - Keran Wang
- Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Filip Scheperjans
- Department of Neurology, Helsinki University Hospital, and Clinicum, University of Helsinki, Haartmaninkatu 4, 00290 Helsinki, Finland
| | - Bryan Killinger
- Graduate College, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|