1
|
Park SJ, Kim KW, Lee EJ. Gut-brain axis and environmental factors in Parkinson's disease: bidirectional link between disease onset and progression. Neural Regen Res 2025; 20:3416-3429. [PMID: 39688568 PMCID: PMC11974660 DOI: 10.4103/nrr.nrr-d-24-00994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Parkinson's disease has long been considered a disorder that primarily affects the brain, as it is defined by the dopaminergic neurodegeneration in the substantia nigra and the brain accumulation of Lewy bodies containing α-synuclein protein. In recent decades, however, accumulating research has revealed that Parkinson's disease also involves the gut and uncovered an intimate and important bidirectional link between the brain and the gut, called the "gut-brain axis." Numerous clinical studies demonstrate that gut dysfunction frequently precedes motor symptoms in Parkinson's disease patients, with findings including impaired intestinal permeability, heightened inflammation, and distinct gut microbiome profiles and metabolites. Furthermore, α-synuclein deposition has been consistently observed in the gut of Parkinson's disease patients, suggesting a potential role in disease initiation. Importantly, individuals with vagotomy have a reduced Parkinson's disease risk. From these observations, researchers have hypothesized that α-synuclein accumulation may initiate in the gut and subsequently propagate to the central dopaminergic neurons through the gut-brain axis, leading to Parkinson's disease. This review comprehensively examines the gut's involvement in Parkinson's disease, focusing on the concept of a gut-origin for the disease. We also examine the interplay between altered gut-related factors and the accumulation of pathological α-synuclein in the gut of Parkinson's disease patients. Given the accessibility of the gut to both dietary and pharmacological interventions, targeting gut-localized α-synuclein represents a promising avenue for developing effective Parkinson's disease therapies.
Collapse
Affiliation(s)
- Soo Jung Park
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Kyung Won Kim
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Chuncheon, South Korea
| | - Eun Jeong Lee
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| |
Collapse
|
2
|
Qin K, Li Y, Liu Y, Xue L, Wang Z, Xian W, Tu R, Yang B, Ning F, Xie A. Divergent amygdala function in proposed brain-first and body-first Parkinson's disease: a resting-state functional magnetic resonance imaging study. J Affect Disord 2025; 382:123-130. [PMID: 40250815 DOI: 10.1016/j.jad.2025.04.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/05/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND The newly proposed α-Synuclein Origin and Connectome (SOC) Model classifies Parkinson's disease (PD) patients into brain-first and body-first subtypes. In brain-first patients, α-synuclein may originate in the amygdala of one cerebral hemisphere and disseminate ipsilaterally via the neural connectome. This study aimed to investigate the differences in clinical characteristics and amygdala function between these two subtypes and to evaluate whether amygdala function could serve as a marker for subtype distinctions. METHODS Resting-state functional MRI data of 66 early-stage PD patients and 17 healthy controls (HC) were retrieved from the Parkinson's Progression Markers Initiative database. PD patients with REM Sleep Behavior Disorder (RBD) were classified as the body-first subtype, while those without RBD were classified as the brain-first subtype. RESULTS We found that body-first patients had a longer disease duration and more severe autonomic dysfunction compared to brain-first patients. Amygdala-related FC in brain-first patients was similar to that in the HC group, with both groups showing stronger FC between the bilateral amygdala and the right postcentral gyrus than body-first patients. Importantly, the abnormal amygdala-related FC was negatively correlated with SCOPA-Aut scores (r = -0.361, P = 0.002) in PD patients. ROC analysis indicated that the area under the curve for the FC was 0.834. CONCLUSION Our findings suggest that the amygdala-related FC may serve as an effective indicator to differentiate brain-first and body-first subtypes. Moreover, functional abnormalities in the amygdala contribute to autonomic dysfunction, rather than depression or anxiety in early-stage PD patients. Further validation of these findings in trials with larger cohorts is needed.
Collapse
Affiliation(s)
- Kunpeng Qin
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaqing Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yumei Liu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li Xue
- Record room, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zihan Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenke Xian
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruizi Tu
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bohan Yang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fangbo Ning
- Department of Neurology, Taian City Central Hospital, Taian, China.
| | - Anmu Xie
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
3
|
Wang X, Chen H, Ma X, Liu H, Wu D, Du W, He J, Li S, Chen H, Wu T, Feng T, Su W. Atrophy of ventral diencephalon is associated with freezing of gait in Parkinson's disease: analysis of two cohorts. NPJ Parkinsons Dis 2025; 11:44. [PMID: 40050652 PMCID: PMC11885666 DOI: 10.1038/s41531-025-00893-5] [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: 05/16/2024] [Accepted: 02/21/2025] [Indexed: 03/09/2025] Open
Abstract
Evidence regarding brain structural atrophy associated with Freezing of Gait (FOG) in Parkinson's disease (PD) is inconsistent. We analyzed cortical thickness and subcortical nuclei volumes using FreeSurfer in two large PD cohorts. In cohort 1 (N = 316), multivariate analyses identified reduced pallidum and ventral diencephalon (VDC) volumes as significantly associated with FOG presence. Validation in the Parkinson's Progression Markers Initiative (PPMI) cohort (cohort 2, N = 94) demonstrated that decreased VDC volume at four-year follow-up independently predicted higher FOG risk, improving the predictive model's accuracy when combined with PIGD score, CSF Aβ42, and caudate DAT uptake (AUC 0.760; Δχ2 = 5.449, P = 0.020; Z = 2.211, P = 0.027). VDC volume is also correlated with FOG severity. These findings suggest that VDC atrophy may underlie FOG mechanisms and serve as a biomarker for its progression in PD patients.
Collapse
Affiliation(s)
- Xuemei Wang
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Huimin Chen
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Xinxin Ma
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Huijing Liu
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Dongdong Wu
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Du
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing He
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuhua Li
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Haibo Chen
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Tao Wu
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Feng
- Center for Movement Disorders, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Wen Su
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
4
|
Faldu KG, Shah JS. Ambroxol Improves Amyloidogenic, NF-κB, and Nrf2 Pathways in a Scopolamine-Induced Cognitive Impairment Rat Model of Alzheimer's Disease. Drug Dev Res 2024; 85:e70017. [PMID: 39533780 DOI: 10.1002/ddr.70017] [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: 06/17/2024] [Revised: 10/13/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Ambroxol (ABX) is used to manage excessive production of mucus in the respiratory system. The present study sought to assess the neuroprotective potential of ambroxol by influencing the amyloidogenic, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways in a rat model of Alzheimer's disease (AD) induced by scopolamine. The AD pathology was induced by chronic administration of scopolamine. The rats were given scopolamine at a dose of 2 mg/kg via intraperitoneal injection daily for 14 days, followed by treatment (ABX 121.5, 135, and 180 mg/kg orally and 5 mg/kg orally of donepezil) for the next 28 days while continuing to receive daily scopolamine injection. The behavior of the rats was evaluated using Modified Y-Maze and Novel object recognition tasks. Analyses were carried out on AD pathological markers [Amyloid beta peptide 1-40, Amyloid beta peptide 1-42, acetylcholinesterase, beta-secretase 1 (BACE1), total tau, and p-tau], inflammatory markers [NF-κB, tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6), and interferon γ], antioxidant markers (Nrf2 and heme Oxygenase 1 (HO-1)], along with synaptophysin and glial fibrillary acidic protein (GFAP) immunohistochemistry and histopathological assessment of the hippocampus. Our findings indicated that ABX reduced impairment in behavior. Levels of Acetylcholinesterase, BACE1, amyloid beta 1-40, amyloid beta 1-42, total tau, p-tau, NF-κB, IFN-γ, IL-6, and TNF-α decreased significantly. There was a significant increase in the levels of HO-1 and Nrf2. It stopped the neuronal degeneration, raised synaptophysin immunoreactivity, and lowered GFAP immunoreactivity. The current research indicates that ambroxol may possess senomorphic properties by impacting the transcription factors NF-κB and senescence-associated secretory phenotype (SASP). Consequently, it could provide neuroprotection through alterations in the Nrf2 and NF-κB signaling pathways in AD.
Collapse
Affiliation(s)
- Khushboo Govind Faldu
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Jigna Samir Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| |
Collapse
|
5
|
Jordi L, Isacson O. Neuronal threshold functions: Determining symptom onset in neurological disorders. Prog Neurobiol 2024; 242:102673. [PMID: 39389338 PMCID: PMC11809673 DOI: 10.1016/j.pneurobio.2024.102673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/18/2024] [Accepted: 10/06/2024] [Indexed: 10/12/2024]
Abstract
Synaptic networks determine brain function. Highly complex interconnected brain synaptic networks provide output even under fluctuating or pathological conditions. Relevant to the treatment of brain disorders, understanding the limitations of such functional networks becomes paramount. Here we use the example of Parkinson's Disease (PD) as a system disorder, with PD symptomatology emerging only when the functional reserves of neurons, and their interconnected networks, are unable to facilitate effective compensatory mechanisms. We have denoted this the "threshold theory" to account for how PD symptoms develop in sequence. In this perspective, threshold functions are delineated in a quantitative, synaptic, and cellular network context. This provides a framework to discuss the development of specific symptoms. PD includes dysfunction and degeneration in many organ systems and both peripheral and central nervous system involvement. The threshold theory accounts for and explains the reasons why parallel gradually emerging pathologies in brain and peripheral systems generate specific symptoms only when functional thresholds are crossed, like tipping points. New and mounting evidence demonstrate that PD and related neurodegenerative diseases are multisystem disorders, which transcends the traditional brain-centric paradigm. We believe that representation of threshold functions will be helpful to develop new medicines and interventions that are specific for both pre- and post-symptomatic periods of neurodegenerative disorders.
Collapse
Affiliation(s)
- Luc Jordi
- Neuroregeneration Institute, McLean Hospital / Harvard Medical School, Belmont, MA 02478, USA.
| | - Ole Isacson
- Neuroregeneration Institute, McLean Hospital / Harvard Medical School, Belmont, MA 02478, USA; Department of Neurology and Program in Neuroscience, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Dundon NM, Stuber A, Bullock T, Garcia JO, Babenko V, Rizor E, Yang D, Giesbrecht B, Grafton ST. Cardiac-Sympathetic Contractility and Neural Alpha-Band Power: Cross-Modal Collaboration during Approach-Avoidance Conflict. J Neurosci 2024; 44:e2008232024. [PMID: 39214705 PMCID: PMC11466073 DOI: 10.1523/jneurosci.2008-23.2024] [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: 10/24/2023] [Revised: 08/09/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
As evidence mounts that the cardiac-sympathetic nervous system reacts to challenging cognitive settings, we ask if these responses are epiphenomenal companions or if there is evidence suggesting a more intertwined role of this system with cognitive function. Healthy male and female human participants performed an approach-avoidance paradigm, trading off monetary reward for painful electric shock, while we recorded simultaneous electroencephalographic and cardiac-sympathetic signals. Participants were reward sensitive but also experienced approach-avoidance "conflict" when the subjective appeal of the reward was near equivalent to the revulsion of the cost. Drift-diffusion model parameters suggested that participants managed conflict in part by integrating larger volumes of evidence into choices (wider decision boundaries). Late alpha-band (neural) dynamics were consistent with widening decision boundaries serving to combat reward sensitivity and spread attention more fairly to all dimensions of available information. Independently, wider boundaries were also associated with cardiac "contractility" (an index of sympathetically mediated positive inotropy). We also saw evidence of conflict-specific "collaboration" between the neural and cardiac-sympathetic signals. In states of high conflict, the alignment (i.e., product) of alpha dynamics and contractility were associated with a further widening of the boundary, independent of either signal's singular association. Cross-trial coherence analyses provided additional evidence that the autonomic systems controlling cardiac-sympathetics might influence the assessment of information streams during conflict by disrupting or overriding reward processing. We conclude that cardiac-sympathetic control might play a critical role, in collaboration with cognitive processes, during the approach-avoidance conflict in humans.
Collapse
Affiliation(s)
- Neil M Dundon
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, University of Freiburg, Freiburg 79104, Germany
| | - Alexander Stuber
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, California 93106
| | - Tom Bullock
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, California 93106
| | - Javier O Garcia
- Humans in Complex Systems Division, US DEVCOM Army Research Laboratory, Aberdeen Proving Ground, Maryland 21005
| | - Viktoriya Babenko
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, California 93106
- BIOPAC Systems Inc., Goleta, California 93117
| | - Elizabeth Rizor
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, California 93106
- Interdepartmental Graduate Program in Dynamical Neuroscience, University of California, Santa Barbara, California 93106
| | - Dengxian Yang
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, California 93106
- Department of Computer Science, University of California, Santa Barbara, California 93106
| | - Barry Giesbrecht
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
- Institute for Collaborative Biotechnologies, University of California, Santa Barbara, California 93106
- Interdepartmental Graduate Program in Dynamical Neuroscience, University of California, Santa Barbara, California 93106
| | - Scott T Grafton
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, California 93106
| |
Collapse
|
7
|
Paulėkas E, Vanagas T, Lagunavičius S, Pajėdienė E, Petrikonis K, Rastenytė D. Navigating the Neurobiology of Parkinson's: The Impact and Potential of α-Synuclein. Biomedicines 2024; 12:2121. [PMID: 39335634 PMCID: PMC11429448 DOI: 10.3390/biomedicines12092121] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease worldwide; therefore, since its initial description, significant progress has been made, yet a mystery remains regarding its pathogenesis and elusive root cause. The widespread distribution of pathological α-synuclein (αSyn) aggregates throughout the body raises inquiries regarding the etiology, which has prompted several hypotheses, with the most prominent one being αSyn-associated proteinopathy. The identification of αSyn protein within Lewy bodies, coupled with genetic evidence linking αSyn locus duplication, triplication, as well as point mutations to familial Parkinson's disease, has underscored the significance of αSyn in initiating and propagating Lewy body pathology throughout the brain. In monogenic and sporadic PD, the presence of early inflammation and synaptic dysfunction leads to αSyn aggregation and neuronal death through mitochondrial, lysosomal, and endosomal functional impairment. However, much remains to be understood about αSyn pathogenesis, which is heavily grounded in biomarkers and treatment strategies. In this review, we provide emerging new evidence on the current knowledge about αSyn's pathophysiological impact on PD, and its presumable role as a specific disease biomarker or main target of disease-modifying therapies, highlighting that this understanding today offers the best potential of disease-modifying therapy in the near future.
Collapse
Affiliation(s)
- Erlandas Paulėkas
- Department of Neurology, Lithuanian University of Health Sciences Kaunas Clinics, LT-50161 Kaunas, Lithuania; (T.V.); (S.L.); (E.P.); (K.P.); (D.R.)
| | | | | | | | | | | |
Collapse
|
8
|
Yoo SW, Ryu DW, Oh Y, Ha S, Lyoo CH, Kim JS. Unraveling olfactory subtypes in Parkinson's disease and their effect on the natural history of the disease. J Neurol 2024; 271:6102-6113. [PMID: 39043904 DOI: 10.1007/s00415-024-12586-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND Hyposmia in Parkinson's disease (PD) had been studied before but had not been detailed by its temporal progression. This study observed how each olfactory subtype evolved in terms of motor symptoms, cardiac sympathetic innervation, and cognition. METHODS Two hundred and three early PD patients were classified as normosmia, hyposmia-converter (hypo-converter), and hyposmia. Their presynaptic monoamine availability at the time of diagnosis was assessed by positron emission tomography imaging using 18F-N-(3-fluoropropyl)-2beta-carbon ethoxy-3beta-(4-iodophenyl) nortropane and compared across the subtypes. Motor symptoms were evaluated in all patients, cardiac denervation was examined in 183 patients, and cognition in 195 patients were assessed using a neuropsychological battery. The domains were re-assessed 2-4 times, and the longitudinal data were analyzed to discern the natural course of each subtype. RESULTS Twenty-nine (14.3%) patients belonged to the normosmia group, 34 (16.7%) to the hypo-converter group, and the rest to the hyposmia (69.0%) group. 85.7% of the total population became hyposmic during an average 3 years of follow-up. The baseline motor symptoms, cardiac denervation, and cognition were comparable across the olfactory subtypes. Across the subtypes, a decline in the presynaptic monoamine densities of the caudate, especially the ventral-anterior subdivisions, correlated inversely with olfaction dysfunction. Over time, motor and cardiac denervation burdens worsened regardless of olfactory subtypes, but hypo-converters experienced faster cognitive deterioration than the other two groups. CONCLUSIONS The results suggest that the olfactory subtypes have differential significance along the disease course, which might reflect the involvement of different neuro-biochemical circuitries.
Collapse
Affiliation(s)
- Sang-Won Yoo
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Dong-Woo Ryu
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Yoonsang Oh
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chul Hyoung Lyoo
- Department of Neurology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joong-Seok Kim
- Department of Neurology, College of Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| |
Collapse
|
9
|
Romero A, Sanchez A, Jones JD, Ledesma K, El-Halawany MS, Hamouda AK, Bill BR. Optimization of Zebrafish Larvae 6-OHDA Exposure for Neurotoxin Induced Dopaminergic Marker Reduction. Zebrafish 2024; 21:287-293. [PMID: 38608227 PMCID: PMC11876810 DOI: 10.1089/zeb.2023.0098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that is clinically assessed by motor symptoms associated with the loss of midbrain dopaminergic neurons affecting the quality of life for over 8.5 million people worldwide. The neurotoxin 6-hydroxydopamine (6-OHDA) has been used to chemically induce a PD-like state in zebrafish larvae by several laboratories; however, highly variable concentration, methodology, and reagents have resulted in conflicting results suggesting a need to investigate these issues of reproducibility. We propose a protocol that addresses the differences in methodology and induces changes in 6 days postfertilization (dpf) larvae utilizing a 24-h exposure at 3 dpf with 30 μM 6-OHDA. Despite ∼50% lethality, no morphological or development differences in surviving fish are observed. Definition of our model is defined by downregulation of the expression of th1 by reverse transcriptase-quantitative polymerase chain reaction, a marker for dopaminergic neurons and a reduction in movement. Additionally, we observed a downregulation of pink1 and an upregulation of sod1 and sod2, indicators of mitochondrial dysfunction and response to reactive oxygen species, respectively.
Collapse
Affiliation(s)
- Adrian Romero
- The University of Texas at Tyler College of Arts and Sciences Department of Biology, Tyler, Texas, USA
- The University of Texas Tyler Ben and Maytee Fisch College of Pharmacy, Tyler, Texas, USA
| | - Armando Sanchez
- The University of Texas at Tyler College of Arts and Sciences Department of Biology, Tyler, Texas, USA
| | - Jocelyn D. Jones
- The University of Texas at Tyler College of Arts and Sciences Department of Biology, Tyler, Texas, USA
| | - Kristel Ledesma
- The University of Texas at Tyler College of Arts and Sciences Department of Biology, Tyler, Texas, USA
| | - Medhat S. El-Halawany
- The University of Texas Tyler Ben and Maytee Fisch College of Pharmacy, Tyler, Texas, USA
| | - Ayman K. Hamouda
- The University of Texas Tyler Ben and Maytee Fisch College of Pharmacy, Tyler, Texas, USA
| | - Brent R. Bill
- The University of Texas at Tyler College of Arts and Sciences Department of Biology, Tyler, Texas, USA
| |
Collapse
|
10
|
You M, Chen N, Yang Y, Cheng L, He H, Cai Y, Liu Y, Liu H, Hong G. The gut microbiota-brain axis in neurological disorders. MedComm (Beijing) 2024; 5:e656. [PMID: 39036341 PMCID: PMC11260174 DOI: 10.1002/mco2.656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 07/23/2024] Open
Abstract
Previous studies have shown a bidirectional communication between human gut microbiota and the brain, known as the microbiota-gut-brain axis (MGBA). The MGBA influences the host's nervous system development, emotional regulation, and cognitive function through neurotransmitters, immune modulation, and metabolic pathways. Factors like diet, lifestyle, genetics, and environment shape the gut microbiota composition together. Most research have explored how gut microbiota regulates host physiology and its potential in preventing and treating neurological disorders. However, the individual heterogeneity of gut microbiota, strains playing a dominant role in neurological diseases, and the interactions of these microbial metabolites with the central/peripheral nervous systems still need exploration. This review summarizes the potential role of gut microbiota in driving neurodevelopmental disorders (autism spectrum disorder and attention deficit/hyperactivity disorder), neurodegenerative diseases (Alzheimer's and Parkinson's disease), and mood disorders (anxiety and depression) in recent years and discusses the current clinical and preclinical gut microbe-based interventions, including dietary intervention, probiotics, prebiotics, and fecal microbiota transplantation. It also puts forward the current insufficient research on gut microbiota in neurological disorders and provides a framework for further research on neurological disorders.
Collapse
Affiliation(s)
- Mingming You
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Nan Chen
- Master of Public HealthSchool of Public HealthXiamen UniversityXiamenChina
| | - Yuanyuan Yang
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Lingjun Cheng
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Hongzhang He
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Yanhua Cai
- Master of Public HealthSchool of Public HealthXiamen UniversityXiamenChina
| | - Yating Liu
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Haiyue Liu
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| | - Guolin Hong
- Xiamen Key Laboratory of Genetic TestingThe Department of Laboratory MedicineThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenChina
| |
Collapse
|
11
|
Sian-Hulsmann J, Riederer P. The 'α-synucleinopathy syndicate': multiple system atrophy and Parkinson's disease. J Neural Transm (Vienna) 2024; 131:585-595. [PMID: 37227594 PMCID: PMC11192696 DOI: 10.1007/s00702-023-02653-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023]
Abstract
Multiple System Atrophy (MSA) and Parkinson's diseases (PD) are elite members of the α-synucleinopathy organization. Aberrant accumulations of the protein α-synuclein characterize them. A plethora of evidence indicates the involvement of these rogue inclusions in a cascade of events that disturb cellular homeostasis resulting in neuronal dysfunction. These two neurodegenerative diseases share many features both clinically and pathologically. Cytotoxic processes commonly induced by reactive free radical species have been associated with oxidative stress and neuroinflammation, frequently reported in both diseases. However, it appears they have characteristic and distinct α-synuclein inclusions. It is glial cytoplasmic inclusions in the case of MSA while Lewy bodies manifest in PD. This is probably related to the etiology of the illness. At present, precise mechanism(s) underlying the characteristic configuration of neurodegeneration are unclear. Furthermore, the "prion-like" transmission from cell to cell prompts the suggestion that perhaps these α-synucleinopathies are prion-like diseases. The possibility of some underlying genetic foul play remains controversial. But as major culprits of pathological processes or even single triggers of PD and MSA are the same-like oxidative stress, iron-induced pathology, mitochondriopathy, loss of respiratory activity, loss of proteasomal function, microglial activation, neuroinflammation-it is not farfetched to assume that in sporadic PD and also in MSA a variety of combinations of susceptibility genes contribute to the regional specificity of pathological onset. These players of pathology, as mentioned above, in a synergistic combination, are responsible for driving the progression of PD, MSA and other neurodegenerative disorders. Elucidating the triggers and progression factors is vital for advocating disease modification or halting its progression in both, MSA and PD.
Collapse
Affiliation(s)
| | - Peter Riederer
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, Würzburg, Germany.
- Department of Psychiatry, University of Southern Denmark Odense, J.B. Winslows Vey 18, 5000, Odense, Denmark.
| |
Collapse
|
12
|
Xu Q, Jiang S, Kang R, Wang Y, Zhang B, Tian J. Deciphering the molecular pathways underlying dopaminergic neuronal damage in Parkinson's disease associated with SARS-CoV-2 infection. Comput Biol Med 2024; 171:108200. [PMID: 38428099 DOI: 10.1016/j.compbiomed.2024.108200] [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: 08/10/2023] [Revised: 01/24/2024] [Accepted: 02/18/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND The COVID-19 pandemic caused by SARS-CoV-2 has led to significant global morbidity and mortality, with potential neurological consequences, such as Parkinson's disease (PD). However, the underlying mechanisms remain elusive. METHODS To address this critical question, we conducted an in-depth transcriptome analysis of dopaminergic (DA) neurons in both COVID-19 and PD patients. We identified common pathways and differentially expressed genes (DEGs), performed enrichment analysis, constructed protein‒protein interaction networks and gene regulatory networks, and employed machine learning methods to develop disease diagnosis and progression prediction models. To further substantiate our findings, we performed validation of hub genes using a single-cell sequencing dataset encompassing DA neurons from PD patients, as well as transcriptome sequencing of DA neurons from a mouse model of MPTP(1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-induced PD. Furthermore, a drug-protein interaction network was also created. RESULTS We gained detailed insights into biological functions and signaling pathways, including ion transport and synaptic signaling pathways. CD38 was identified as a potential key biomarker. Disease diagnosis and progression prediction models were specifically tailored for PD. Molecular docking simulations and molecular dynamics simulations were employed to predict potential therapeutic drugs, revealing that genistein holds significant promise for exerting dual therapeutic effects on both PD and COVID-19. CONCLUSIONS Our study provides innovative strategies for advancing PD-related research and treatment in the context of the ongoing COVID-19 pandemic by elucidating the common pathogenesis between COVID-19 and PD in DA neurons.
Collapse
Affiliation(s)
- Qiuhan Xu
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Sisi Jiang
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Ruiqing Kang
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Yiling Wang
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Baorong Zhang
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| | - Jun Tian
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| |
Collapse
|
13
|
Fornstedt Wallin B. Oxidation of dopamine and related catechols in dopaminergic brain regions in Parkinson's disease and during ageing in non-Parkinsonian subjects. J Neural Transm (Vienna) 2024; 131:213-228. [PMID: 38238531 DOI: 10.1007/s00702-023-02718-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/28/2023] [Indexed: 02/18/2024]
Abstract
The present study was performed to examine if catechol oxidation is higher in brains from patients with Parkinson's disease compared to age-matched controls, and if catechol oxidation increases with age. Brain tissue from Parkinson patients and age-matched controls was examined for oxidation of dopamine, 3,4-dihydroxyphenylacetic acid (DOPAC) and 3,4-dihydroxyphenylalanine (DOPA) to corresponding quinones, by measurement of 5-S-cysteinyl-dopamine, 5-S-cysteinyl-DOPAC and 5-S-cysteinyl-DOPA. The cysteinyl catechols are assumed to be biomarkers for DA, DOPAC and DOPA autoxidation and part of the biosynthetic pathway of neuromelanin. The concentrations of the 5-S-cysteinyl catechols were lower, whereas the 5-S-cysteinyl-DA/DA and 5-S-cysteinyl-DOPAC/DOPAC ratios tended to be higher in the Parkinson group compared to controls, which was interpreted as a higher degree of oxidation. High 5-S-cysteinyl-DA/DA ratios were found in the substantia nigra of a sub-population of the Parkinson group. Based on 5-S-cysteinyl-DA/DA ratios, dopamine oxidation was found to increase statistically significantly with age in the caudate nucleus, and non-significantly in the substantia nigra. In conclusion, the occurrence of 5-S-cysteinyl-DA, 5-S-cysteinyl-DOPAC and 5-S-cysteinyl-DOPA was demonstrated in dopaminergic brain areas of humans, a tendency for higher oxidation of DA in the Parkinson group compared to controls was observed as well as a statistically significant increase in DA oxidation with age. Possibly, autoxidation of DA and other catechols are involved in both normal and pathological ageing of the brain. This study confirms one earlier but small study, as well as complements one study on non-PD cases and one study on both PD cases and controls on NM bound or integrated markers or catechols.
Collapse
Affiliation(s)
- Bodil Fornstedt Wallin
- Department of Pharmacology, University of Göteborg (at the time of the study), Göteborg, Sweden.
| |
Collapse
|
14
|
Pasricha TS, Guerrero-Lopez IL, Kuo B. Management of Gastrointestinal Symptoms in Parkinson's Disease: A Comprehensive Review of Clinical Presentation, Workup, and Treatment. J Clin Gastroenterol 2024; 58:211-220. [PMID: 38260966 PMCID: PMC10855995 DOI: 10.1097/mcg.0000000000001961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024]
Abstract
Gastrointestinal symptoms in Parkinson's disease (PD) are among the most prevalent and debilitating of complications and present unique diagnostic and management challenges. Patients with PD commonly experience dysphagia, nausea, bloating, and constipation related to pathologic involvement of the enteric nervous system. In turn, gastrointestinal complications may impact motor fluctuations and the efficacy of levodopa therapy. This review will explore the common gastrointestinal manifestations of PD with an emphasis on clinical presentation, workup, and treatment strategies.
Collapse
Affiliation(s)
- Trisha S. Pasricha
- Division of Gastroenterology, Massachusetts General Hospital
- Harvard Medical School, Boston, MA
| | | | - Braden Kuo
- Division of Gastroenterology, Massachusetts General Hospital
- Harvard Medical School, Boston, MA
| |
Collapse
|
15
|
Cooper O, Hallett P, Isacson O. Upstream lipid and metabolic systems are potential causes of Alzheimer's disease, Parkinson's disease and dementias. FEBS J 2024; 291:632-645. [PMID: 36165619 PMCID: PMC10040476 DOI: 10.1111/febs.16638] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/02/2022] [Accepted: 09/26/2022] [Indexed: 11/28/2022]
Abstract
Brain health requires circuits, cells and molecular pathways to adapt when challenged and to promptly reset once the challenge has resolved. Neurodegeneration occurs when adaptability becomes confined, causing challenges to overwhelm neural circuitry. Studies of rare and common neurodegenerative diseases suggest that the accumulation of lipids can compromise circuit adaptability. Using microglia as an example, we review data that suggest increased lipid concentrations cause dysfunctional inflammatory responses to immune challenges, leading to Alzheimer's disease, Parkinson's disease and dementia. We highlight current approaches to treat lipid metabolic and clearance pathways and identify knowledge gaps towards restoring adaptive homeostasis in individuals who are at-risk of losing cognition.
Collapse
Affiliation(s)
- Oliver Cooper
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| | - Penny Hallett
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| | - Ole Isacson
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| |
Collapse
|
16
|
Dorsey ER, De Miranda BR, Horsager J, Borghammer P. The Body, the Brain, the Environment, and Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2024; 14:363-381. [PMID: 38607765 PMCID: PMC11091648 DOI: 10.3233/jpd-240019] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/14/2024]
Abstract
The brain- and body-first models of Lewy body disorders predict that aggregated alpha-synuclein pathology usually begins in either the olfactory system or the enteric nervous system. In both scenarios the pathology seems to arise in structures that are closely connected to the outside world. Environmental toxicants, including certain pesticides, industrial chemicals, and air pollution are therefore plausible trigger mechanisms for Parkinson's disease and dementia with Lewy bodies. Here, we propose that toxicants inhaled through the nose can lead to pathological changes in alpha-synuclein in the olfactory system that subsequently spread and give rise to a brain-first subtype of Lewy body disease. Similarly, ingested toxicants can pass through the gut and cause alpha-synuclein pathology that then extends via parasympathetic and sympathetic pathways to ultimately produce a body-first subtype. The resulting spread can be tracked by the development of symptoms, clinical assessments, in vivo imaging, and ultimately pathological examination. The integration of environmental exposures into the brain-first and body-first models generates testable hypotheses, including on the prevalence of the clinical conditions, their future incidence, imaging patterns, and pathological signatures. The proposed link, though, has limitations and leaves many questions unanswered, such as the role of the skin, the influence of the microbiome, and the effects of ongoing exposures. Despite these limitations, the interaction of exogenous factors with the nose and the gut may explain many of the mysteries of Parkinson's disease and open the door toward the ultimate goal -prevention.
Collapse
Affiliation(s)
- E. Ray Dorsey
- Department of Neurology and Center for Health and Technology, University of Rochester Medical Center, Rochester, NY, USA
| | - Briana R. De Miranda
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jacob Horsager
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| | - Per Borghammer
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
17
|
Hu M, Skjærbæk C, Borghammer P. Approaches to Early Parkinson's Disease Subtyping. JOURNAL OF PARKINSON'S DISEASE 2024; 14:S297-S306. [PMID: 39331104 PMCID: PMC11492007 DOI: 10.3233/jpd-230419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/09/2024] [Indexed: 09/28/2024]
Abstract
Parkinson's disease (PD) unfolds with pathological processes and neurodegeneration well before the emergence of noticeable motor symptoms, providing a window for early identification. The extended prodromal phase allows the use of risk stratification measures and prodromal markers to pinpoint individuals likely to develop PD. Importantly, a growing body of evidence emphasizes the heterogeneity within prodromal and clinically diagnosed PD. The disease likely comprises distinct subtypes exhibiting diverse clinical manifestations, pathophysiological mechanisms, and patterns of α-synuclein progression in the central and peripheral nervous systems. There is a pressing need to refine the definition and early identification of these prodromal subtypes. This requires a comprehensive strategy that integrates genetic, pathological, imaging, and multi-omics markers, alongside careful observation of subtle motor and non-motor symptoms. Such multi-dimensional classification of early PD subtypes will improve our understanding of underlying disease pathophysiology, improve predictions of clinical endpoints, progression trajectory and medication response, contribute to drug discovery and personalized medicine by identifying subtype-specific disease mechanisms, and facilitate drug trials by reducing confounding effects of heterogeneity. Here we explore different subtyping methodologies in prodromal and clinical PD, focusing on clinical, imaging, genetic and molecular subtyping approaches. We also emphasize the need for refined, theoretical a priori disease models. These will be prerequisite to understanding the biological underpinnings of biological subtypes, which have been defined by large scale data-driven approaches and multi-omics fingerprints.
Collapse
Affiliation(s)
- Michele Hu
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Casper Skjærbæk
- Department of Nuclear Medicine & PET, Aarhus University Hospital, Aarhus, Denmark
| | - Per Borghammer
- Department of Nuclear Medicine & PET, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
18
|
Parmar M, Perrier AL. Introduction to stem cell biology and its role in treating neurologic disorders. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:3-14. [PMID: 39341661 DOI: 10.1016/b978-0-323-90120-8.00005-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Regenerative medicine is an emerging and rapidly evolving field of research and therapeutics aimed to restore, maintain, and improve body functions. In the adult mammalian brain, very few neurons, if any, are generated after disease onset or an injury, and its ability to self-repair is therefore limited. Replacing neurons that are lost during neurodegenerative diseases or due to injury therefore represents one of the major challenges to modern medicine. In this introductory chapter, we describe the basic biology of stem cells and outline how stem cells and cell reprogramming can be utilized to create new neurons for therapeutic purposes that are discussed in detail in other chapters in this handbook.
Collapse
Affiliation(s)
- Malin Parmar
- Developmental and Regenerative Neurobiology, Wallenberg Neuroscience Center, Lund Stem Cell Center, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Anselme L Perrier
- Université Paris-Saclay, CEA, CNRS, Laboratoire des Maladies Neurodégénératives: mécanismes, thérapies, imagerie, Fontenay-aux-Roses, France; Université Paris-Saclay, CEA, Molecular Imaging Research Center, Fontenay-aux-Roses, France
| |
Collapse
|
19
|
Zhang X, Tang B, Guo J. Parkinson's disease and gut microbiota: from clinical to mechanistic and therapeutic studies. Transl Neurodegener 2023; 12:59. [PMID: 38098067 PMCID: PMC10722742 DOI: 10.1186/s40035-023-00392-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/27/2023] [Indexed: 12/17/2023] Open
Abstract
Parkinson's disease (PD) is one of the most prevalent neurodegenerative diseases. The typical symptomatology of PD includes motor symptoms; however, a range of nonmotor symptoms, such as intestinal issues, usually occur before the motor symptoms. Various microorganisms inhabiting the gastrointestinal tract can profoundly influence the physiopathology of the central nervous system through neurological, endocrine, and immune system pathways involved in the microbiota-gut-brain axis. In addition, extensive evidence suggests that the gut microbiota is strongly associated with PD. This review summarizes the latest findings on microbial changes in PD and their clinical relevance, describes the underlying mechanisms through which intestinal bacteria may mediate PD, and discusses the correlations between gut microbes and anti-PD drugs. In addition, this review outlines the status of research on microbial therapies for PD and the future directions of PD-gut microbiota research.
Collapse
Affiliation(s)
- Xuxiang Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China.
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, China.
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
20
|
Thomas R, Connolly KJ, Brekk OR, Hinrich AJ, Hastings ML, Isacson O, Hallett PJ. Viral-like TLR3 induction of cytokine networks and α-synuclein are reduced by complement C3 blockade in mouse brain. Sci Rep 2023; 13:15164. [PMID: 37704739 PMCID: PMC10499893 DOI: 10.1038/s41598-023-41240-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023] Open
Abstract
Inflammatory processes and mechanisms are of central importance in neurodegenerative diseases. In the brain, α-synucleinopathies such as Parkinson's disease (PD) and Lewy body dementia (LBD) show immune cytokine network activation and increased toll like receptor 3 (TLR3) levels for viral double-stranded RNA (dsRNA). Brain inflammatory reactions caused by TLR3 activation are also relevant to understand pathogenic cascades by viral SARS-CoV-2 infection causing post- COVID-19 brain-related syndromes. In the current study, following regional brain TLR3 activation induced by dsRNA in mice, an acute complement C3 response was seen at 2 days. A C3 splice-switching antisense oligonucleotide (ASO) that promotes the splicing of a non-productive C3 mRNA, prevented downstream cytokines, such as IL-6, and α-synuclein changes. This report is the first demonstration that α-synuclein increases occur downstream of complement C3 activation. Relevant to brain dysfunction, post-COVID-19 syndromes and pathological changes leading to PD and LBD, viral dsRNA TLR3 activation in the presence of C3 complement blockade further revealed significant interactions between complement systems, inflammatory cytokine networks and α-synuclein changes.
Collapse
Affiliation(s)
- Ria Thomas
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Kyle J Connolly
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Oeystein R Brekk
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA
| | - Anthony J Hinrich
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Michelle L Hastings
- Center for Genetic Diseases, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, 60064, USA
| | - Ole Isacson
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA.
| | - Penelope J Hallett
- Neuroregeneration Institute, McLean Hospital/Harvard Medical School, Belmont, MA, 02478, USA.
| |
Collapse
|
21
|
Krohn F, Lancini E, Ludwig M, Leiman M, Guruprasath G, Haag L, Panczyszyn J, Düzel E, Hämmerer D, Betts M. Noradrenergic neuromodulation in ageing and disease. Neurosci Biobehav Rev 2023; 152:105311. [PMID: 37437752 DOI: 10.1016/j.neubiorev.2023.105311] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/29/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023]
Abstract
The locus coeruleus (LC) is a small brainstem structure located in the lower pons and is the main source of noradrenaline (NA) in the brain. Via its phasic and tonic firing, it modulates cognition and autonomic functions and is involved in the brain's immune response. The extent of degeneration to the LC in healthy ageing remains unclear, however, noradrenergic dysfunction may contribute to the pathogenesis of Alzheimer's (AD) and Parkinson's disease (PD). Despite their differences in progression at later disease stages, the early involvement of the LC may lead to comparable behavioural symptoms such as preclinical sleep problems and neuropsychiatric symptoms as a result of AD and PD pathology. In this review, we draw attention to the mechanisms that underlie LC degeneration in ageing, AD and PD. We aim to motivate future research to investigate how early degeneration of the noradrenergic system may play a pivotal role in the pathogenesis of AD and PD which may also be relevant to other neurodegenerative diseases.
Collapse
Affiliation(s)
- F Krohn
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - E Lancini
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany.
| | - M Ludwig
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - M Leiman
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - G Guruprasath
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - L Haag
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - J Panczyszyn
- Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - E Düzel
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neuroscience, University College London, London UK-WC1E 6BT, UK; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| | - D Hämmerer
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neuroscience, University College London, London UK-WC1E 6BT, UK; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany; Department of Psychology, University of Innsbruck, A-6020 Innsbruck, Austria
| | - M Betts
- German Center for Neurodegenerative Diseases (DZNE), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research (IKND), Otto-von-Guericke University Magdeburg, Magdeburg, Germany; CBBS Center for Behavioral Brain Sciences, University of Magdeburg, Magdeburg, Germany
| |
Collapse
|
22
|
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
|
23
|
Sun YH, Wu YL, Liao BY. Phenotypic heterogeneity in human genetic diseases: ultrasensitivity-mediated threshold effects as a unifying molecular mechanism. J Biomed Sci 2023; 30:58. [PMID: 37525275 PMCID: PMC10388531 DOI: 10.1186/s12929-023-00959-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 07/26/2023] [Indexed: 08/02/2023] Open
Abstract
Phenotypic heterogeneity is very common in genetic systems and in human diseases and has important consequences for disease diagnosis and treatment. In addition to the many genetic and non-genetic (e.g., epigenetic, environmental) factors reported to account for part of the heterogeneity, we stress the importance of stochastic fluctuation and regulatory network topology in contributing to phenotypic heterogeneity. We argue that a threshold effect is a unifying principle to explain the phenomenon; that ultrasensitivity is the molecular mechanism for this threshold effect; and discuss the three conditions for phenotypic heterogeneity to occur. We suggest that threshold effects occur not only at the cellular level, but also at the organ level. We stress the importance of context-dependence and its relationship to pleiotropy and edgetic mutations. Based on this model, we provide practical strategies to study human genetic diseases. By understanding the network mechanism for ultrasensitivity and identifying the critical factor, we may manipulate the weak spot to gently nudge the system from an ultrasensitive state to a stable non-disease state. Our analysis provides a new insight into the prevention and treatment of genetic diseases.
Collapse
Affiliation(s)
- Y Henry Sun
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Zhunan, Miaoli, Taiwan.
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
| | - Yueh-Lin Wu
- Institute of Molecular and Genomic Medicine, National Health Research Institute, Zhunan, Miaoli, Taiwan
- Division of Nephrology, Department of Internal Medicine, Wei-Gong Memorial Hospital, Miaoli, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei City, Taiwan
| | - Ben-Yang Liao
- Institute of Population Health Sciences, National Health Research Institute, Zhunan, Miaoli, Taiwan
| |
Collapse
|
24
|
Müller-Nedebock AC, Dekker MCJ, Farrer MJ, Hattori N, Lim SY, Mellick GD, Rektorová I, Salama M, Schuh AFS, Stoessl AJ, Sue CM, Tan AH, Vidal RL, Klein C, Bardien S. Different pieces of the same puzzle: a multifaceted perspective on the complex biological basis of Parkinson's disease. NPJ Parkinsons Dis 2023; 9:110. [PMID: 37443150 DOI: 10.1038/s41531-023-00535-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/22/2023] [Indexed: 07/15/2023] Open
Abstract
The biological basis of the neurodegenerative movement disorder, Parkinson's disease (PD), is still unclear despite it being 'discovered' over 200 years ago in Western Medicine. Based on current PD knowledge, there are widely varying theories as to its pathobiology. The aim of this article was to explore some of these different theories by summarizing the viewpoints of laboratory and clinician scientists in the PD field, on the biological basis of the disease. To achieve this aim, we posed this question to thirteen "PD experts" from six continents (for global representation) and collated their personal opinions into this article. The views were varied, ranging from toxin exposure as a PD trigger, to LRRK2 as a potential root cause, to toxic alpha-synuclein being the most important etiological contributor. Notably, there was also growing recognition that the definition of PD as a single disease should be reconsidered, perhaps each with its own unique pathobiology and treatment regimen.
Collapse
Affiliation(s)
- Amica C Müller-Nedebock
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa
| | - Marieke C J Dekker
- Department of Internal Medicine, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Matthew J Farrer
- Norman Fixel Institute for Neurological Diseases, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Nobutaka Hattori
- Research Institute of Disease of Old Age, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
- Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama, 351-0106, Japan
| | - Shen-Yang Lim
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- The Mah Pooi Soo & Tan Chin Nam Centre for Parkinson's & Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - George D Mellick
- Griffith Institute of Drug Discovery (GRIDD), Griffith University, Brisbane, QLD, Australia
| | - Irena Rektorová
- First Department of Neurology and International Clinical Research Center, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Applied Neuroscience Research Group, CEITEC, Masaryk University, Brno, Czech Republic
| | - Mohamed Salama
- Institute of Global Health and Human Ecology (I-GHHE), The American University in Cairo (AUC), New Cairo, 11835, Egypt
- Faculty of Medicine, Mansoura University, Dakahleya, Egypt
- Atlantic Senior Fellow for Equity in Brain Health at the Global Brain Health Institute (GBHI), Trinity College Dublin (TCD), Dublin, Ireland
| | - Artur F S Schuh
- Departamento de Farmacologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Serviço de Neurologia, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - A Jon Stoessl
- Pacific Parkinson's Research Centre, Department of Medicine (Division of Neurology), Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Carolyn M Sue
- Neuroscience Research Australia; Faculty of Medicine, University of New South Wales; Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst; Department of Neurology, Prince of Wales Hospital, South Eastern Sydney Local Health District, Randwick, NSW, Australia
| | - Ai Huey Tan
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- The Mah Pooi Soo & Tan Chin Nam Centre for Parkinson's & Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Rene L Vidal
- Instituto de Neurociencia Biomédica (BNI), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Centro FONDAP de Gerociencia, Salud Mental y Metabolismo (GERO), Santiago, Chile
- Centro de Biología Integrativa, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Christine Klein
- Institute of Neurogenetics, University of Lübeck and University Hospital Schleswig-Holstein, Lübeck, Germany.
| | - Soraya Bardien
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
- South African Medical Research Council/Stellenbosch University Genomics of Brain Disorders Research Unit, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
25
|
Borghammer P. The brain-first vs. body-first model of Parkinson's disease with comparison to alternative models. J Neural Transm (Vienna) 2023; 130:737-753. [PMID: 37062013 DOI: 10.1007/s00702-023-02633-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/03/2023] [Indexed: 04/17/2023]
Abstract
The ultimate origin of Lewy body disorders, including Parkinson's disease (PD) and Dementia with Lewy bodies (DLB), is still incompletely understood. Although a large number of pathogenic mechanisms have been implicated, accumulating evidence support that aggregation and neuron-to-neuron propagation of alpha-synuclein may be the core feature of these disorders. The synuclein, origin, and connectome (SOC) disease model of Lewy body disorders was recently introduced. This model is based on the hypothesis that in the majority of patients, the first alpha-synuclein pathology arises in single location and spreads from there. The most common origin sites are the enteric nervous system and the olfactory system. The SOC model predicts that gut-first pathology leads to a clinical body-first subtype characterized by prodromal autonomic symptoms and REM sleep behavior disorder. In contrast, olfactory-first pathology leads to a brain-first subtype with fewer non-motor symptoms before diagnosis. The SOC model further predicts that body-first patients are older, more commonly develop symmetric dopaminergic degeneration, and are at increased risk of dementia-compared to brain-first patients. In this review, the SOC model is explained and compared to alternative models of the pathogenesis of Lewy body disorders, including the Braak staging system, and the Unified Staging System for Lewy Body Disorders. Postmortem evidence from brain banks and clinical imaging data of dopaminergic and cardiac sympathetic loss is reviewed. It is concluded that these datasets seem to be more compatible with the SOC model than with those alternative disease models of Lewy body disorders.
Collapse
Affiliation(s)
- Per Borghammer
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, J220, 8200, Aarhus, Denmark.
| |
Collapse
|
26
|
Brembati V, Faustini G, Longhena F, Bellucci A. Alpha synuclein post translational modifications: potential targets for Parkinson's disease therapy? Front Mol Neurosci 2023; 16:1197853. [PMID: 37305556 PMCID: PMC10248004 DOI: 10.3389/fnmol.2023.1197853] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative disorder with motor symptoms. The neuropathological alterations characterizing the brain of patients with PD include the loss of dopaminergic neurons of the nigrostriatal system and the presence of Lewy bodies (LB), intraneuronal inclusions that are mainly composed of alpha-synuclein (α-Syn) fibrils. The accumulation of α-Syn in insoluble aggregates is a main neuropathological feature in PD and in other neurodegenerative diseases, including LB dementia (LBD) and multiple system atrophy (MSA), which are therefore defined as synucleinopathies. Compelling evidence supports that α-Syn post translational modifications (PTMs) such as phosphorylation, nitration, acetylation, O-GlcNAcylation, glycation, SUMOylation, ubiquitination and C-terminal cleavage, play important roles in the modulation α-Syn aggregation, solubility, turnover and membrane binding. In particular, PTMs can impact on α-Syn conformational state, thus supporting that their modulation can in turn affect α-Syn aggregation and its ability to seed further soluble α-Syn fibrillation. This review focuses on the importance of α-Syn PTMs in PD pathophysiology but also aims at highlighting their general relevance as possible biomarkers and, more importantly, as innovative therapeutic targets for synucleinopathies. In addition, we call attention to the multiple challenges that we still need to face to enable the development of novel therapeutic approaches modulating α-Syn PTMs.
Collapse
Affiliation(s)
| | | | | | - Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
27
|
Riederer P, Nagatsu T, Youdim MBH, Wulf M, Dijkstra JM, Sian-Huelsmann J. Lewy bodies, iron, inflammation and neuromelanin: pathological aspects underlying Parkinson's disease. J Neural Transm (Vienna) 2023; 130:627-646. [PMID: 37062012 PMCID: PMC10121516 DOI: 10.1007/s00702-023-02630-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/29/2023] [Indexed: 04/17/2023]
Abstract
Since the description of some peculiar symptoms by James Parkinson in 1817, attempts have been made to define its cause or at least to enlighten the pathology of "Parkinson's disease (PD)." The vast majority of PD subtypes and most cases of sporadic PD share Lewy bodies (LBs) as a characteristic pathological hallmark. However, the processes underlying LBs generation and its causal triggers are still unknown. ɑ-Synuclein (ɑ-syn, encoded by the SNCA gene) is a major component of LBs, and SNCA missense mutations or duplications/triplications are causal for rare hereditary forms of PD. Thus, it is imperative to study ɑ-syn protein and its pathology, including oligomerization, fibril formation, aggregation, and spreading mechanisms. Furthermore, there are synergistic effects in the underlying pathogenic mechanisms of PD, and multiple factors-contributing with different ratios-appear to be causal pathological triggers and progression factors. For example, oxidative stress, reduced antioxidative capacity, mitochondrial dysfunction, and proteasomal disturbances have each been suggested to be causal for ɑ-syn fibril formation and aggregation and to contribute to neuroinflammation and neural cell death. Aging is also a major risk factor for PD. Iron, as well as neuromelanin (NM), show age-dependent increases, and iron is significantly increased in the Parkinsonian substantia nigra (SN). Iron-induced pathological mechanisms include changes of the molecular structure of ɑ-syn. However, more recent PD research demonstrates that (i) LBs are detected not only in dopaminergic neurons and glia but in various neurotransmitter systems, (ii) sympathetic nerve fibres degenerate first, and (iii) at least in "brain-first" cases dopaminergic deficiency is evident before pathology induced by iron and NM. These recent findings support that the ɑ-syn/LBs pathology as well as iron- and NM-induced pathology in "brain-first" cases are important facts of PD pathology and via their interaction potentiate the disease process in the SN. As such, multifactorial toxic processes posted on a personal genetic risk are assumed to be causal for the neurodegenerative processes underlying PD. Differences in ratios of multiple factors and their spatiotemporal development, and the fact that common triggers of PD are hard to identify, imply the existence of several phenotypical subtypes, which is supported by arguments from both the "bottom-up/dual-hit" and "brain-first" models. Therapeutic strategies are necessary to avoid single initiation triggers leading to PD.
Collapse
Affiliation(s)
- Peter Riederer
- Clinic and Policlinic for Psychiatry, Psychosomatics and Psychotherapy, University Hospital Wuerzburg, Margarete-Höppel-Platz 1, 97080, Würzburg, Germany.
- Department of Psychiatry, University of Southern Denmark Odense, J.B. Winslows Vey 18, 5000, Odense, Denmark.
| | - Toshiharu Nagatsu
- Center for Research Promotion and Support, School of Medicine, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | | | - Max Wulf
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, 44801, Bochum, Germany
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, 44801, Bochum, Germany
| | | | | |
Collapse
|
28
|
Chahine LM, Merchant K, Siderowf A, Sherer T, Tanner C, Marek K, Simuni T. Proposal for a Biologic Staging System of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2023; 13:297-309. [PMID: 37066922 DOI: 10.3233/jpd-225111] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
The Parkinson's disease (PD) research field has seen the advent of several promising biomarkers and a deeper understanding of the clinical features of the disease from the earliest stages of pathology to manifest disease. Despite progress, a biologically based PD staging system does not exist. Such staging would be a useful framework within which to model the disease, develop and validate biomarkers, guide therapeutic development, and inform clinical trials design. We propose that the presence of aggregated neuronal α-synuclein, dopaminergic neuron dysfunction/degeneration, and clinical signs and symptoms identifies a group of individuals that have Lewy body pathology, which in early stages manifests with what is now referred to as prodromal non-motor features and later stages with the manifestations of PD and related Lewy body diseases as defined by clinical diagnostic criteria. Based on the state of the field, we herein propose a definition and staging of PD based on biology. We present the biologic basis for such a staging system and review key assumptions and evidence that support the proposed approach. We identify gaps in knowledge and delineate crucial research priorities that will inform the ultimate integrated biologic staging system for PD.
Collapse
Affiliation(s)
- Lana M Chahine
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kalpana Merchant
- Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andrew Siderowf
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Todd Sherer
- The Michael J Fox Foundation for Parkinson's Research, New York, NY, USA
| | - Caroline Tanner
- Department of Neurology, Weill Institute for Neurosciences, University of San Francisco, San Francisco, CA, USA
| | | | - Tanya Simuni
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
29
|
Tokunbo OS, Arogundade TT, Abayomi TA, Lewu SF, Abayomi OA, Obembe OO, Bayo-Olugbami AA, Ilesanmi DO, Keji ST, Enaibe BU. African Walnut (Tetracarpidium conophorum) Extract upregulates Glococerebrosidase activity and circumvents Parkinsonian changes in the Hippocampus via theActivation of Heatshock Proteins. J Chem Neuroanat 2023; 130:102271. [PMID: 37019342 DOI: 10.1016/j.jchemneu.2023.102271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/01/2023] [Accepted: 04/02/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Neurodegenerative illnesses like Parkinson's and Alzheimer's are largely caused by the accumulation of aggregated proteins. Heat shock proteins (HSPs), which are molecular chaperons, have been linked with the modulation of β-glucocerebrosidase (GCase) function encoded by GBA1 and Synucleinopathies. Herein, the chaperonic properties of African walnut ethanolic extract (WNE) in manganese-induced Parkinsonian neuropathology in the hippocampus was examined. METHODOLOGY 48 adult male rats weighing 185g±10g were randomly assigned into 6 (A - F) groups (n=8) and treated orally as follows: A-PBS (1ml daily for 28 days), B-WNE (200mg/kg daily for 28 days), C- WNE (400mg/kg daily for 28 days), D-Mn (100mg/kg daily for 28 days), E-Mn plus WNE (100mg/kg Mn + 200mg/kg WNE daily concomitantly for 28 days), F-Mn plus WNE (100mg/kg Mn + 400mg/kg WNE daily concomitantly for 28 days). RESULTS Rats treated with WNE showed increased levels of HSP70 and HSP90 in comparison with the Mn-intoxicated group. GCase activity also increased significantly in animals treated with WNE. Our results further revealed the therapeutic tendencies of WNE against Mn toxicity by modulating oligomeric α-synuclein levels, redox activity, and glucose bioenergetics. Furthermore, immunohistochemical evaluation revealed reduced expression of neurofibrillary tangles, and reactive astrogliosis following WNE treatment. CONCLUSION The ethanolic extract of African Walnut induced the activation of HSPs and increased the expression of GBA1 gene in the hippocampus. Activated heat shock proteins suppressed neurodegenerative changes due to Manganese toxicity. WNE was also shown to modulate neuroinflammatory, bioenergetics and neural redox balance in Parkinson-like neuropathology. This study was limited to the use of crude walnut extract and the evaluation of non-motor cascades of Parkinson's disease.
Collapse
|
30
|
Kim J, Inbo H, Kim HS, Kim W, Jang SJ, Min K, Kim SH, Bae SH, Jeong YH, Kim B, Kim C, Schwarz SC, Schwarz J, Cho KG, Chung SS, Moon J. First Clinical Report on the Treatment of Parkinson's Disease with Fetal Midbrain Precursor Cells. Mov Disord 2023; 38:589-603. [PMID: 36692025 DOI: 10.1002/mds.29316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/14/2022] [Accepted: 12/16/2022] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Because human fetal ventral mesencephalic tissue grafts provide promising results in ameliorating Parkinson's disease-implicated motor dysfunctions, human fetal midbrain-derived dopamine neuronal precursor cells are considered good candidates for cell-based therapy for Parkinson's disease in that large quantities of cells can be supplied through a good manufacturing practice-compliant system. OBJECTIVE We conducted a prospective, phase I/IIa, dose-escalation, open-label "first-in-human" clinical trial with fetal neural precursor cells to assess their safety and therapeutic efficacy in patients with idiopathic Parkinson's disease. METHODS Fifteen patients were assigned to receive three different doses of cells (4 × 106 , 12 × 106 , and 40 × 106 cells) and completed a 12-month follow-up. The primary outcome was safety, by measuring the presence of grade 3 or higher cells according to National Cancer Institute guidelines and any contaminated cells. Secondary outcomes assessed motor and neurocognitive function, as well as the level of dopamine transporters, by positron emission tomography-computed tomography. RESULTS Although a pronation-supination and hand/arm movement performance was remarkably enhanced in all three groups (all P < 0.05), the medium- and high-dose-treated groups exhibited significant improvement in Unified Parkinson's Disease Rating Scale Part III only up to 26.16% and 40%, respectively, at 12 months after transplantation without any serious clinical complications or graft-induced dyskinesia in all patients. However, the motor improvements did not correlate with increase in the dopamine transporter on positron emission tomography images. CONCLUSIONS Our results primarily demonstrate the safety and plausible dose-dependent efficacy of human fetal midbrain-derived dopamine neuronal precursor cells for idiopathic Parkinson's disease. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Joopyoung Kim
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Han Inbo
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Hyun Sook Kim
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - WonChan Kim
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Su Jin Jang
- Department of Nuclear Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Kyunghoon Min
- Department of Rehabilitation Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Sang Heum Kim
- Department of Neuroradiology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Sang-Hun Bae
- Department of Biotechnology, CHA University, Seongnam, Republic of Korea
| | - Yun-Hwa Jeong
- Department of Biotechnology, CHA University, Seongnam, Republic of Korea
| | - Borah Kim
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Chul Kim
- Department of Biotechnology, CHA University, Seongnam, Republic of Korea
| | - Sigrid C Schwarz
- Department of Neurology, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany.,German Center for Neurodegenerative Diseases, Technical University Munich, Munich, Germany
| | - Johannes Schwarz
- Department of Neurology, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany.,Geriatric Hospital Haag, Haag, Germany
| | - Kyung Gi Cho
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Sang-Sup Chung
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Jisook Moon
- Department of Biotechnology, CHA University, Seongnam, Republic of Korea
| |
Collapse
|
31
|
Youn J, Won JH, Kim M, Kwon J, Moon SH, Kim M, Ahn JH, Mun JK, Park H, Cho JW. Extra-Basal Ganglia Brain Structures Are Related to Motor Reserve in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2023; 13:39-48. [PMID: 36565134 PMCID: PMC9912725 DOI: 10.3233/jpd-223542] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 12/04/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND The "motor reserve" is an emerging concept based on the discrepancy between the severity of parkinsonism and dopaminergic degeneration; however, the related brain structures have not yet been elucidated. OBJECTIVE We investigated brain structures relevant to the motor reserve in Parkinson's disease (PD) in this study. METHODS Patients with drug-naïve, early PD were enrolled, who then underwent dopamine transporter (DAT) scan and diffusion tensor imaging (DTI). The severity of motor symptoms was evaluated with the Unified Parkinson's Disease Rating Scale score of bradykinesia and rigidity on the more affected side and dopaminergic degeneration of DAT uptake of the more affected putamen. Individual motor reserve estimate (MRE) was evaluated based on the discrepancy between the severity of motor symptoms and dopaminergic degeneration. Using DTI and the Brainnetome atlas, brain structures correlated with MRE were identified. RESULTS We enrolled 193 patients with drug-naïve PD (mean disease duration of 15.6±13.2 months), and the MRE successfully predicted the increase of levodopa equivalent dose after two years. In the DTI analysis, fractional anisotropy values of medial, inferior frontal, and temporal lobes, limbic structures, nucleus accumbens, and thalamus were positively correlated with the MRE, while no brain structures were correlated with mean diffusivity. Additionally, degree centrality derived from the structural connectivity of the frontal and temporal lobes and limbic structures was positively correlated with the MRE. CONCLUSION Our results show empirical evidence for MR in PD and brain structures relevant to MR, particularly, the extra-basal ganglia system including the limbic and frontal structures.
Collapse
Affiliation(s)
- Jinyoung Youn
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Ji Hye Won
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
| | - Mansu Kim
- Department of Artificial Intelligence, Gwangju Institute of Science and Technology, Gwangju, Korea
| | - Junmo Kwon
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon, Korea
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
| | - Seung Hwan Moon
- Nuclear Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Minkyeong Kim
- Department of Neurology, Gyeongsang National University Hospital, Jinju, Korea
| | - Jong Hyun Ahn
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Jun Kyu Mun
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
| | - Hyunjin Park
- Center for Neuroscience Imaging Research, Institute for Basic Science, Suwon, Korea
- School of Electronic and Electrical Engineering, Sungkyunkwan University, Suwon, Korea
| | - Jin Whan Cho
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Korea
| |
Collapse
|
32
|
Abd Elghani F, Safory H, Hamza H, Savyon M, Farhoud M, Toren‐Hershoviz M, Vitic Z, Ebanks K, Shani V, Bisharat S, Shaulov L, Brodski C, Song Z, Bandopadhyay R, Engelender S. SIAH proteins regulate the degradation and intra-mitochondrial aggregation of PINK1: Implications for mitochondrial pathology in Parkinson's disease. Aging Cell 2022; 21:e13731. [PMID: 36307912 PMCID: PMC9741505 DOI: 10.1111/acel.13731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 09/17/2022] [Accepted: 10/04/2022] [Indexed: 01/25/2023] Open
Abstract
Parkinson's disease (PD) is characterized by degeneration of neurons, particularly dopaminergic neurons in the substantia nigra. PD brains show accumulation of α-synuclein in Lewy bodies and accumulation of dysfunctional mitochondria. However, the mechanisms leading to mitochondrial pathology in sporadic PD are poorly understood. PINK1 is a key for mitophagy activation and recycling of unfit mitochondria. The activation of mitophagy depends on the accumulation of uncleaved PINK1 at the outer mitochondrial membrane and activation of a cascade of protein ubiquitination at the surface of the organelle. We have now found that SIAH3, a member of the SIAH proteins but lacking ubiquitin-ligase activity, is increased in PD brains and cerebrospinal fluid and in neurons treated with α-synuclein preformed fibrils (α-SynPFF). We also observed that SIAH3 is aggregated together with PINK1 in the mitochondria of PD brains. SIAH3 directly interacts with PINK1, leading to their intra-mitochondrial aggregation in cells and neurons and triggering a cascade of toxicity with PINK1 inactivation along with mitochondrial depolarization and neuronal death. We also found that SIAH1 interacts with PINK1 and promotes ubiquitination and proteasomal degradation of PINK1. Similar to the dimerization of SIAH1/SIAH2, SIAH3 interacts with SIAH1, promoting its translocation to mitochondria and preventing its ubiquitin-ligase activity toward PINK1. Our results support the notion that the increase in SIAH3 and intra-mitochondrial aggregation of SIAH3-PINK1 may mediate α-synuclein pathology by promoting proteotoxicity and preventing the elimination of dysfunctional mitochondria. We consider it possible that PINK1 activity is decreased in sporadic PD, which impedes proper mitochondrial renewal in the disease.
Collapse
Affiliation(s)
- Fatimah Abd Elghani
- Department of Biochemistry, Rappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Hazem Safory
- Department of Biochemistry, Rappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Haya Hamza
- Department of Biochemistry, Rappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Mor Savyon
- Department of Biochemistry, Rappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Malik Farhoud
- Department of Biochemistry, Rappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Michal Toren‐Hershoviz
- Department of Biochemistry, Rappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Zagorka Vitic
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health SciencesBen‐Gurion University of the NegevBe'er ShevaIsrael
| | - Kirsten Ebanks
- Reta Lila Weston Institute of Neurological StudiesUCL Queen Square Institute of NeurologyLondonUK
| | - Vered Shani
- Department of Biochemistry, Rappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Sleman Bisharat
- Department of Biochemistry, Rappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Lihi Shaulov
- Department of Biochemistry, Rappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Claude Brodski
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health SciencesBen‐Gurion University of the NegevBe'er ShevaIsrael
| | - Zhiyin Song
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Frontier Science Center for Immunology and MetabolismWuhan UniversityWuhanChina
| | - Rina Bandopadhyay
- Reta Lila Weston Institute of Neurological StudiesUCL Queen Square Institute of NeurologyLondonUK
| | - Simone Engelender
- Department of Biochemistry, Rappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| |
Collapse
|
33
|
Du J, Zhou X, Liang Y, Zhao L, Dai C, Zhong Y, Liu H, Liu G, Mo L, Tan C, Liu X, Chen L. Levodopa responsiveness and white matter alterations in Parkinson's disease: A DTI-based study and brain network analysis: A cross-sectional study. Brain Behav 2022; 12:e2825. [PMID: 36423257 PMCID: PMC9759147 DOI: 10.1002/brb3.2825] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/24/2022] [Accepted: 11/01/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Patients with Parkinson's disease (PD) present various responsiveness to levodopa, but the cause of such differences in levodopa responsiveness is unclear. Previous studies related the damage of brain white matter (WM) to levodopa responsiveness in PD patients, but no study investigated the relationship between the structural brain network change in PD patients and their levodopa responsiveness. METHODS PD patients were recruited and evaluated using the Unified Parkinson's Disease Rating Scale (UPDRS). Each patient received a diffusion tensor imaging (DTI) scan and an acute levodopa challenge test. The improvement rate of UPDRS-III was calculated. PD patients were grouped into irresponsive group (improvement rate < 30%) and responsive group (improvement rate ≥ 30%). Tract-based spatial statistics (TBSS), deterministic tracing (DT), region of interest (ROI) analysis, and automatic fiber identification (AFQ) analyses were performed. The structural brain network was also constructed and the topological parameters were calculated. RESULTS Fifty-four PD patients were included. TBSS identified significant differences in fractional anisotropy (FA) values in the corpus callosum and other regions of the brain. DT and ROI analysis of the corpus callosum found a significant difference in FA between the two groups. Graph theory analysis showed statistical differences in global efficiency, local efficiency, and characteristic path length. CONCLUSION PD patients with poor responsiveness to levodopa had WM damage in multiple brain areas, especially the corpus callosum, which might cause disruption of information integration of the structural brain network.
Collapse
Affiliation(s)
- Juncong Du
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Xuan Zhou
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Yi Liang
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Lili Zhao
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Chengcheng Dai
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Yuke Zhong
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Hang Liu
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Guohui Liu
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Lijuan Mo
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Changhong Tan
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Xi Liu
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Lifen Chen
- Department of NeurologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| |
Collapse
|
34
|
Ryu DW, Yoo SW, Oh YS, Lee KS, Ha S, Kim JS. Comparison of disease progression between brain-predominant Parkinson's disease versus Parkinson's disease with body-involvement phenotypes. Neurobiol Dis 2022; 174:105883. [DOI: 10.1016/j.nbd.2022.105883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 11/28/2022] Open
|
35
|
Bjørklund G, Zou L, Peana M, Chasapis CT, Hangan T, Lu J, Maes M. The Role of the Thioredoxin System in Brain Diseases. Antioxidants (Basel) 2022; 11:2161. [PMID: 36358532 PMCID: PMC9686621 DOI: 10.3390/antiox11112161] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/23/2022] [Accepted: 10/28/2022] [Indexed: 08/08/2023] Open
Abstract
The thioredoxin system, consisting of thioredoxin (Trx), thioredoxin reductase (TrxR), and NADPH, plays a fundamental role in the control of antioxidant defenses, cell proliferation, redox states, and apoptosis. Aberrations in the Trx system may lead to increased oxidative stress toxicity and neurodegenerative processes. This study reviews the role of the Trx system in the pathophysiology and treatment of Alzheimer's, Parkinson's and Huntington's diseases, brain stroke, and multiple sclerosis. Trx system plays an important role in the pathophysiology of those disorders via multiple interactions through oxidative stress, apoptotic, neuro-immune, and pro-survival pathways. Multiple aberrations in Trx and TrxR systems related to other redox systems and their multiple reciprocal relationships with the neurodegenerative, neuro-inflammatory, and neuro-oxidative pathways are here analyzed. Genetic and environmental factors (nutrition, metals, and toxins) may impact the function of the Trx system, thereby contributing to neuropsychiatric disease. Aberrations in the Trx and TrxR systems could be a promising drug target to prevent and treat neurodegenerative, neuro-inflammatory, neuro-oxidative stress processes, and related brain disorders.
Collapse
Affiliation(s)
- Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Toften 24, 8610 Mo i Rana, Norway
| | - Lili Zou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China
| | - Massimiliano Peana
- Department of Chemical, Physical, Mathematical and Natural Sciences, University of Sassari, Via Vienna 2, 07100 Sassari, Italy
| | - Christos T. Chasapis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Tony Hangan
- Faculty of Medicine, Ovidius University of Constanta, 900470 Constanta, Romania
| | - Jun Lu
- School of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
36
|
Molecular and Cellular Interactions in Pathogenesis of Sporadic Parkinson Disease. Int J Mol Sci 2022; 23:ijms232113043. [PMID: 36361826 PMCID: PMC9657547 DOI: 10.3390/ijms232113043] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/16/2022] [Accepted: 10/25/2022] [Indexed: 11/23/2022] Open
Abstract
An increasing number of the population all around the world suffer from age-associated neurodegenerative diseases including Parkinson’s disease (PD). This disorder presents different signs of genetic, epigenetic and environmental origin, and molecular, cellular and intracellular dysfunction. At the molecular level, α-synuclein (αSyn) was identified as the principal molecule constituting the Lewy bodies (LB). The gut microbiota participates in the pathogenesis of PD and may contribute to the loss of dopaminergic neurons through mitochondrial dysfunction. The most important pathogenetic link is an imbalance of Ca2+ ions, which is associated with redox imbalance in the cells and increased generation of reactive oxygen species (ROS). In this review, genetic, epigenetic and environmental factors that cause these disorders and their cause-and-effect relationships are considered. As a constituent of environmental factors, the example of organophosphates (OPs) is also reviewed. The role of endothelial damage in the pathogenesis of PD is discussed, and a ‘triple hit hypothesis’ is proposed as a modification of Braak’s dual hit one. In the absence of effective therapies for neurodegenerative diseases, more and more evidence is emerging about the positive impact of nutritional structure and healthy lifestyle on the state of blood vessels and the risk of developing these diseases.
Collapse
|
37
|
Zhang D, Yao J, Ma J, Gao L, Sun J, Fang J, He H, Wu T. Connectivity of corticostriatal circuits in nonmanifesting LRRK2 G2385R and R1628P carriers. CNS Neurosci Ther 2022; 28:2024-2031. [PMID: 35934920 PMCID: PMC9627388 DOI: 10.1111/cns.13933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 07/17/2022] [Accepted: 07/21/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Neuroimaging studies have shown that the functional connectivity (FC) of corticostriatal circuits in nonmanifesting leucine-rich repeat kinase 2 (LRRK2) G2019S mutation carriers mirrors neural changes in idiopathic Parkinson's disease (PD). In contrast, neural network changes in LRRK2 G2385R and R1628P mutations are unclear. We aimed to investigate the FC of corticostriatal circuits in nonmanifesting LRRK2 G2385R and R1628P mutation carriers (NMCs). METHODS Twenty-three NMCs, 28 PD patients, and 29 nonmanifesting noncarriers (NMNCs) were recruited. LRRK2 mutation analysis was performed on all participants. Clinical evaluation included MDS-UPDRS. RESULTS When compared to NMNCs, NMCs showed significantly reduced FC between the caudate nucleus and superior frontal gyrus and cerebellum, and between the nucleus accumbens and parahippocampal gyrus, amygdala, and insula. We also found increased striatum-cortical FC in NMCs. CONCLUSIONS Although the corticostriatal circuits have characteristic changes similar to PD, the relatively intact function of the sensorimotor striatum-cortical loop may result in less possibility of developing parkinsonian motor symptoms for the NMCs. This study helps explain why LRRK2 G2385R and R1628P mutations are risk factors rather than pathogenic mutations for PD and suggests that various LRRK2 mutations have distinct effects on neural networks.
Collapse
Affiliation(s)
- Dongling Zhang
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina,China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Junye Yao
- Center for Brain Imaging Science and TechnologyCollege of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
| | - Jinghong Ma
- Department of Neurobiology, Beijing Institute of GeriatricsXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Linlin Gao
- Department of Neurobiology, Beijing Institute of GeriatricsXuanwu Hospital of Capital Medical UniversityBeijingChina
| | - Junyan Sun
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina,China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Jiliang Fang
- Department of Radiology, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Hongjian He
- Center for Brain Imaging Science and TechnologyCollege of Biomedical Engineering and Instrument ScienceZhejiang UniversityHangzhouChina
| | - Tao Wu
- Department of Neurology, Center for Movement Disorders, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina,China National Clinical Research Center for Neurological DiseasesBeijingChina,Parkinson's Disease Center, Beijing Institute for Brain DisordersCapital Medical UniversityBeijingChina
| |
Collapse
|
38
|
Sidoroff V, Bower P, Stefanova N, Fanciulli A, Stankovic I, Poewe W, Seppi K, Wenning GK, Krismer F. Disease-Modifying Therapies for Multiple System Atrophy: Where Are We in 2022? JOURNAL OF PARKINSON'S DISEASE 2022; 12:1369-1387. [PMID: 35491799 PMCID: PMC9398078 DOI: 10.3233/jpd-223183] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple system atrophy is a rapidly progressive and fatal neurodegenerative disorder. While numerous preclinical studies suggested efficacy of potentially disease modifying agents, none of those were proven to be effective in large-scale clinical trials. Three major strategies are currently pursued in preclinical and clinical studies attempting to slow down disease progression. These target α-synuclein, neuroinflammation, and restoration of neurotrophic support. This review provides a comprehensive overview on ongoing preclinical and clinical developments of disease modifying therapies. Furthermore, we will focus on potential shortcomings of previous studies that can be avoided to improve data quality in future studies of this rare disease.
Collapse
Affiliation(s)
- Victoria Sidoroff
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Pam Bower
- The Multiple System Atrophy Coalition, Inc., McLean, VA, USA
| | - Nadia Stefanova
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Iva Stankovic
- Neurology Clinic, University Clinical Center of Serbia, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Werner Poewe
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Klaus Seppi
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gregor K Wenning
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Krismer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
39
|
Tan AH, Lim SY, Lang AE. The microbiome-gut-brain axis in Parkinson disease - from basic research to the clinic. Nat Rev Neurol 2022; 18:476-495. [PMID: 35750883 DOI: 10.1038/s41582-022-00681-2] [Citation(s) in RCA: 177] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 12/12/2022]
Abstract
Evidence for a close bidirectional link between the brain and the gut has led to a paradigm shift in neurology, especially in the case of Parkinson disease (PD), in which gastrointestinal dysfunction is a prominent feature. Over the past decade, numerous high-quality preclinical and clinical publications have shed light on the highly complex relationship between the gut and the brain in PD, providing potential for the development of new biomarkers and therapeutics. With the advent of high-throughput sequencing, the role of the gut microbiome has been specifically highlighted. Here, we provide a critical review of the literature on the microbiome-gut-brain axis in PD and present perspectives that will be useful for clinical practice. We begin with an overview of the gut-brain axis in PD, including the potential roles and interrelationships of the vagus nerve, α-synuclein in the enteric nervous system, altered intestinal permeability and inflammation, and gut microbes and their metabolic activities. The sections that follow synthesize the proposed roles of gut-related factors in the development and progression of, in responses to PD treatment, and as therapeutic targets. Finally, we summarize current knowledge gaps and challenges and delineate future directions for the field.
Collapse
Affiliation(s)
- Ai Huey Tan
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia. .,Mah Pooi Soo & Tan Chin Nam Centre for Parkinson's & Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Shen Yang Lim
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Mah Pooi Soo & Tan Chin Nam Centre for Parkinson's & Related Disorders, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Anthony E Lang
- Edmond J. Safra Program in Parkinson's Disease, Toronto Western Hospital, Toronto, Ontario, Canada.,Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Lee H, Wang Z, Tillekeratne A, Lukish N, Puliyadi V, Zeger S, Gallagher M, Knierim JJ. Loss of functional heterogeneity along the CA3 transverse axis in aging. Curr Biol 2022; 32:2681-2693.e4. [PMID: 35597233 PMCID: PMC9233142 DOI: 10.1016/j.cub.2022.04.077] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 04/18/2022] [Accepted: 04/26/2022] [Indexed: 01/05/2023]
Abstract
Age-related deficits in pattern separation have been postulated to bias the output of hippocampal memory processing toward pattern completion, which can cause deficits in accurate memory retrieval. Although the CA3 region of the hippocampus is often conceptualized as a homogeneous network involved in pattern completion, growing evidence demonstrates a functional gradient in CA3 along the transverse axis, as pattern-separated outputs (dominant in the more proximal CA3) transition to pattern-completed outputs (dominant in the more distal CA3). We examined the neural representations along the CA3 transverse axis in young (Y), aged memory-unimpaired (AU), and aged memory-impaired (AI) rats when different changes were made to the environment. Functional heterogeneity in CA3 was observed in Y and AU rats when the environmental similarity was high (altered cues or altered environment shapes in the same room), with more orthogonalized representations in proximal CA3 than in distal CA3. In contrast, AI rats showed reduced orthogonalization in proximal CA3 but showed normal (i.e., generalized) representations in distal CA3, with little evidence of a functional gradient. Under experimental conditions when the environmental similarity was low (different rooms), representations in proximal and distal CA3 remapped in all rats, showing that CA3 of AI rats is able to encode distinctive representations for inputs with greater dissimilarity. These experiments support the hypotheses that the age-related bias toward hippocampal pattern completion is due to the loss in AI rats of the normal transition from pattern separation to pattern completion along the CA3 transverse axis.
Collapse
Affiliation(s)
- Heekyung Lee
- Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218,Correspondence: ;
| | - Zitong Wang
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205
| | - Arjuna Tillekeratne
- Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218
| | - Nick Lukish
- Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218
| | - Vyash Puliyadi
- Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218,Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD
| | - Scott Zeger
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21205
| | - Michela Gallagher
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD,Kavli Neuroscience Discovery Institute, Johns Hopkins University
| | - James J. Knierim
- Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218,Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD,Kavli Neuroscience Discovery Institute, Johns Hopkins University,Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205,Lead Contact,Correspondence: ;
| |
Collapse
|
41
|
Engelender S, Stefanis L, Oddo S, Bellucci A. Can We Treat Neurodegenerative Proteinopathies by Enhancing Protein Degradation? Mov Disord 2022; 37:1346-1359. [PMID: 35579450 DOI: 10.1002/mds.29058] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 12/16/2022] Open
Abstract
Neurodegenerative proteinopathies are defined as a class of neurodegenerative disorders, with either genetic or sporadic age-related onset, characterized by the pathological accumulation of aggregated protein deposits. These mainly include Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD) as well as frontotemporal lobar degeneration (FTLD). The deposition of abnormal protein aggregates in the brain of patients affected by these disorders is thought to play a causative role in neuronal loss and disease progression. On that account, the idea of improving the clearance of pathological protein aggregates has taken hold as a potential therapeutic strategy. Among the possible approaches to pursue for reducing disease protein accumulation, there is the stimulation of the main protein degradation machineries of eukaryotic cells: the ubiquitin proteasomal system (UPS) and autophagy lysosomal pathway (ALP). Of note, several clinical trials testing the efficacy of either UPS- or ALP-active compounds are currently ongoing. Here, we discuss the main gaps and controversies emerging from experimental studies and clinical trials assessing the therapeutic efficacy of modulators of either the UPS or ALP in neurodegenerative proteinopathies, to gather whether they may constitute a real gateway from these disorders. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Simone Engelender
- Department of Biochemistry, The B. Rappaport Faculty of Medicine and Institute of Medical Research, Technion-Israel Institute of Technology, Haifa, Israel
| | - Leonidas Stefanis
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,First Department of Neurology, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Salvatore Oddo
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Messina, Italy
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
42
|
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
|
43
|
Chavarría C, Ivagnes R, Souza JM. Extracellular Alpha-Synuclein: Mechanisms for Glial Cell Internalization and Activation. Biomolecules 2022; 12:655. [PMID: 35625583 PMCID: PMC9138387 DOI: 10.3390/biom12050655] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022] Open
Abstract
Alpha-synuclein (α-syn) is a small protein composed of 140 amino acids and belongs to the group of intrinsically disordered proteins. It is a soluble protein that is highly expressed in neurons and expressed at low levels in glial cells. The monomeric protein aggregation process induces the formation of oligomeric intermediates and proceeds towards fibrillar species. These α-syn conformational species have been detected in the extracellular space and mediate consequences on surrounding neurons and glial cells. In particular, higher-ordered α-syn aggregates are involved in microglial and oligodendrocyte activation, as well as in the induction of astrogliosis. These phenomena lead to mitochondrial dysfunction, reactive oxygen and nitrogen species formation, and the induction of an inflammatory response, associated with neuronal cell death. Several receptors participate in cell activation and/or in the uptake of α-syn, which can vary depending on the α-syn aggregated state and cell types. The receptors involved in this process are of outstanding relevance because they may constitute potential therapeutic targets for the treatment of PD and related synucleinopathies. This review article focuses on the mechanism associated with extracellular α-syn uptake in glial cells and the consequent glial cell activation that contributes to the neuronal death associated with synucleinopathies.
Collapse
Affiliation(s)
| | | | - José M. Souza
- Departamento de Bioquímica, Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Av. Gral. Flores 2125, 11400 Montevideo, Uruguay; (C.C.); (R.I.)
| |
Collapse
|
44
|
Redefining the hypotheses driving Parkinson's diseases research. NPJ Parkinsons Dis 2022; 8:45. [PMID: 35440633 PMCID: PMC9018840 DOI: 10.1038/s41531-022-00307-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 03/04/2022] [Indexed: 12/20/2022] Open
Abstract
Parkinson’s disease (PD) research has largely focused on the disease as a single entity centred on the development of neuronal pathology within the central nervous system. However, there is growing recognition that PD is not a single entity but instead reflects multiple diseases, in which different combinations of environmental, genetic and potential comorbid factors interact to direct individual disease trajectories. Moreover, an increasing body of recent research implicates peripheral tissues and non-neuronal cell types in the development of PD. These observations are consistent with the hypothesis that the initial causative changes for PD development need not occur in the central nervous system. Here, we discuss how the use of neuronal pathology as a shared, qualitative phenotype minimises insights into the possibility of multiple origins and aetiologies of PD. Furthermore, we discuss how considering PD as a single entity potentially impairs our understanding of the causative molecular mechanisms, approaches for patient stratification, identification of biomarkers, and the development of therapeutic approaches to PD. The clear consequence of there being distinct diseases that collectively form PD, is that there is no single biomarker or treatment for PD development or progression. We propose that diagnosis should shift away from the clinical definitions, towards biologically defined diseases that collectively form PD, to enable informative patient stratification. N-of-one type, clinical designs offer an unbiased, and agnostic approach to re-defining PD in terms of a group of many individual diseases.
Collapse
|
45
|
Faustini G, Longhena F, Masato A, Bassareo V, Frau R, Klingstedt T, Shirani H, Brembati V, Parrella E, Vezzoli M, Nilsson KPR, Pizzi M, Spillantini MG, Bubacco L, Bellucci A. Synapsin III gene silencing redeems alpha-synuclein transgenic mice from Parkinson's disease-like phenotype. Mol Ther 2022; 30:1465-1483. [PMID: 35038583 PMCID: PMC9077321 DOI: 10.1016/j.ymthe.2022.01.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/20/2021] [Accepted: 01/12/2022] [Indexed: 10/19/2022] Open
Abstract
Fibrillary aggregated α-synuclein (α-syn) deposition in Lewy bodies (LB) characterizes Parkinson's disease (PD) and is believed to trigger dopaminergic synaptic failure and a retrograde terminal-to-cell body neuronal degeneration. We described that the neuronal phosphoprotein synapsin III (Syn III) cooperates with α-syn to regulate dopamine (DA) release and can be found in the insoluble α-syn fibrils composing LB. Moreover, we showed that α-syn aggregates deposition, and the associated onset of synaptic deficits and neuronal degeneration occurring following adeno-associated viral vectors-mediated overexpression of human α-syn in the nigrostriatal system are hindered in Syn III knock out mice. This supports that Syn III facilitates α-syn aggregation. Here, in an interventional experimental design, we found that by inducing the gene silencing of Syn III in human α-syn transgenic mice at PD-like stage with advanced α-syn aggregation and overt striatal synaptic failure, we could lower α-syn aggregates and striatal fibers loss. In parallel, we observed recovery from synaptic vesicles clumping, DA release failure, and motor functions impairment. This supports that Syn III consolidates α-syn aggregates, while its downregulation enables their reduction and redeems the PD-like phenotype. Strategies targeting Syn III could thus constitute a therapeutic option for PD.
Collapse
Affiliation(s)
- Gaia Faustini
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Francesca Longhena
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Anna Masato
- Department of Biology, University of Padova, Via Ugo Bassi 58b, 35121 Padua, Italy
| | - Valentina Bassareo
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Blocco A, Cagliari, 09124 Cagliari, Italy
| | - Roberto Frau
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria Blocco A, Cagliari, 09124 Cagliari, Italy
| | - Therése Klingstedt
- Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| | - Hamid Shirani
- Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| | - Viviana Brembati
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Edoardo Parrella
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Marika Vezzoli
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - K Peter R Nilsson
- Department of Physics, Chemistry and Biology, Linköping University, 581 83 Linköping, Sweden
| | - Marina Pizzi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Maria Grazia Spillantini
- Department of Clinical Neurosciences, University of Cambridge, Clifford Albutt Building, Cambridge CB2 0AH, UK
| | - Luigi Bubacco
- Department of Biology, University of Padova, Via Ugo Bassi 58b, 35121 Padua, Italy
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123 Brescia, Italy.
| |
Collapse
|
46
|
Brain region-specific susceptibility of Lewy body pathology in synucleinopathies is governed by α-synuclein conformations. Acta Neuropathol 2022; 143:453-469. [PMID: 35141810 PMCID: PMC8960659 DOI: 10.1007/s00401-022-02406-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/12/2022] [Accepted: 02/01/2022] [Indexed: 12/29/2022]
Abstract
The protein α-synuclein, a key player in Parkinson’s disease (PD) and other synucleinopathies, exists in different physiological conformations: cytosolic unfolded aggregation-prone monomers and helical aggregation-resistant multimers. It has been shown that familial PD-associated missense mutations within the α-synuclein gene destabilize the conformer equilibrium of physiologic α-synuclein in favor of unfolded monomers. Here, we characterized the relative levels of unfolded and helical forms of cytosolic α-synuclein in post-mortem human brain tissue and showed that the equilibrium of α-synuclein conformations is destabilized in sporadic PD and DLB patients. This disturbed equilibrium is decreased in a brain region-specific manner in patient samples pointing toward a possible “prion-like” propagation of the underlying pathology and forms distinct disease-specific patterns in the two different synucleinopathies. We are also able to show that a destabilization of multimers mechanistically leads to increased levels of insoluble, pathological α-synuclein, while pharmacological stabilization of multimers leads to a “prion-like” aggregation resistance. Together, our findings suggest that these disease-specific patterns of α-synuclein multimer destabilization in sporadic PD and DLB are caused by both regional neuronal vulnerability and “prion-like” aggregation transmission enabled by the destabilization of local endogenous α-synuclein protein.
Collapse
|
47
|
Predicting Parkinson’s Disease Progression: Evaluation of Ensemble Methods in Machine Learning. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:2793361. [PMID: 35154618 PMCID: PMC8831050 DOI: 10.1155/2022/2793361] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/13/2022] [Accepted: 01/15/2022] [Indexed: 01/12/2023]
Abstract
Parkinson’s disease (PD) is a complex neurodegenerative disease. Accurate diagnosis of this disease in the early stages is crucial for its initial treatment. This paper aims to present a comparative study on the methods developed by machine learning techniques in PD diagnosis. We rely on clustering and prediction learning approaches to perform the comparative study. Specifically, we use different clustering techniques for PD data clustering and support vector regression ensembles to predict Motor-UPDRS and Total-UPDRS. The results are then compared with the other prediction learning approaches, multiple linear regression, neurofuzzy, and support vector regression techniques. The comparative study is performed on a real-world PD dataset. The prediction results of data analysis on a PD real-world dataset revealed that expectation-maximization with the aid of SVR ensembles can provide better prediction accuracy in relation to decision trees, deep belief network, neurofuzzy, and support vector regression combined with other clustering techniques in the prediction of Motor-UPDRS and Total-UPDRS.
Collapse
|
48
|
Motor and non-motor circuit disturbances in early Parkinson disease: which happens first? Nat Rev Neurosci 2022; 23:115-128. [PMID: 34907352 DOI: 10.1038/s41583-021-00542-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2021] [Indexed: 12/15/2022]
Abstract
For the last two decades, pathogenic concepts in Parkinson disease (PD) have revolved around the toxicity and spread of α-synuclein. Thus, α-synuclein would follow caudo-rostral propagation from the periphery to the central nervous system, first producing non-motor manifestations (such as constipation, sleep disorders and hyposmia), and subsequently impinging upon the mesencephalon to account for the cardinal motor features before reaching the neocortex as the disease evolves towards dementia. This model is the prevailing theory of the principal neurobiological mechanism of disease. Here, we scrutinize the temporal evolution of motor and non-motor manifestations in PD and suggest that, even though the postulated bottom-up mechanisms are likely to be involved, early involvement of the nigrostriatal system is a key and prominent pathophysiological mechanism. Upcoming studies of detailed clinical manifestations with newer neuroimaging techniques will allow us to more closely define, in vivo, the role of α-synuclein aggregates with respect to neuronal loss during the onset and progression of PD.
Collapse
|
49
|
Yan M, Xiong M, Dai L, Zhang X, Zha Y, Deng X, Yu Z, Zhang Z. Cofilin 1 promotes the pathogenicity and transmission of pathological α-synuclein in mouse models of Parkinson's disease. NPJ Parkinsons Dis 2022; 8:1. [PMID: 35013321 PMCID: PMC8748615 DOI: 10.1038/s41531-021-00272-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 12/20/2021] [Indexed: 11/09/2022] Open
Abstract
The pathological hallmark of Parkinson's disease (PD) is the presence of Lewy bodies (LBs) with aggregated α-synuclein being the major component. The abnormal α-synuclein aggregates transfer between cells, recruit endogenous α-synuclein into toxic LBs, and finally trigger neuronal injury. However, the molecular mechanisms mediating the aggregation and transmission of pathological α-synuclein remain unknown. Previously we found that cofilin 1, a member of the actin-binding protein, promotes the aggregation and pathogenicity of α-synuclein in vitro. Here we further investigated the effect of cofilin 1 in mouse models of PD. We found that the mixed fibrils composed of cofilin 1 and α-synuclein are more pathogenic to mice and more prone to propagation than pure α-synuclein fibrils. Overexpression of cofilin 1 enhances the seeding and spreading of α-synuclein aggregates, and induces PD-like behavioral impairments in mice. Together, these results illustrate the important role of cofilin 1 in the pathogenicity and transmission of α-synuclein during the onset and progression of PD.
Collapse
Affiliation(s)
- Mingmin Yan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Neurology, Hubei NO. 3 People's Hospital of Jianghan University, Wuhan, 430060, China
| | - Min Xiong
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xingyu Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yunhong Zha
- The People's Hospital of China Three Gorges University, the First People's Hospital of Yichang, Yichang, 443000, China
| | - Xiaorong Deng
- Department of Neurology, Hubei NO. 3 People's Hospital of Jianghan University, Wuhan, 430060, China
| | - Zhui Yu
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
50
|
A double-hit in vivo model of GBA viral microRNA-mediated downregulation and human alpha-synuclein overexpression demonstrates nigrostriatal degeneration. Neurobiol Dis 2022; 163:105612. [PMID: 34995756 DOI: 10.1016/j.nbd.2022.105612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/19/2021] [Accepted: 01/03/2022] [Indexed: 01/30/2023] Open
Abstract
Preclinical and clinical studies support a strong association between mutations in the GBA1 gene that encodes beta-glucocerebrosidase (GCase) (EC 3.2.1.45; glucosylceramidase beta) and Parkinson's disease (PD). Alpha-synuclein (AS), a key player in PD pathogenesis, and GBA1 mutations may independently and synergistically cause lysosomal dysfunction and thus, embody clinically well-validated targets of the neurodegenerative disease process in PD. However, in vivo models, recapitulating pathological features of PD that can be used to dissect the nature of the complex relationship between GCase and AS on the nigrostriatal axis, the region particularly vulnerable in PD, are direly needed. To address this, we implemented a bidirectional approach in mice to examine the effects of: 1) GCase overexpression (wild-type and mutant N370S GBA) on endogenous AS levels and 2) downregulation of endogenous GCase (Gba) combined with AS overexpression. Striatal delivery of viral-mediated GCase overexpression revealed minimal effects on cortical and nigrostriatal AS tissue levels and no significant effect on dopaminergic system integrity. On the other hand, microRNA (miR)-mediated Gba1 downregulation (miR Gba), combined with virus-mediated human AS overexpression (+AS), yields decreased GCase activity in the cortex, mimicking levels seen in GBA1 heterozygous carriers (30-40%), increased astrogliosis and microgliosis, decreased striatal dopamine levels (50% compared to controls) and loss of nigral dopaminergic neurons (~33%)- effects that were all reversible with miR rescue. Most importantly, the synergistic neurodegeneration of miR Gba + AS correlated with augmented AS accumulation and extracellular release in the striatum. Collectively, our results suggest that GCase downregulation alone is not sufficient to recapitulate key pathological features of PD in vivo, but its synergistic interplay with AS, via increased AS levels and extracellular release, drives nigrostriatal neurodegeneration. Furthermore, we report a novel double-hit GBA-AS model that can be used to identify putative mechanisms driving PD pathophysiology and can be subsequently used to test novel therapeutic approaches.
Collapse
|