1
|
Colijn MA, Vrijsen S, Au PYB, Abou El Asrar R, Houdou M, Van den Haute C, Sarna J, Montgomery G, Vangheluwe P. Kufor-Rakeb syndrome-associated psychosis: a novel loss-of-function ATP13A2 variant and response to antipsychotic therapy. Neurogenetics 2024; 25:405-415. [PMID: 39023817 PMCID: PMC11534834 DOI: 10.1007/s10048-024-00767-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/18/2024] [Indexed: 07/20/2024]
Abstract
Biallelic (autosomal recessive) pathogenic variants in ATP13A2 cause a form of juvenile-onset parkinsonism, termed Kufor-Rakeb syndrome. In addition to motor symptoms, a variety of other neurological and psychiatric symptoms may occur in affected individuals, including supranuclear gaze palsy and cognitive decline. Although psychotic symptoms are often reported, response to antipsychotic therapy is not well described in previous case reports/series. As such, we describe treatment response in an individual with Kufor-Rakeb syndrome-associated psychosis. His disease was caused by a homozygous novel loss-of-function ATP13A2 variant (NM_022089.4, c.1970_1975del) that was characterized in this study. Our patient exhibited a good response to quetiapine monotherapy, which he has so far tolerated well. We also reviewed the literature and summarized all previous descriptions of antipsychotic treatment response. Although its use has infrequently been described in Kufor-Rakeb syndrome, quetiapine is commonly used in other degenerative parkinsonian disorders, given its lower propensity to cause extrapyramidal symptoms. As such, quetiapine should be considered in the treatment of Kufor-Rakeb syndrome-associated psychosis when antipsychotic therapy is deemed necessary.
Collapse
Affiliation(s)
- Mark Ainsley Colijn
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada.
- Mathison Centre for Mental Health Research and Education, University of Calgary, Calgary, AB, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| | - Stephanie Vrijsen
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, B-3000, Belgium
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network , Chevy Chase, MD, 20815, USA
| | - Ping Yee Billie Au
- Department of Medical Genetics, Cumming School of Medicine, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Rania Abou El Asrar
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, B-3000, Belgium
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network , Chevy Chase, MD, 20815, USA
| | - Marine Houdou
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, B-3000, Belgium
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network , Chevy Chase, MD, 20815, USA
| | - Chris Van den Haute
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network , Chevy Chase, MD, 20815, USA
- Leuven Viral Vector Core KU Leuven, Leuven, B-3000, Belgium
- Research Group for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Leuven, B-3000, Belgium
| | - Justyna Sarna
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Greg Montgomery
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | - Peter Vangheluwe
- Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, B-3000, Belgium
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network , Chevy Chase, MD, 20815, USA
| |
Collapse
|
2
|
Sikora J, Dovero S, Kinet R, Arotcarena ML, Bohic S, Bezard E, Fernagut PO, Dehay B. Nigral ATP13A2 depletion induces Parkinson's disease-related neurodegeneration in a pilot study in non-human primates. NPJ Parkinsons Dis 2024; 10:141. [PMID: 39090150 PMCID: PMC11294619 DOI: 10.1038/s41531-024-00757-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
Lysosomal impairment is strongly implicated in Parkinson's disease (PD). Among the several PD-linked genes, the ATP13A2 gene, associated with the PARK9 locus, encodes a transmembrane lysosomal P5-type ATPase. Mutations in the ATP13A2 gene were primarily identified as the cause of Kufor-Rakeb syndrome (KRS), a juvenile-onset form of PD. Subsequently, an increasing list of several mutations has been described. These mutations result in truncation of the ATP13A2 protein, leading to a loss of function but surprisingly causing heterogeneity and variability in the clinical symptoms associated with different brain pathologies. In vitro studies show that its loss compromises lysosomal function, contributing to cell death. To understand the role of ATP13A2 dysfunction in disease, we disrupted its expression through a viral vector-based approach in nonhuman primates. Here, in this pilot study, we injected bilaterally into the substantia nigra of macaques, a lentiviral vector expressing an ATP13A2 small hairpin RNA. Animals were terminated five months later, and brains were harvested and compared with historical non-injected control brains to evaluate cerebral pathological markers known to be affected in KRS and PD. We characterised the pattern of dopaminergic loss in the striatum and the substantia nigra, the regional distribution of α-synuclein immunoreactivity in several brain structures, and its pathological status (i.e., S129 phosphorylation), the accumulation of heavy metals in nigral sections and occurrence of lysosomal dysfunction. This proof-of-concept experiment highlights the potential value of lentivirus-mediated ATP13A2 silencing to induce significant and ongoing degeneration in the nigrostriatal pathway, α-synuclein pathology, and iron accumulation in nonhuman primates.
Collapse
Affiliation(s)
- Joanna Sikora
- Univ. Bordeaux, CNRS, IMN, Bordeaux, France
- Univ. De Poitiers, INSERM, LNEC, Poitiers, France
| | | | - Rémi Kinet
- Univ. Bordeaux, CNRS, IMN, Bordeaux, France
| | | | - Sylvain Bohic
- Univ. Grenoble Alpes, Synchrotron Radiation for Biomedicine (STROBE), Grenoble, France
| | | | | | | |
Collapse
|
3
|
Amini E, Rohani M, Jalessi M, Azad Z, Valzania F, Cavallieri F, Farhadi M, Gholibeigian Z. Olfactory status in neurodegeneration with brain iron accumulation disorders. Neurol Sci 2024; 45:647-654. [PMID: 37651040 DOI: 10.1007/s10072-023-07037-8] [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: 03/03/2023] [Accepted: 08/22/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Olfactory dysfunction has been suggested as a diagnostic and discriminative biomarker in some neurodegenerative disorders. However, there are few studies regarding the olfactory status in rare diseases including neurodegeneration with brain iron accumulation (NBIA) disorders. METHODS Genetically-confirmed NBIA patients were enrolled. Neurological and cognitive examinations were conducted according to the Pantothenate Kinase-Associated Neurodegeneration-Disease Rating Scale (PKAN-DRS) and the Mini-Mental State Examination (MMSE) questionnaire, respectively. Olfaction was assessed in three domains of odor threshold (OT), odor discrimination (OD), odor identification (OI), and total sum (TDI) score by the Sniffin' Sticks test. The olfactory scores were compared to a control group and a normative data set. RESULTS Thirty-seven patients, including 22 PKAN, 6 Kufor Rakeb syndrome, 4 Mitochondrial membrane Protein-Associated Neurodegeneration (MPAN), 5 cases of other 4 subtypes, and 37 controls were enrolled. The mean PKAN-DRS score was 51.83±24.93. Sixteen patients (55.2%) had normal cognition based on MMSE. NBIA patients had significantly lower olfactory scores compared to the controls in TDI and all three subtests, and 60% of them were hyposmic according to the normative data. Including only the cognitively-normal patients, still, OI and TDI scores were significantly lower compared to the controls. The phospholipase A2-Associated Neurodegeneration (PLAN) and MPAN patients had a significantly lower OI score compared to the cognitively-matched PKAN patients. CONCLUSION Olfactory impairment as a common finding in various subtypes of NBIA disorder can potentially be considered a discriminative biomarker. Better OI in PKAN compared to PLAN and MPAN patients may be related to the different underlying pathologies.
Collapse
Affiliation(s)
- Elahe Amini
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Rohani
- Department of Neurology, The Five Senses Health Institute, Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
- Skull Base Research Center, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Jalessi
- Skull Base Research Center, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Zahra Azad
- Skull Base Research Center, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Franco Valzania
- Neurology Unit, Neuromotor & Rehabilitation Department, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Francesco Cavallieri
- Neurology Unit, Neuromotor & Rehabilitation Department, Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Mohammad Farhadi
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Gholibeigian
- Skull Base Research Center, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Croucher KM, Fleming SM. ATP13A2 (PARK9) and basal ganglia function. Front Neurol 2024; 14:1252400. [PMID: 38249738 PMCID: PMC10796451 DOI: 10.3389/fneur.2023.1252400] [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: 07/05/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024] Open
Abstract
ATP13A2 is a lysosomal protein involved in polyamine transport with loss of function mutations associated with multiple neurodegenerative conditions. These include early onset Parkinson's disease, Kufor-Rakeb Syndrome, neuronal ceroid lipofuscinosis, hereditary spastic paraplegia, and amyotrophic lateral sclerosis. While ATP13A2 mutations may result in clinical heterogeneity, the basal ganglia appear to be impacted in the majority of cases. The basal ganglia is particularly vulnerable to environmental exposures such as heavy metals, pesticides, and industrial agents which are also established risk factors for many neurodegenerative conditions. Not surprisingly then, impaired function of ATP13A2 has been linked to heavy metal toxicity including manganese, iron, and zinc. This review discusses the role of ATP13A2 in basal ganglia function and dysfunction, potential common pathological mechanisms in ATP13A2-related disorders, and how gene x environment interactions may contribute to basal ganglia dysfunction.
Collapse
Affiliation(s)
- Kristina M. Croucher
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
- Biomedical Sciences Graduate Program, Kent State University, Kent, OH, United States
| | - Sheila M. Fleming
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States
| |
Collapse
|
5
|
Wang T, Sun Y, Dettmer U. Astrocytes in Parkinson's Disease: From Role to Possible Intervention. Cells 2023; 12:2336. [PMID: 37830550 PMCID: PMC10572093 DOI: 10.3390/cells12192336] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons. While neuronal dysfunction is central to PD, astrocytes also play important roles, both positive and negative, and such roles have not yet been fully explored. This literature review serves to highlight these roles and how the properties of astrocytes can be used to increase neuron survivability. Astrocytes normally have protective functions, such as releasing neurotrophic factors, metabolizing glutamate, transferring healthy mitochondria to neurons, or maintaining the blood-brain barrier. However, in PD, astrocytes can become dysfunctional and contribute to neurotoxicity, e.g., via impaired glutamate metabolism or the release of inflammatory cytokines. Therefore, astrocytes represent a double-edged sword. Restoring healthy astrocyte function and increasing the beneficial effects of astrocytes represents a promising therapeutic approach. Strategies such as promoting neurotrophin release, preventing harmful astrocyte reactivity, or utilizing regional astrocyte diversity may help restore neuroprotection.
Collapse
Affiliation(s)
- Tianyou Wang
- Collège Jean-de-Brébeuf, 3200 Chemin de la Côte-Sainte-Catherine, Montreal, QC H3T 1C1, Canada
| | - Yingqi Sun
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK;
| | - Ulf Dettmer
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| |
Collapse
|
6
|
Nechushtai L, Frenkel D, Pinkas-Kramarski R. Autophagy in Parkinson's Disease. Biomolecules 2023; 13:1435. [PMID: 37892117 PMCID: PMC10604695 DOI: 10.3390/biom13101435] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Parkinson's disease (PD) is a devastating disease associated with accumulation of α-synuclein (α-Syn) within dopaminergic neurons, leading to neuronal death. PD is characterized by both motor and non-motor clinical symptoms. Several studies indicate that autophagy, an important intracellular degradation pathway, may be involved in different neurodegenerative diseases including PD. The autophagic process mediates the degradation of protein aggregates, damaged and unneeded proteins, and organelles, allowing their clearance, and thereby maintaining cell homeostasis. Impaired autophagy may cause the accumulation of abnormal proteins. Incomplete or impaired autophagy may explain the neurotoxic accumulation of protein aggregates in several neurodegenerative diseases including PD. Indeed, studies have suggested the contribution of impaired autophagy to α-Syn accumulation, the death of dopaminergic neurons, and neuroinflammation. In this review, we summarize the recent literature on the involvement of autophagy in PD pathogenesis.
Collapse
Affiliation(s)
| | | | - Ronit Pinkas-Kramarski
- Department of Neurobiology, School of Neurobiology, Biochemistry and Biophysics, Tel-Aviv University, Ramat-Aviv, Tel Aviv 69978, Israel; (L.N.); (D.F.)
| |
Collapse
|
7
|
Gurram S, Holla VV, Kumari R, Dhar D, Kamble N, Yadav R, Muthusamy B, Pal PK. Dystonic Opisthotonus in Kufor-Rakeb Syndrome: Expanding the Phenotypic and Genotypic Spectrum. J Mov Disord 2023; 16:343-346. [PMID: 37488066 PMCID: PMC10548071 DOI: 10.14802/jmd.23098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/10/2023] [Accepted: 07/25/2023] [Indexed: 07/26/2023] Open
Affiliation(s)
- Sandeep Gurram
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Vikram V Holla
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Riyanka Kumari
- Institute of Bioinformatics, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, India
| | - Debjyoti Dhar
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Nitish Kamble
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Ravi Yadav
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| | - Babylakshmi Muthusamy
- Institute of Bioinformatics, Bengaluru, India
- Manipal Academy of Higher Education, Manipal, India
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, India
| |
Collapse
|
8
|
Fujii T, Nagamori S, Wiriyasermkul P, Zheng S, Yago A, Shimizu T, Tabuchi Y, Okumura T, Fujii T, Takeshima H, Sakai H. Parkinson's disease-associated ATP13A2/PARK9 functions as a lysosomal H +,K +-ATPase. Nat Commun 2023; 14:2174. [PMID: 37080960 PMCID: PMC10119128 DOI: 10.1038/s41467-023-37815-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 03/31/2023] [Indexed: 04/22/2023] Open
Abstract
Mutations in the human ATP13A2 (PARK9), a lysosomal ATPase, cause Kufor-Rakeb Syndrome, an early-onset form of Parkinson's disease (PD). Here, we demonstrate that ATP13A2 functions as a lysosomal H+,K+-ATPase. The K+-dependent ATPase activity and the lysosomal K+-transport activity of ATP13A2 are inhibited by an inhibitor of sarco/endoplasmic reticulum Ca2+-ATPase, thapsigargin, and K+-competitive inhibitors of gastric H+,K+-ATPase, such as vonoprazan and SCH28080. Interestingly, these H+,K+-ATPase inhibitors cause lysosomal alkalinization and α-synuclein accumulation, which are pathological hallmarks of PD. Furthermore, PD-associated mutants of ATP13A2 show abnormal expression and function. Our results suggest that the H+/K+-transporting function of ATP13A2 contributes to acidification and α-synuclein degradation in lysosomes.
Collapse
Affiliation(s)
- Takuto Fujii
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
| | - Shushi Nagamori
- Center for SI Medical Research and Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, 105-8461, Japan
| | - Pattama Wiriyasermkul
- Center for SI Medical Research and Department of Laboratory Medicine, The Jikei University School of Medicine, Tokyo, 105-8461, Japan
| | - Shizhou Zheng
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Asaka Yago
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Takahiro Shimizu
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan
| | - Yoshiaki Tabuchi
- Division of Molecular Genetics Research, Life Science Research Center, University of Toyama, Toyama, 930-0194, Japan
| | - Tomoyuki Okumura
- Department of Surgery and Science, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Tsutomu Fujii
- Department of Surgery and Science, Faculty of Medicine, University of Toyama, Toyama, 930-0194, Japan
| | - Hiroshi Takeshima
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Hideki Sakai
- Department of Pharmaceutical Physiology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194, Japan.
| |
Collapse
|
9
|
Mu J, Xue C, Fu L, Yu Z, Nie M, Wu M, Chen X, Liu K, Bu R, Huang Y, Yang B, Han J, Jiang Q, Chan KC, Zhou R, Li H, Huang A, Wang Y, Liu Z. Conformational cycle of human polyamine transporter ATP13A2. Nat Commun 2023; 14:1978. [PMID: 37031211 PMCID: PMC10082790 DOI: 10.1038/s41467-023-37741-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 03/28/2023] [Indexed: 04/10/2023] Open
Abstract
Dysregulation of polyamine homeostasis strongly associates with human diseases. ATP13A2, which is mutated in juvenile-onset Parkinson's disease and autosomal recessive spastic paraplegia 78, is a transporter with a critical role in balancing the polyamine concentration between the lysosome and the cytosol. Here, to better understand human ATP13A2-mediated polyamine transport, we use single-particle cryo-electron microscopy to solve high-resolution structures of human ATP13A2 in six intermediate states, including the putative E2 structure for the P5 subfamily of the P-type ATPases. These structures comprise a nearly complete conformational cycle spanning the polyamine transport process and capture multiple substrate binding sites distributed along the transmembrane regions, suggesting a potential polyamine transport pathway. Integration of high-resolution structures, biochemical assays, and molecular dynamics simulations allows us to obtain a better understanding of the structural basis of how hATP13A2 transports polyamines, providing a mechanistic framework for ATP13A2-related diseases.
Collapse
Affiliation(s)
- Jianqiang Mu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Chenyang Xue
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Lei Fu
- Shanghai Institute for Advanced Study, Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, 310027, Hangzhou, China
| | - Zongjun Yu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Minhan Nie
- School of Pharmaceutical Sciences, Sun Yat-sen University, No.132 Wai Huan Dong Lu, Guangzhou Higher Education Mega Center, 510006, Guangzhou, China
| | - Mengqi Wu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Xinmeng Chen
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Kun Liu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Ruiqian Bu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Ying Huang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Baisheng Yang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Jianming Han
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Qianru Jiang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Kevin C Chan
- Shanghai Institute for Advanced Study, Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, 310027, Hangzhou, China
| | - Ruhong Zhou
- Shanghai Institute for Advanced Study, Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, 310027, Hangzhou, China
| | - Huilin Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, No.132 Wai Huan Dong Lu, Guangzhou Higher Education Mega Center, 510006, Guangzhou, China
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, Guangdong, China
| | - Ancheng Huang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Yong Wang
- Shanghai Institute for Advanced Study, Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, 310027, Hangzhou, China.
- The Provincial International Science and Technology Cooperation Base on Engineering Biology, International Campus of Zhejiang University, 314400, Haining, China.
| | - Zhongmin Liu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China.
| |
Collapse
|
10
|
Gao H, Sun H, Yan N, Zhao P, Xu H, Zheng W, Zhang X, Wang T, Guo C, Zhong M. ATP13A2 Declines Zinc-Induced Accumulation of α-Synuclein in a Parkinson’s Disease Model. Int J Mol Sci 2022; 23:ijms23148035. [PMID: 35887392 PMCID: PMC9318580 DOI: 10.3390/ijms23148035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/11/2022] [Accepted: 07/18/2022] [Indexed: 02/01/2023] Open
Abstract
Parkinson’s disease (PD) is characterized by the presence of Lewy bodies caused by α-synuclein. The imbalance of zinc homeostasis is a major cause of PD, promoting α-synuclein accumulation. ATP13A2, a transporter found in acidic vesicles, plays an important role in Zn2+ homeostasis and is highly expressed in Lewy bodies in PD-surviving neurons. ATP13A2 is involved in the transport of zinc ions in lysosomes and exosomes and inhibits the aggregation of α-synuclein. However, the potential mechanism underlying the regulation of zinc homeostasis and α-synuclein accumulation by ATP13A2 remains unexplored. We used α-synuclein-GFP transgenic mice and HEK293 α-synuclein-DsRed cell line as models. The spatial exploration behavior of mice was significantly reduced, and phosphorylation levels of α-synuclein increased upon high Zn2+ treatment. High Zn2+ also inhibited the autophagy pathway by reducing LAMP2a levels and changing the expression of LC3 and P62, by reducing mitochondrial membrane potential and increasing the expression of cytochrom C, and by activating the ERK/P38 apoptosis signaling pathway, ultimately leading to increased caspase 3 levels. These protein changes were reversed after ATP13A2 overexpression, whereas ATP13A2 knockout exacerbated α-synuclein phosphorylation levels. These results suggest that ATP13A2 may have a protective effect on Zn2+-induced abnormal aggregation of α-synuclein, lysosomal dysfunction, and apoptosis.
Collapse
Affiliation(s)
- Huiling Gao
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; (H.G.); (H.S.); (P.Z.); (T.W.); (C.G.)
| | - Hehong Sun
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; (H.G.); (H.S.); (P.Z.); (T.W.); (C.G.)
| | - Nan Yan
- School of Medical Applied Technology, Shenyang Medical College, Shenyang 110034, China;
| | - Pu Zhao
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; (H.G.); (H.S.); (P.Z.); (T.W.); (C.G.)
| | - He Xu
- Department of Anatomy, Histology and Embryology, School of Medicine, Shenzhen University, Shenzhen 518060, China;
| | - Wei Zheng
- Department of Histology and Embryology, School of Basic Medical Sciences, China Medical University, Shenyang 110122, China;
| | - Xiaoyu Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China;
| | - Tao Wang
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; (H.G.); (H.S.); (P.Z.); (T.W.); (C.G.)
| | - Chuang Guo
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; (H.G.); (H.S.); (P.Z.); (T.W.); (C.G.)
| | - Manli Zhong
- College of Life and Health Sciences, Northeastern University, Shenyang 110169, China; (H.G.); (H.S.); (P.Z.); (T.W.); (C.G.)
- Correspondence:
| |
Collapse
|
11
|
Robustness of the Krebs Cycle under Physiological Conditions and in Cancer: New Clues for Evaluating Metabolism-Modifying Drug Therapies. Biomedicines 2022; 10:biomedicines10051199. [PMID: 35625935 PMCID: PMC9138339 DOI: 10.3390/biomedicines10051199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/10/2022] Open
Abstract
The Krebs cycle in cells that contain mitochondria is necessary for both energy production and anabolic processes. In given cell/condition, the Krebs cycle is dynamic but remains at a steady state. In this article, we first aimed at comparing the properties of a closed cycle versus the same metabolism in a linear array. The main finding is that, unlike a linear metabolism, the closed cycle can reach a steady state (SS) regardless of the nature and magnitude of the disturbance. When the cycle is modeled with input and output reactions, the “open” cycle is robust and reaches a steady state but with exceptions that lead to sustained accumulation of intermediate metabolites, i.e., conditions at which no SS can be achieved. The modeling of the cycle in cancer, trying to obtain marked reductions in flux, shows that these reductions are limited and therefore the Warburg effect is moderate at most. In general, our results of modeling the cycle in different conditions and looking for the achievement, or not, of SS, suggest that the cycle may have a regulation, not yet discovered, to go from an open cycle to a closed one. Said regulation could allow for reaching the steady state, thus avoiding the unwanted effects derived from the aberrant accumulation of metabolites in the mitochondria. The information in this paper might be useful to evaluate metabolism-modifying medicines.
Collapse
|
12
|
Ruiz-Barrio I, Horta-Barba A, Illán-Gala I, Kulisevsky J, Pagonabarraga J. Genotype-Phenotype Correlation in Progressive Supranuclear Palsy Syndromes: Clinical and Radiological Similarities and Specificities. Front Neurol 2022; 13:861585. [PMID: 35557621 PMCID: PMC9087829 DOI: 10.3389/fneur.2022.861585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/23/2022] [Indexed: 11/24/2022] Open
Abstract
The progressive supranuclear palsy (PSP) syndrome encompasses different entities. PSP disease of sporadic origin is the most frequent presentation, but different genetic mutations can lead either to monogenic variants of PSP disease, or to other conditions with a different pathophysiology that eventually may result in PSP phenotype. PSP syndrome of monogenic origin is poorly understood due to the low prevalence and variable expressivity of some mutations. Through this review, we describe how early age of onset, family history of early dementia, parkinsonism, dystonia, or motor neuron disease among other clinical features, as well as some neuroimaging signatures, may be the important clues to suspect PSP syndrome of monogenic origin. In addition, a diagnostic algorithm is proposed that may be useful to guide the genetic diagnosis once there is clinical suspicion of a monogenic PSP syndrome.
Collapse
Affiliation(s)
- Iñigo Ruiz-Barrio
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Andrea Horta-Barba
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ignacio Illán-Gala
- Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Sant Pau Memory Unit, Neurology Department, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Javier Pagonabarraga
- Movement Disorders Unit, Neurology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Sant Pau Biomedical Research Institute (IIB-Sant Pau), Barcelona, Spain.,Centro de Investigación en Red - Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
13
|
Sim SI, von Bülow S, Hummer G, Park E. Structural basis of polyamine transport by human ATP13A2 (PARK9). Mol Cell 2021; 81:4635-4649.e8. [PMID: 34715013 DOI: 10.1016/j.molcel.2021.08.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/17/2021] [Accepted: 08/11/2021] [Indexed: 02/03/2023]
Abstract
Polyamines are small, organic polycations that are ubiquitous and essential to all forms of life. Currently, how polyamines are transported across membranes is not understood. Recent studies have suggested that ATP13A2 and its close homologs, collectively known as P5B-ATPases, are polyamine transporters at endo-/lysosomes. Loss-of-function mutations of ATP13A2 in humans cause hereditary early-onset Parkinson's disease. To understand the polyamine transport mechanism of ATP13A2, we determined high-resolution cryoelectron microscopy (cryo-EM) structures of human ATP13A2 in five distinct conformational intermediates, which together, represent a near-complete transport cycle of ATP13A2. The structural basis of the polyamine specificity was revealed by an endogenous polyamine molecule bound to a narrow, elongated cavity within the transmembrane domain. The structures show an atypical transport path for a water-soluble substrate, in which polyamines may exit within the cytosolic leaflet of the membrane. Our study provides important mechanistic insights into polyamine transport and a framework to understand the functions and mechanisms of P5B-ATPases.
Collapse
Affiliation(s)
- Sue Im Sim
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Sören von Bülow
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany; Institute for Biophysics, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Eunyong Park
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biosciences, University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
14
|
Gagliardi M, Procopio R, Nicoletti G, Morelli M, Brighina L, Quattrone A, Bonapace G, Malanga D, Quattrone A, Annesi G. Mutation analysis of the ATP13A2 gene in patients with PD and MSA from Italy. J Neurol Sci 2021; 430:120031. [PMID: 34695705 DOI: 10.1016/j.jns.2021.120031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/15/2021] [Accepted: 10/12/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Monica Gagliardi
- Institute for Biomedical Research and Innovation, National Research Council, Mangone, CS, Italy.
| | - Radha Procopio
- Institute of Neurology, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Giuseppe Nicoletti
- Institute of Molecular Bioimaging and Physiology, National Research Council, Section of Germaneto, Catanzaro, Italy
| | - Maurizio Morelli
- Institute of Neurology, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Laura Brighina
- Department of Neurology, Milan Center for Neuroscience, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Andrea Quattrone
- Institute of Neurology, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| | - Giuseppe Bonapace
- Department of Medical and Surgical Science, Pediatrics Unit, University Magna Graecia, Catanzaro, Italy
| | - Donatella Malanga
- Laboratory of Molecular Oncology, Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy; Interdepartmental Center of Services (CIS), Magna Graecia University, Catanzaro, Italy
| | - Aldo Quattrone
- Institute of Molecular Bioimaging and Physiology, National Research Council, Section of Germaneto, Catanzaro, Italy; Neuroscience Center, University Magna Graecia, Catanzaro, Italy
| | - Grazia Annesi
- Institute for Biomedical Research and Innovation, National Research Council, Mangone, CS, Italy
| |
Collapse
|
15
|
Riboldi GM, Frattini E, Monfrini E, Frucht SJ, Fonzo AD. A Practical Approach to Early-Onset Parkinsonism. JOURNAL OF PARKINSONS DISEASE 2021; 12:1-26. [PMID: 34569973 PMCID: PMC8842790 DOI: 10.3233/jpd-212815] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Early-onset parkinsonism (EO parkinsonism), defined as subjects with disease onset before the age of 40 or 50 years, can be the main clinical presentation of a variety of conditions that are important to differentiate. Although rarer than classical late-onset Parkinson’s disease (PD) and not infrequently overlapping with forms of juvenile onset PD, a correct diagnosis of the specific cause of EO parkinsonism is critical for offering appropriate counseling to patients, for family and work planning, and to select the most appropriate symptomatic or etiopathogenic treatments. Clinical features, radiological and laboratory findings are crucial for guiding the differential diagnosis. Here we summarize the most important conditions associated with primary and secondary EO parkinsonism. We also proposed a practical approach based on the current literature and expert opinion to help movement disorders specialists and neurologists navigate this complex and challenging landscape.
Collapse
Affiliation(s)
- Giulietta M Riboldi
- The Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, Department of Neurology, NYU Langone Health, New York, NY, USA
| | - Emanuele Frattini
- IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy.,Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation , University of Milan, Milan, Italy
| | - Edoardo Monfrini
- IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy.,Dino Ferrari Center, Neuroscience Section, Department of Pathophysiology and Transplantation , University of Milan, Milan, Italy
| | - Steven J Frucht
- The Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, Department of Neurology, NYU Langone Health, New York, NY, USA
| | - Alessio Di Fonzo
- IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neurology Unit, Milan, Italy
| |
Collapse
|
16
|
Behl T, Madaan P, Sehgal A, Singh S, Sharma N, Bhatia S, Al-Harrasi A, Chigurupati S, Alrashdi I, Bungau SG. Elucidating the Neuroprotective Role of PPARs in Parkinson's Disease: A Neoteric and Prospective Target. Int J Mol Sci 2021; 22:10161. [PMID: 34576325 PMCID: PMC8467926 DOI: 10.3390/ijms221810161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 12/13/2022] Open
Abstract
One of the utmost frequently emerging neurodegenerative diseases, Parkinson's disease (PD) must be comprehended through the forfeit of dopamine (DA)-generating nerve cells in the substantia nigra pars compacta (SN-PC). The etiology and pathogenesis underlying the emergence of PD is still obscure. However, expanding corroboration encourages the involvement of genetic and environmental factors in the etiology of PD. The destruction of numerous cellular components, namely oxidative stress, ubiquitin-proteasome system (UPS) dysfunction, autophagy-lysosome system dysfunction, neuroinflammation and programmed cell death, and mitochondrial dysfunction partake in the pathogenesis of PD. Present-day pharmacotherapy can alleviate the manifestations, but no therapy has been demonstrated to cease disease progression. Peroxisome proliferator-activated receptors (PPARs) are ligand-directed transcription factors pertaining to the class of nuclear hormone receptors (NHR), and are implicated in the modulation of mitochondrial operation, inflammation, wound healing, redox equilibrium, and metabolism of blood sugar and lipids. Numerous PPAR agonists have been recognized to safeguard nerve cells from oxidative destruction, inflammation, and programmed cell death in PD and other neurodegenerative diseases. Additionally, various investigations suggest that regular administration of PPAR-activating non-steroidal anti-inflammatory drugs (NSAIDs) (ibuprofen, indomethacin), and leukotriene receptor antagonists (montelukast) were related to the de-escalated evolution of neurodegenerative diseases. The present review elucidates the emerging evidence enlightening the neuroprotective outcomes of PPAR agonists in in vivo and in vitro models experiencing PD. Existing articles up to the present were procured through PubMed, MEDLINE, etc., utilizing specific keywords spotlighted in this review. Furthermore, the authors aim to provide insight into the neuroprotective actions of PPAR agonists by outlining the pharmacological mechanism. As a conclusion, PPAR agonists exhibit neuroprotection through modulating the expression of a group of genes implicated in cellular survival pathways, and may be a propitious target in the therapy of incapacitating neurodegenerative diseases like PD.
Collapse
Affiliation(s)
- Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (P.M.); (A.S.); (S.S.); (N.S.)
| | - Piyush Madaan
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (P.M.); (A.S.); (S.S.); (N.S.)
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (P.M.); (A.S.); (S.S.); (N.S.)
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (P.M.); (A.S.); (S.S.); (N.S.)
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (P.M.); (A.S.); (S.S.); (N.S.)
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz 616, Nizwa P.O. Box 33, Oman; (S.B.); (A.A.-H.)
- School of Health Science, University of Petroleum and Energy Studies, Dehradun 248007, India
| | - Ahmed Al-Harrasi
- Natural & Medical Sciences Research Centre, University of Nizwa, Birkat Al Mauz 616, Nizwa P.O. Box 33, Oman; (S.B.); (A.A.-H.)
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 52571, Saudi Arabia;
| | - Ibrahim Alrashdi
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne NE1 7RU, UK;
| | - Simona Gabriela Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410073 Oradea, Romania
| |
Collapse
|
17
|
Torrealba-Acosta G, Yu E, Lobo-Prada T, Ruíz-Martínez J, Gorostidi-Pagola A, Gan-Or Z, Carazo-Céspedes K, Trempe JF, Mata IF, Fornaguera-Trías J. Clinical and Genetic Analysis of Costa Rican Patients With Parkinson's Disease. Front Neurol 2021; 12:656342. [PMID: 34421783 PMCID: PMC8371686 DOI: 10.3389/fneur.2021.656342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/18/2021] [Indexed: 12/16/2022] Open
Abstract
Background: Most research in genomics of Parkinson's disease (PD) has been done in subjects of European ancestry, leading to sampling bias and leaving Latin American populations underrepresented. We sought to clinically characterize PD patients of Costa Rican origin and to sequence familial PD and atypical parkinsonism-associated genes in cases and controls. Methods: We enrolled 118 PD patients with 97 unrelated controls. Collected information included demographics, exposure to risk and protective factors, and motor and cognitive assessments. We sequenced coding and untranslated regions in familial PD and atypical parkinsonism-associated genes including GBA, SNCA, VPS35, LRRK2, GCH1, PRKN, PINK1, DJ-1, VPS13C, and ATP13A2. Results: Mean age of PD probands was 62.12 ± 13.51 years; 57.6% were male. The frequency of risk and protective factors averaged ~45%. Physical activity significantly correlated with better motor performance despite years of disease. Increased years of education were significantly associated with better cognitive function, whereas hallucinations, falls, mood disorders, and coffee consumption correlated with worse cognitive performance. We did not identify an association between tested genes and PD or any damaging homozygous or compound heterozygous variants. Rare variants in LRRK2 were nominally associated with PD; six were located between amino acids p.1620 and 1623 in the C-terminal-of-ROC (COR) domain of Lrrk2. Non-synonymous GBA variants (p.T369M, p.N370S, and p.L444P) were identified in three healthy individuals. One PD patient carried a pathogenic GCH1 variant, p.K224R. Discussion: This is the first study that describes sociodemographics, risk factors, clinical presentation, and genetics of Costa Rican patients with PD, adding information to genomics research in a Latino population.
Collapse
Affiliation(s)
- Gabriel Torrealba-Acosta
- Department of Neurology and Neurosurgery, Baylor College of Medicine, Houston, TX, United States.,Neurosciences Research Center, Universidad de Costa Rica, San José, Costa Rica
| | - Eric Yu
- Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Tanya Lobo-Prada
- Neurosciences Research Center, Universidad de Costa Rica, San José, Costa Rica.,Department of Biochemistry, Medicine School, Universidad de Costa Rica, San José, Costa Rica
| | - Javier Ruíz-Martínez
- Group of Neurodegenerative Diseases, Biodonostia Health Research Institute, San Sebastian, Spain.,CIBERNED, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain.,Movement Disorders Unit, Neurology Department, Donostialdea Integrated Health Organisation, Osakidetza Basque Health Service, San Sebastian, Spain
| | - Ana Gorostidi-Pagola
- CIBERNED, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain.,Movement Disorders Unit, Neurology Department, Donostialdea Integrated Health Organisation, Osakidetza Basque Health Service, San Sebastian, Spain.,Genomic Platform, Biodonostia Health Research Institute, San Sebastian, Spain
| | - Ziv Gan-Or
- Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Kenneth Carazo-Céspedes
- Department of Neurology, Hospital San Juan de Dios, Caja Costarricense de Seguro Social, San José, Costa Rica
| | - Jean-François Trempe
- Department of Pharmacology and Therapeutics and Centre de Recherche en Biologie Structurale, McGill University, Montreal, QC, Canada
| | - Ignacio F Mata
- Cleveland Clinic Foundation, Genomic Medicine, Lerner Research Institute, Cleveland, OH, United States
| | - Jaime Fornaguera-Trías
- Neurosciences Research Center, Universidad de Costa Rica, San José, Costa Rica.,Department of Biochemistry, Medicine School, Universidad de Costa Rica, San José, Costa Rica
| |
Collapse
|
18
|
Ganguly J, Jog M. Tauopathy and Movement Disorders-Unveiling the Chameleons and Mimics. Front Neurol 2020; 11:599384. [PMID: 33250855 PMCID: PMC7674803 DOI: 10.3389/fneur.2020.599384] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
The spectrum of tauopathy encompasses heterogenous group of neurodegenerative disorders characterized by neural or glial deposition of pathological protein tau. Clinically they can present as cognitive syndromes, movement disorders, motor neuron disease, or mixed. The heterogeneity in clinical presentation, genetic background, and underlying pathology make it difficult to classify and clinically approach tauopathy. In the literature, tauopathies are thus mostly highlighted from pathological perspective. From clinical standpoint, cognitive syndromes are often been focussed while reviewing tauopathies. However, the spectrum of tauopathy has also evolved significantly in the domain of movement disorders and has transgressed beyond the domain of primary tauopathies. Secondary tauopathies from neuroinflammation or autoimmune insults and some other "novel" tauopathies are increasingly being reported in the current literature, while some of them are geographically isolated. Because of the overlapping clinical phenotypes, it often becomes difficult for the clinician to diagnose them clinically and have to wait for the pathological confirmation by autopsy. However, each of these tauopathies has some clinical and radiological signatures those can help in clinical diagnosis and targeted genetic testing. In this review, we have exposed the heterogeneity of tauopathy from a movement disorder perspective and have provided a clinical approach to diagnose them ante mortem before confirmatory autopsy. Additionally, phenotypic variability of these disorders (chameleons) and the look-alikes (mimics) have been discussed with potential clinical pointers for each of them. The review provides a framework within which new and as yet undiscovered entities can be classified in the future.
Collapse
Affiliation(s)
| | - Mandar Jog
- Movement Disorder Centre, London Health Sciences Centre, University of Western Ontario, London, ON, Canada
| |
Collapse
|
19
|
The Emerging Role of the Lysosome in Parkinson's Disease. Cells 2020; 9:cells9112399. [PMID: 33147750 PMCID: PMC7692401 DOI: 10.3390/cells9112399] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/22/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022] Open
Abstract
Lysosomal function has a central role in maintaining neuronal homeostasis, and, accordingly, lysosomal dysfunction has been linked to neurodegeneration and particularly to Parkinson’s disease (PD). Lysosomes are the converging step where the substrates delivered by autophagy and endocytosis are degraded in order to recycle their primary components to rebuild new macromolecules. Genetic studies have revealed the important link between the lysosomal function and PD; several of the autosomal dominant and recessive genes associated with PD as well as several genetic risk factors encode for lysosomal, autophagic, and endosomal proteins. Mutations in these PD-associated genes can cause lysosomal dysfunction, and since α-synuclein degradation is mostly lysosomal-dependent, among other consequences, lysosomal impairment can affect α-synuclein turnover, contributing to increase its intracellular levels and therefore promoting its accumulation and aggregation. Recent studies have also highlighted the bidirectional link between Parkinson’s disease and lysosomal storage diseases (LSD); evidence includes the presence of α-synuclein inclusions in the brain regions of patients with LSD and the identification of several lysosomal genes involved in LSD as genetic risk factors to develop PD.
Collapse
|
20
|
Chen H, Jin YH, Xue YY, Chen YL, Chen YJ, Tao QQ, Wu ZY. Novel ATP13A2 and PINK1 variants identified in Chinese patients with Parkinson’s disease by whole-exome sequencing. Neurosci Lett 2020; 733:135075. [DOI: 10.1016/j.neulet.2020.135075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 05/17/2020] [Accepted: 05/19/2020] [Indexed: 12/15/2022]
|
21
|
Foulquier F, Legrand D. Biometals and glycosylation in humans: Congenital disorders of glycosylation shed lights into the crucial role of Golgi manganese homeostasis. Biochim Biophys Acta Gen Subj 2020; 1864:129674. [PMID: 32599014 DOI: 10.1016/j.bbagen.2020.129674] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/18/2020] [Accepted: 06/24/2020] [Indexed: 12/24/2022]
Abstract
About half of the eukaryotic proteins bind biometals that participate in their structure and functions in virtually all physiological processes, including glycosylation. After reviewing the biological roles and transport mechanisms of calcium, magnesium, manganese, zinc and cobalt acting as cofactors of the metalloproteins involved in sugar metabolism and/or glycosylation, the paper will outline the pathologies resulting from a dysregulation of these metals homeostasis and more particularly Congenital Disorders of Glycosylation (CDGs) caused by ion transporter defects. Highlighting of CDGs due to defects in SLC39A8 (ZIP8) and TMEM165, two proteins transporting manganese from the extracellular space to cytosol and from cytosol to the Golgi lumen, respectively, has emphasized the importance of manganese homeostasis for glycosylation. Based on our current knowledge of TMEM165 structure and functions, this review will draw a picture of known and putative mechanisms regulating manganese homeostasis in the secretory pathway.
Collapse
Affiliation(s)
- François Foulquier
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, Lille F-59000, France
| | - Dominique Legrand
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, Lille F-59000, France.
| |
Collapse
|
22
|
Martín-Jiménez R, Lurette O, Hebert-Chatelain E. Damage in Mitochondrial DNA Associated with Parkinson's Disease. DNA Cell Biol 2020; 39:1421-1430. [PMID: 32397749 DOI: 10.1089/dna.2020.5398] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Mitochondria are the only organelles that contain their own genetic material (mtDNA). Mitochondria are involved in several key physiological functions, including ATP production, Ca2+ homeostasis, and metabolism of neurotransmitters. Since these organelles perform crucial processes to maintain neuronal homeostasis, mitochondrial dysfunctions can lead to various neurodegenerative diseases. Several mitochondrial proteins involved in ATP production are encoded by mtDNA. Thus, any mtDNA alteration can ultimately lead to mitochondrial dysfunction and cell death. Accumulation of mutations, deletions, and rearrangements in mtDNA has been observed in animal models and patients suffering from Parkinson's disease (PD). Also, specific inherited variations associated with mtDNA genetic groups (known as mtDNA haplogroups) are associated with lower or higher risk of developing PD. Consequently, mtDNA alterations should now be considered important hallmarks of this neurodegenerative disease. This review provides an update about the role of mtDNA alterations in the physiopathology of PD.
Collapse
Affiliation(s)
- Rebeca Martín-Jiménez
- Department of Biology and Université de Moncton, Moncton, Canada
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Université de Moncton, Moncton, Canada
| | - Olivier Lurette
- Department of Biology and Université de Moncton, Moncton, Canada
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Université de Moncton, Moncton, Canada
| | - Etienne Hebert-Chatelain
- Department of Biology and Université de Moncton, Moncton, Canada
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Université de Moncton, Moncton, Canada
| |
Collapse
|
23
|
Franco R, Navarro G, Martínez-Pinilla E. Lessons on Differential Neuronal-Death-Vulnerability from Familial Cases of Parkinson's and Alzheimer's Diseases. Int J Mol Sci 2019; 20:ijms20133297. [PMID: 31277513 PMCID: PMC6651599 DOI: 10.3390/ijms20133297] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 12/19/2022] Open
Abstract
The main risk of Alzheimer’s disease (AD) and Parkinson’s disease (PD), the two most common neurodegenerative pathologies, is aging. In contrast to sporadic cases, whose symptoms appear at >60 years of age, familial PD or familial AD affects younger individuals. Finding early biological markers of these diseases as well as efficacious treatments for both symptom relief and delaying disease progression are of paramount relevance. Familial early-onset PD/AD are due to genetic factors, sometimes a single mutation in a given gene. Both diseases have neuronal loss and abnormal accumulations of specific proteins in common, but in different brain regions. Despite shared features, the mechanisms underlying the pathophysiological processes are not known. This review aims at finding, among the genetic-associated cases of PD and AD, common trends that could be of interest to discover reliable biomarkers and efficacious therapies, especially those aimed at affording neuroprotection, i.e., the prevention of neuronal death.
Collapse
Affiliation(s)
- Rafael Franco
- Chemistry School, University of Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain.
| | - Gemma Navarro
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain
- Department of Biochemistry and Physiology, Faculty of Pharmacy, University of Barcelona, 02028 Barcelona, Spain
| | - Eva Martínez-Pinilla
- Departamento de Morfología y Biología Celular, Facultad de Medicina, Universidad de Oviedo, 33006 Oviedo, Spain.
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33003 Oviedo, Asturias, Spain.
- Instituto de Salud del Principado de Asturias (ISPA), 33011 Oviedo, Asturias, Spain.
| |
Collapse
|
24
|
Selvaraj S, Piramanayagam S. Impact of gene mutation in the development of Parkinson's disease. Genes Dis 2019; 6:120-128. [PMID: 31193965 PMCID: PMC6545447 DOI: 10.1016/j.gendis.2019.01.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 01/31/2019] [Indexed: 01/09/2023] Open
Abstract
Parkinson's disease (PD) is the second most common age related neurodegenerative disorder worldwide and presents as a progressive movement disorder. Globally seven million to 10 million people have Parkinson's disease. Parkinsonism is typically sporadic in nature. Loss of dopaminergic neurons from substantia nigra pars compacta (SNpc) and the neuronal intracellular Lewy body inclusions are the major cause of PD. Gene mutation and protein aggregation play a pivotal role in the degeneration of dopamine neurons. But the actual cause of dopamine degeneration remains unknown. However, several rare familial forms of PD are associated with genetic loci, and the recognition of causal mutations has provided insight into the disease process. Yet, the molecular pathways and gene transformation that trigger neuronal susceptibility are inadequately comprehended. The discovery of a mutation in new genes has provided a basis for much of the ongoing molecular work in the PD field and testing of targeted therapeutics. Single gene mutation in a dominantly or recessively inherited gene results a great impact in the development of Parkinson's disease. In this review, we summarize the molecular genetics of PD.
Collapse
Affiliation(s)
- Suganya Selvaraj
- Computational Biology Lab, Department of Bioinformatics, Bharathiar University, Coimbatore, 641046, India
| | - Shanmughavel Piramanayagam
- Professor, Computational Biology Lab, Department of Bioinformatics, Bharathiar University, Coimbatore, 641046, India
| |
Collapse
|
25
|
Schmutz I, Jagannathan V, Bartenschlager F, Stein VM, Gruber AD, Leeb T, Katz ML. ATP13A2 missense variant in Australian Cattle Dogs with late onset neuronal ceroid lipofuscinosis. Mol Genet Metab 2019; 127:95-106. [PMID: 30956123 PMCID: PMC6548654 DOI: 10.1016/j.ymgme.2018.11.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/27/2018] [Accepted: 11/29/2018] [Indexed: 02/06/2023]
Abstract
The neuronal ceroid lipofuscinoses (NCLs) are lysosomal storage disorders characterized by progressive neurodegeneration and declines in neurological functions. Pathogenic sequence variants in at least 13 genes underlie different forms of NCL, almost all of which are recessively inherited. To date 13 sequence variants in 8 canine orthologs of human NCL genes have been found to occur in 11 dog breeds in which they result in progressive neurological disorders similar to human NCLs. Canine NCLs can serve as models for preclinical evaluation of therapeutic interventions for these disorders. In most NCLs, the onset of neurological signs occurs in childhood, but some forms have adult onsets. Among these is CLN12 disease, also known as Kufor-Rakeb syndrome, PARK9, and spastic paraplegia78. These disorders result from variants in ATP13A2 which encodes a putative transmembrane ion transporter important for lysosomal function. Three Australian Cattle Dogs (a female and two of her offspring) were identified with a progressive neurological disorder with an onset of clinical signs at approximately 6 years of age. The affected dogs exhibited clinical courses and histopathology characteristic of the NCLs. Whole genome sequence analysis of one of these dogs revealed a homozygous c.1118C > T variant in ATP13A2 that predicts a nonconservative p.(Thr373Ile) amino acid substitution. All 3 affected dogs were homozygous for this variant, which was heterozygous in 42 of 394 unaffected Australian Cattle Dogs, the remainder of which were homozygous for the c.1118C allele. The high frequency of the mutant allele in this breed suggests that further screening for this variant should identify additional homozygous dogs and indicates that it would be advisable to perform such screening prior to breeding Australian Cattle Dogs.
Collapse
Affiliation(s)
- Isabelle Schmutz
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland
| | - Vidhya Jagannathan
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland
| | - Florian Bartenschlager
- Department of Veterinary Pathology, College of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany
| | - Veronika M Stein
- Department of Clinical Veterinary Sciences, Vetsuisse Faculty, University of Bern, CH-3012 Bern, Switzerland
| | - Achim D Gruber
- Department of Veterinary Pathology, College of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany
| | - Tosso Leeb
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland
| | - Martin L Katz
- Mason Eye Institute, University of Missouri School of Medicine, Columbia, MO, USA.
| |
Collapse
|
26
|
Ysselstein D, Shulman JM, Krainc D. Emerging links between pediatric lysosomal storage diseases and adult parkinsonism. Mov Disord 2019; 34:614-624. [PMID: 30726573 PMCID: PMC6520126 DOI: 10.1002/mds.27631] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/08/2019] [Accepted: 01/15/2019] [Indexed: 01/01/2023] Open
Abstract
Lysosomal storage disorders comprise a clinically heterogeneous group of autosomal-recessive or X-linked genetic syndromes caused by disruption of lysosomal biogenesis or function resulting in accumulation of nondegraded substrates. Although lysosomal storage disorders are diagnosed predominantly in children, many show variable expressivity with clinical presentations possible later in life. Given the important role of lysosomes in neuronal homeostasis, neurological manifestations, including movement disorders, can accompany many lysosomal storage disorders. Over the last decade, evidence from genetics, clinical epidemiology, cell biology, and biochemistry have converged to implicate links between lysosomal storage disorders and adult-onset movement disorders. The strongest evidence comes from mutations in Glucocerebrosidase, which cause Gaucher's disease and are among the most common and potent risk factors for PD. However, recently, many additional lysosomal storage disorder genes have been similarly implicated, including SMPD1, ATP13A2, GALC, and others. Examination of these links can offer insight into pathogenesis of PD and guide development of new therapeutic strategies. We systematically review the emerging genetic links between lysosomal storage disorders and PD. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Daniel Ysselstein
- Department of Neurology, Ken and Ruth Davee Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Joshua M. Shulman
- Departments of Neurology, Neuroscience, and Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
- Jan and Dan Duncan Neurologic Research Institute, Texas Children’s Hospital, Houston, TX
| | - Dimitri Krainc
- Department of Neurology, Ken and Ruth Davee Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
27
|
Senkevich KA, Miliukhina IV, Pchelina SN. [The genetic predictors of cognitive impairment in Parkinson's disease]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 118:109-117. [PMID: 30251988 DOI: 10.17116/jnevro2018118081109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder that can be both sporadic and familial. A number of studies are devoted to the study of non-motor symptoms in PD today. Cognitive deficits, and especially dementia, are one of the most severe and disabling non-motor symptoms of PD. More than a quarter of patients in the early stages of PD have a moderate cognitive impairment, more than half of patients with PD develop dementia within 10 years from the date of diagnosis. Using genome-wide association studies (GWAS), a number of genes associated with cognitive impairment have been identified based on a comparison of genetic and clinical phenotypes. These genes can be divided into three groups: genes that lead to the development of PD and are inherited according to the laws of Mendel (SNCA), genes that are risk factors for PD development (GBA, MAPT) and genes associated with the development of cognitive impairment, but not with PD (COMT, APOE, BDNF). This review examines the effect of genetic variants in the above-mentioned genes on cognitive functions in patients with PD. The elucidation of the genetic basis of cognitive deficits in PD could help in choice of treatment tactics and in development of new therapeutic strategies.
Collapse
Affiliation(s)
- K A Senkevich
- Institute of Experimental Medicine, St. Petersburg, Russia; Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia; St. Petersburg Nuclear Physics Institute named by Konstantinov of NRC 'Kurchatov Institute', Gatchina, Russia
| | - I V Miliukhina
- Institute of Experimental Medicine, St. Petersburg, Russia; Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia
| | - S N Pchelina
- Institute of Experimental Medicine, St. Petersburg, Russia; Pavlov First Saint Petersburg State Medical University, St. Petersburg, Russia; St. Petersburg Nuclear Physics Institute named by Konstantinov of NRC 'Kurchatov Institute', Gatchina, Russia
| |
Collapse
|
28
|
Suleiman J, El-Hattab AW. ATP13A2-related juvenile-onset Parkinson disease. Brain Dev 2019; 41:223. [PMID: 30144971 DOI: 10.1016/j.braindev.2018.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 08/07/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Jehan Suleiman
- Division of Pediatric Neurology, Tawam Hospital, Al Ain, United Arab Emirates
| | - Ayman W El-Hattab
- Division of Clinical Genetics and Metabolic Disorders, Tawam Hospital, Al-Ain, United Arab Emirates.
| |
Collapse
|
29
|
Suleiman J, Hamwi N, El-Hattab AW. ATP13A2 novel mutations causing a rare form of juvenile-onset Parkinson disease. Brain Dev 2018; 40:824-826. [PMID: 29903538 DOI: 10.1016/j.braindev.2018.05.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/10/2018] [Accepted: 05/24/2018] [Indexed: 12/15/2022]
Abstract
Parkinson disease is a common neurodegenerative disease that typically starts around the age of 60 years; however, juvenile-onset disease can occur rarely. Although Parkinson disease is typically sporadic; in rare occasions, it can be caused by a single gene defect that is inherited in an autosomal dominant, autosomal recessive, or X-linked manner. Herein, we describe a 10-year-old child who had juvenile-onset parkinsonism with rigidity, bradykinesia, dystonia, gait disturbance, and cognitive impairment. Whole exome sequencing showed compound heterozygosity for two previously unreported novel mutations in ATP13A2 (PARK9): a paternally inherited c.1321A>T (p.I441F) and a maternally inherited c.3205G>A (p.A1069T). ATP13A2 mutations are rare cause of autosomal recessive juvenile-onset Parkinson disease. Family co-segregation study and the clinical phenotype support that p.I441F and p.A1069T are indeed disease-causing mutations.
Collapse
Affiliation(s)
- Jehan Suleiman
- Division of Neurology, Department of Pediatrics, Tawam Hospital, and Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Nadia Hamwi
- Department of Pediatrics, Tawam Hospital, Al Ain, United Arab Emirates
| | - Ayman W El-Hattab
- Division of Clinical Genetics and Metabolic Disorders, Tawam Hospital, Al Ain, United Arab Emirates.
| |
Collapse
|
30
|
The genetics of Parkinson disease. Ageing Res Rev 2018; 42:72-85. [PMID: 29288112 DOI: 10.1016/j.arr.2017.12.007] [Citation(s) in RCA: 358] [Impact Index Per Article: 51.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 12/18/2017] [Accepted: 12/20/2017] [Indexed: 12/21/2022]
Abstract
About 15% of patients with Parkinson disease (PD) have family history and 5-10% have a monogenic form of the disease with Mendelian inheritance. To date, at least 23 loci and 19 disease-causing genes for parkinsonism have been found, but many more genetic risk loci and variants for sporadic PD phenotype have been identified in various association studies. Investigating the mutated protein products has uncovered potential pathogenic pathways that provide insights into mechanisms of neurodegeneration in familial and sporadic PD. To commemorate the 200th anniversary of Parkinson's publication of An Essay on the Shaking Palsy, we provide a comprehensive and critical overview of the current clinical, neuropathological, and genetic understanding of genetic forms of PD. We also discuss advances in screening for genetic PD-related risk factors and how they impact genetic counseling and contribute to the development of potential disease-modifying therapies.
Collapse
|
31
|
Shi CH, Zhang SY, Yang ZH, Yang J, Shang DD, Mao CY, Liu H, Hou HM, Shi MM, Wu J, Xu YM. A novel RAB39B gene mutation in X-linked juvenile parkinsonism with basal ganglia calcification. Mov Disord 2017; 31:1905-1909. [PMID: 27943471 DOI: 10.1002/mds.26828] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 08/29/2016] [Accepted: 09/12/2016] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES Mutations in RAB39B have been reported as a potential cause of X-linked Parkinson's disease (PD), a rare form of familial PD. We conducted a genetic analysis on RAB39B to evaluate whether RAB39B mutations are related to PD in the Chinese population. METHODS In this study, 2 patients from an X-linked juvenile parkinsonism pedigree were clinically characterized and underwent whole-exome sequencing. A comprehensive screening for RAB39B mutations in 505 sporadic patients with PD and 510 healthy controls in a Chinese population was also performed. RESULTS A novel mutation, c. 536dupA (p.E179fsX48), in RAB39B was identified in the juvenile parkinsonism pedigree. Brain MRI and CT scans in the 2 patients revealed calcification within the bilateral globus pallidus. No other potentially disease-causing RAB39B mutations were found in sporadic PD patients and controls. CONCLUSIONS X-linked juvenile parkinsonism could be caused by a RAB39B mutation, and basal ganglia calcification may be a novel clinical feature of RAB39B-related parkinsonism. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chang-He Shi
- Department of Neurology, The First affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Shu-Yu Zhang
- Department of Neurology, The First affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhi-Hua Yang
- Department of Neurology, The First affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.,Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Yang
- Department of Neurology, The First affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Dan-Dan Shang
- Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan, China
| | - Cheng-Yuan Mao
- Department of Neurology, The First affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Hao Liu
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Hai-Man Hou
- Department of Neurology, The First affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Meng-Meng Shi
- Department of Neurology, The First affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Jun Wu
- Department of Neurology, The First affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Yu-Ming Xu
- Department of Neurology, The First affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
32
|
Bouhouche A, Tesson C, Regragui W, Rahmani M, Drouet V, Tibar H, Souirti Z, Ben El Haj R, Bouslam N, Yahyaoui M, Brice A, Benomar A, Lesage S. Mutation Analysis of Consanguineous Moroccan Patients with Parkinson's Disease Combining Microarray and Gene Panel. Front Neurol 2017; 8:567. [PMID: 29163333 PMCID: PMC5674924 DOI: 10.3389/fneur.2017.00567] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/10/2017] [Indexed: 01/20/2023] Open
Abstract
During the last two decades, 15 different genes have been reported to be responsible for the monogenic form of Parkinson’s disease (PD), representing a worldwide frequency of 5–10%. Among them, 10 genes have been associated with autosomal recessive PD, with PRKN and PINK1 being the most frequent. In a cohort of 145 unrelated Moroccan PD patients enrolled since 2013, 19 patients were born from a consanguineous marriage, of which 15 were isolated cases and 4 familial. One patient was homozygous for the common LRRK2 G2019S mutation and the 18 others who did not carry this mutation were screened for exon rearrangements in the PRKN gene using Affymetrix Cytoscan HD microarray. Two patients were determined homozygous for PRKN exon-deletions, while another patient presented with compound heterozygous inheritance (3/18, 17%). Two other patients showed a region of homozygosity covering the 1p36.12 locus and were sequenced for the candidate PINK1 gene, which revealed two homozygous point mutations: the known Q456X mutation in exon 7 and a novel L539F variation in exon 8. The 13 remaining patients were subjected to next-generation sequencing (NGS) that targeted a panel of 22 PD-causing genes and overlapping phenotypes. NGS data showed that two unrelated consanguineous patients with juvenile-onset PD (12 and 13 years) carried the same homozygous stop mutation W258X in the ATP13A2 gene, possibly resulting from a founder effect; and one patient with late onset (76 years) carried a novel heterozygous frameshift mutation in SYNJ1. Clinical analysis showed that patients with the ATP13A2 mutation developed juvenile-onset PD with a severe phenotype, whereas patients having either PRKN or PINK1 mutations displayed early-onset PD with a relatively mild phenotype. By identifying pathogenic mutations in 45% (8/18) of our consanguineous Moroccan PD series, we demonstrate that the combination of chromosomal microarray analysis and NGS is a powerful approach to pinpoint the genetic bases of autosomal recessive PD, particularly in countries with a high rate of consanguinity.
Collapse
Affiliation(s)
- Ahmed Bouhouche
- Research Team in Neurology and Neurogenetics, Faculty of Medicine and Pharmacy, Genomics Center of Human Pathologies, University Mohammed V, Rabat, Morocco
| | - Christelle Tesson
- Sorbonne Universités, UPMC Université Paris 6 UMR_S 1127, INSERM U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Wafaa Regragui
- Research Team in Neurology and Neurogenetics, Faculty of Medicine and Pharmacy, Genomics Center of Human Pathologies, University Mohammed V, Rabat, Morocco
| | - Mounia Rahmani
- Research Team in Neurology and Neurogenetics, Faculty of Medicine and Pharmacy, Genomics Center of Human Pathologies, University Mohammed V, Rabat, Morocco
| | - Valérie Drouet
- Sorbonne Universités, UPMC Université Paris 6 UMR_S 1127, INSERM U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Houyam Tibar
- Research Team in Neurology and Neurogenetics, Faculty of Medicine and Pharmacy, Genomics Center of Human Pathologies, University Mohammed V, Rabat, Morocco
| | - Zouhayr Souirti
- Clinical Neurosciences Laboratory, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Rafiqua Ben El Haj
- Research Team in Neurology and Neurogenetics, Faculty of Medicine and Pharmacy, Genomics Center of Human Pathologies, University Mohammed V, Rabat, Morocco
| | - Naima Bouslam
- Research Team in Neurology and Neurogenetics, Faculty of Medicine and Pharmacy, Genomics Center of Human Pathologies, University Mohammed V, Rabat, Morocco
| | - Mohamed Yahyaoui
- Research Team in Neurology and Neurogenetics, Faculty of Medicine and Pharmacy, Genomics Center of Human Pathologies, University Mohammed V, Rabat, Morocco
| | - Alexis Brice
- Sorbonne Universités, UPMC Université Paris 6 UMR_S 1127, INSERM U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Ali Benomar
- Research Team in Neurology and Neurogenetics, Faculty of Medicine and Pharmacy, Genomics Center of Human Pathologies, University Mohammed V, Rabat, Morocco
| | - Suzanne Lesage
- Sorbonne Universités, UPMC Université Paris 6 UMR_S 1127, INSERM U 1127, CNRS UMR 7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| |
Collapse
|
33
|
Xi J, Yang X, Zhao Q, Zheng J, An R, Tian S, Huang H, Hu F, Ning P, Xu Y. Absence of association of the Ala58Val (rs17571) CTSD gene variant with Parkinson's disease or amyotrophic lateral sclerosis in a Han Chinese population. Neurosci Lett 2017; 662:181-184. [PMID: 28917980 DOI: 10.1016/j.neulet.2017.09.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 07/24/2017] [Accepted: 09/12/2017] [Indexed: 02/05/2023]
Abstract
Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD) are neurodegenerative diseases that may share genetic risk factors. The exon variant Aal58Val (rs17571) in CTSD was recently associated with AD, leading us to examine whether it also affects risk of ALS and PD. The rs17571 variant was genotyped using the ligase detection reaction in 569 Han Chinese patients with PD, 301 patients with ALS, and healthy controls age- and gender-matched to each patient group. The frequencies of genotypes and alleles were similar between each disease group and its respective control group. Similar results were obtained when patients were stratified by gender, age at disease onset or type of symptoms at disease onset. These results suggest that the CTSD rs17571 variant may not be associated with risk of ALS or PD in Han Chinese.
Collapse
Affiliation(s)
- Jing Xi
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China
| | - Xinglong Yang
- Department of Geriatric Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunan Province, 650032, PR China.
| | - Quanzhen Zhao
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China
| | - Jinhua Zheng
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China
| | - Ran An
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China
| | - Sijia Tian
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China
| | - Hongyan Huang
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China
| | - Fayunn Hu
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China
| | - Pingping Ning
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China
| | - Yanming Xu
- Department of Neurology, West China Hospital, Sichuan University, 37 Guo Xue Xiang, Chengdu, Sichuan Province, 610041, PR China.
| |
Collapse
|
34
|
Barow E, Schneider SA, Bhatia KP, Ganos C. Oculogyric crises: Etiology, pathophysiology and therapeutic approaches. Parkinsonism Relat Disord 2017; 36:3-9. [DOI: 10.1016/j.parkreldis.2016.11.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 10/27/2016] [Accepted: 11/21/2016] [Indexed: 12/14/2022]
|
35
|
Arun S, Liu L, Donmez G. Mitochondrial Biology and Neurological Diseases. Curr Neuropharmacol 2016; 14:143-54. [PMID: 26903445 PMCID: PMC4825945 DOI: 10.2174/1570159x13666150703154541] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 01/20/2015] [Accepted: 07/02/2015] [Indexed: 01/02/2023] Open
Abstract
Mitochondria are extremely active organelles that perform a variety of roles in the cell including energy production, regulation of calcium homeostasis, apoptosis, and population maintenance through fission and fusion. Mitochondrial dysfunction in the form of oxidative stress and mutations can contribute to the pathogenesis of various neurodegenerative diseases such as Parkinson’s (PD), Alzheimer’s (AD), and Huntington’s diseases (HD). Abnormalities of Complex I function in the electron transport chain have been implicated in some neurodegenerative diseases, inhibiting ATP production and generating reactive oxygen species that can cause major damage to mitochondria Mutations in both nuclear and mitochondrial DNA can contribute to neurodegenerative disease, although the pathogenesis of these conditions tends to focus on nuclear mutations. In PD, nuclear genome mutations in the PINK1 and parkin genes have been implicated in neurodegeneration [1], while mutations in APP, PSEN1 and PSEN2 have been implicated in a variety of clinical symptoms of AD [5]. Mutant htt protein is known to cause HD [2]. Much progress has been made to determine some causes of these neurodegenerative diseases, though permanent treatments have yet to be developed. In this review, we discuss the roles of mitochondrial dysfunction in the pathogenesis of these diseases.
Collapse
Affiliation(s)
| | | | - Gizem Donmez
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave. Boston MA, 02111, USA.
| |
Collapse
|
36
|
Parkinson's Disease: The Mitochondria-Iron Link. PARKINSONS DISEASE 2016; 2016:7049108. [PMID: 27293957 PMCID: PMC4886095 DOI: 10.1155/2016/7049108] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 12/14/2022]
Abstract
Mitochondrial dysfunction, iron accumulation, and oxidative damage are conditions often found in damaged brain areas of Parkinson's disease. We propose that a causal link exists between these three events. Mitochondrial dysfunction results not only in increased reactive oxygen species production but also in decreased iron-sulfur cluster synthesis and unorthodox activation of Iron Regulatory Protein 1 (IRP1), a key regulator of cell iron homeostasis. In turn, IRP1 activation results in iron accumulation and hydroxyl radical-mediated damage. These three occurrences-mitochondrial dysfunction, iron accumulation, and oxidative damage-generate a positive feedback loop of increased iron accumulation and oxidative stress. Here, we review the evidence that points to a link between mitochondrial dysfunction and iron accumulation as early events in the development of sporadic and genetic cases of Parkinson's disease. Finally, an attempt is done to contextualize the possible relationship between mitochondria dysfunction and iron dyshomeostasis. Based on published evidence, we propose that iron chelation-by decreasing iron-associated oxidative damage and by inducing cell survival and cell-rescue pathways-is a viable therapy for retarding this cycle.
Collapse
|
37
|
Martino D, Melzi V, Franco G, Kandasamy N, Monfrini E, Di Fonzo A. Juvenile dystonia-parkinsonism syndrome caused by a novel p.S941Tfs1X ATP13A2 (PARK9) mutation. Parkinsonism Relat Disord 2015; 21:1378-80. [DOI: 10.1016/j.parkreldis.2015.09.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 09/14/2015] [Accepted: 09/18/2015] [Indexed: 10/23/2022]
|
38
|
Prashanth LK, Murugan S, Kamath V, Gupta R, Jadav R, Sreekantaswamy S, Ramprasad VL. First Report of Kufor-Rakeb Syndrome (PARK 9) from India, and a Novel Nonsense Mutation in ATP13A2 Gene. Mov Disord Clin Pract 2015; 2:326-327. [PMID: 30838237 DOI: 10.1002/mdc3.12175] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/04/2015] [Accepted: 03/11/2015] [Indexed: 12/23/2022] Open
Affiliation(s)
- L K Prashanth
- Department of Neurology Apollo Hospitals Bangalore India
| | | | - Vikram Kamath
- Department of Neurology Apollo Hospitals Bangalore India
| | | | - Rakesh Jadav
- Department of Neurology Apollo Hospitals Bangalore India
| | | | | |
Collapse
|
39
|
Fernández-Moriano C, González-Burgos E, Gómez-Serranillos MP. Mitochondria-Targeted Protective Compounds in Parkinson's and Alzheimer's Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:408927. [PMID: 26064418 PMCID: PMC4429198 DOI: 10.1155/2015/408927] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 03/25/2015] [Accepted: 03/27/2015] [Indexed: 12/21/2022]
Abstract
Mitochondria are cytoplasmic organelles that regulate both metabolic and apoptotic signaling pathways; their most highlighted functions include cellular energy generation in the form of adenosine triphosphate (ATP), regulation of cellular calcium homeostasis, balance between ROS production and detoxification, mediation of apoptosis cell death, and synthesis and metabolism of various key molecules. Consistent evidence suggests that mitochondrial failure is associated with early events in the pathogenesis of ageing-related neurodegenerative disorders including Parkinson's disease and Alzheimer's disease. Mitochondria-targeted protective compounds that prevent or minimize mitochondrial dysfunction constitute potential therapeutic strategies in the prevention and treatment of these central nervous system diseases. This paper provides an overview of the involvement of mitochondrial dysfunction in Parkinson's and Alzheimer's diseases, with particular attention to in vitro and in vivo studies on promising endogenous and exogenous mitochondria-targeted protective compounds.
Collapse
Affiliation(s)
- Carlos Fernández-Moriano
- Department of Pharmacology, Faculty of Pharmacy, University Complutense of Madrid, 28040 Madrid, Spain
| | - Elena González-Burgos
- Department of Pharmacology, Faculty of Pharmacy, University Complutense of Madrid, 28040 Madrid, Spain
| | | |
Collapse
|
40
|
Blesa J, Przedborski S. Parkinson's disease: animal models and dopaminergic cell vulnerability. Front Neuroanat 2014; 8:155. [PMID: 25565980 PMCID: PMC4266040 DOI: 10.3389/fnana.2014.00155] [Citation(s) in RCA: 353] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/27/2014] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that affects about 1.5% of the global population over 65 years of age. A hallmark feature of PD is the degeneration of the dopamine (DA) neurons in the substantia nigra pars compacta (SNc) and the consequent striatal DA deficiency. Yet, the pathogenesis of PD remains unclear. Despite tremendous growth in recent years in our knowledge of the molecular basis of PD and the molecular pathways of cell death, important questions remain, such as: (1) why are SNc cells especially vulnerable; (2) which mechanisms underlie progressive SNc cell loss; and (3) what do Lewy bodies or α-synuclein reveal about disease progression. Understanding the variable vulnerability of the dopaminergic neurons from the midbrain and the mechanisms whereby pathology becomes widespread are some of the primary objectives of research in PD. Animal models are the best tools to study the pathogenesis of PD. The identification of PD-related genes has led to the development of genetic PD models as an alternative to the classical toxin-based ones, but does the dopaminergic neuronal loss in actual animal models adequately recapitulate that of the human disease? The selection of a particular animal model is very important for the specific goals of the different experiments. In this review, we provide a summary of our current knowledge about the different in vivo models of PD that are used in relation to the vulnerability of the dopaminergic neurons in the midbrain in the pathogenesis of PD.
Collapse
Affiliation(s)
- Javier Blesa
- Department of Pathology and Cell Biology, Center for Motor Neuron Biology and Disease, College of Physicians and Surgeons, Columbia UniversityNew York, NY, USA
| | | |
Collapse
|
41
|
Li G, Zhang Z, Xia H, Yang X. Analysis of Thr12Met and Ala1144Thr mutations of the ATP13A2 gene in Parkinson's disease patients in Xinjiang Uygur and Han ethnic groups. Med Sci Monit 2014; 20:2177-82. [PMID: 25374329 PMCID: PMC4301217 DOI: 10.12659/msm.892821] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background It has been reported that the ATP13A2 gene is one of the most susceptible pathogenic genes of Parkinson’s disease (PD). PARK9 mutations are found in early-onset PD and familial PD patients. Uygur and Han PD patients in the Xinjiang area were recruited as research subjects to study the differences in the Thr12Met and Ala1144Thr loci mutations of the ATP13A2 gene in these PD populations. This study explored the mutations at the Thr12Met and Ala1144Thr gene loci of the ATP13A2 gene in Parkinson’s disease patients in the Uygur and Han populations in the Xinjiang province. Material/Methods The polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method was used to analyze the Thr12Met and Ala1144Thr mutations of the ATP13A2 gene in a case-control study of 200 age- and sex- matched Uygur and Han PD patients. Results Of the 200 PD patients were studied, 2 from the Han group had a Thr12Met mutation, but Ala1144Thr mutations were not found. Among the Uygur PD patients, no Thr12Met or Ala1144Thr mutations were found. Conclusions Thr12Met and Ala1144Thr mutations of the ATP13A2 gene are rare in the Uygur PD patients in Xinjiang. Overall, the mutation rates of Thr12Met and Ala1144Thr in the Uygur and Han PD patients in the Xinjiang region are low.
Collapse
Affiliation(s)
- Guihua Li
- Department of Neurology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Zhenzhong Zhang
- Department of Neurology, Traditional Chinese Medicine Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Huan Xia
- Department of Medicine, Third Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China (mainland)
| | - Xinling Yang
- Department of Neurology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Urumqi, Xinjiang, China (mainland)
| |
Collapse
|