51
|
Abushouk AI, Negida A, Ahmed H, Abdel-Daim MM. Neuroprotective mechanisms of plant extracts against MPTP induced neurotoxicity: Future applications in Parkinson's disease. Biomed Pharmacother 2016; 85:635-645. [PMID: 27890431 DOI: 10.1016/j.biopha.2016.11.074] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/03/2016] [Accepted: 11/16/2016] [Indexed: 12/19/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease after Alzheimer's disease, affecting about seven to 10 million patients worldwide. The major pathological features of PD are loss of dopaminergic neurons in the nigrostriatal pathway and accumulation of alpha-synuclein molecules, forming Lewy bodies. Until now, there is no effective cure for PD, and investigators are searching for neuroprotective strategies to stop or slow the disease progression. The MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) induced neurotoxicity of the nigrostriatal pathway has been used to initiate PD in animal models. Multiple experimental studies showed the ability of several plant extracts to protect against MPTP induced neurotoxicity through activation of catalase, superoxide dismutase, and glutathione reductase enzymes, which reduce the cellular concentration of free radicals, preventing intracellular Ca++ release and subsequent apoptosis signaling. Other neuroprotective mechanisms of plant extracts include promoting autophagy of alpha-synuclein molecules and exerting an antiapoptotic activity via inhibition of proteolytic poly (ADP-ribose) polymerase and preventing caspase cleavage. The variety of neuroprotective mechanisms of natural plant extracts may allow researchers to target PD progression in different pathological stages and may be through multiple pathways. Further investigations are required to translate these neuroprotective mechanisms into safe and effective treatments for PD.
Collapse
Affiliation(s)
- Abdelrahman Ibrahim Abushouk
- Faculty of Medicine, Ain Shams University, Cairo, Egypt; NovaMed Medical research Association, Cairo, Egypt; Medical Research Group of Egypt, Cairo, Egypt
| | - Ahmed Negida
- Medical Research Group of Egypt, Cairo, Egypt; Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt; Student Research Unit, Zagazig University, Zagazig, El-Sharkia, Egypt
| | - Hussien Ahmed
- Medical Research Group of Egypt, Cairo, Egypt; Faculty of Medicine, Zagazig University, Zagazig, El-Sharkia, Egypt; Student Research Unit, Zagazig University, Zagazig, El-Sharkia, Egypt
| | - Mohamed M Abdel-Daim
- Pharmacology department, Faculty of veterinary medicine, Suez Canal University, Ismailia, 41522, Egypt.
| |
Collapse
|
52
|
Hayano Y, Takasu K, Koyama Y, Yamada M, Ogawa K, Minami K, Asaki T, Kitada K, Kuwabara S, Yamashita T. Dorsal horn interneuron-derived Netrin-4 contributes to spinal sensitization in chronic pain via Unc5B. J Exp Med 2016; 213:2949-2966. [PMID: 27856613 PMCID: PMC5154943 DOI: 10.1084/jem.20160877] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 09/01/2016] [Accepted: 10/24/2016] [Indexed: 11/28/2022] Open
Abstract
Hayano et al. show that Netrin-4, which is originally identified as an axon guidance molecule, is capable of enhancing sensitivity to sensory input and can contribute to neuropathic pain. The findings provide evidence for a previously unknown pain-inducing signal from spinal cord interneurons. Because of the incomplete understanding of the molecular mechanisms that underlie chronic pain, the currently available treatments for this type of pain remain inefficient. In this study, we show that Netrin-4, a member of the axon guidance molecule family, was expressed in dorsal horn inner lamina II excitatory interneurons in the rat spinal cord. A similar expression pattern for Netrin-4 was also observed in human spinal cord. Behavioral analysis revealed that tactile and heat hyperalgesia after peripheral nerve injury or inflammation were abolished in Netrin-4–mutant rats. Transient suppression of Netrin-4 or its receptor Unc5B after injury could also prevent allodynia. Conversely, intrathecal administration of Netrin-4 protein to naive rats enhanced excitatory synaptic transmission in the dorsal horn and induced allodynia, suggesting that Netrin-4 is involved in spinal sensitization. Furthermore, the Unc5B receptor and subsequent activation of the tyrosine phosphatase SHP2 mediated Netrin-4–induced pain signaling in the spinal cord. These results identify Netrin-4 as a novel protein regulating spinal sensitization leading to chronic pain. Our findings provide evidence for the function of Netrin in the adult nervous system, as well as a previously unknown function in inducing pain signals from dorsal horn interneurons.
Collapse
Affiliation(s)
- Yasufumi Hayano
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Keiko Takasu
- Pain and Neuroscience, Discovery Research Laboratory for Core Therapeutic Areas, Shionogi & Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| | - Yoshihisa Koyama
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Moe Yamada
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - Koichi Ogawa
- Pain and Neuroscience, Discovery Research Laboratory for Core Therapeutic Areas, Shionogi & Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| | - Kazuhisa Minami
- Pain and Neuroscience, Discovery Research Laboratory for Core Therapeutic Areas, Shionogi & Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| | - Toshiyuki Asaki
- Pain and Neuroscience, Discovery Research Laboratory for Core Therapeutic Areas, Shionogi & Co., Ltd., Toyonaka, Osaka 561-0825, Japan
| | - Kazuhiro Kitada
- Department of Biological Sciences, Graduate School of Science, Hokkaido University, Kita-ku, Sapporo 060-0810, Japan
| | - Satoshi Kuwabara
- Department of Neurology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba 260-8670, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
53
|
Zhang M, Mu H, Shang Z, Kang K, Lv H, Duan L, Li J, Chen X, Teng Y, Jiang Y, Zhang R. Genome-wide pathway-based association analysis identifies risk pathways associated with Parkinson's disease. Neuroscience 2016; 340:398-410. [PMID: 27840232 DOI: 10.1016/j.neuroscience.2016.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 11/03/2016] [Accepted: 11/03/2016] [Indexed: 01/02/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease. It is generally believed that it is influenced by both genetic and environmental factors, but the precise pathogenesis of PD is unknown to date. In this study, we performed a pathway analysis based on genome-wide association study (GWAS) to detect risk pathways of PD in three GWAS datasets. We first mapped all SNP markers to autosomal genes in each GWAS dataset. Then, we evaluated gene risk values using the minimum P-value of the tagSNPs. We took a pathway as a unit to identify the risk pathways based on the cumulative risks of the genes in the pathway. Finally, we combine the analysis results of the three datasets to detect the high risk pathways associated with PD. We found there were five same pathways in the three datasets. Besides, we also found there were five pathways which were shared in two datasets. Most of these pathways are associated with nervoussystem. Five pathways had been reported to be PD-related pathways in the previous literature. Our findings also implied that there was a close association between immune response and PD. Continued investigation of these pathways will further help us explain the pathogenesis of PD.
Collapse
Affiliation(s)
- Mingming Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hongbo Mu
- College of Science, Northeast Forestry University, Harbin, China
| | - Zhenwei Shang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Kai Kang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, China
| | - Hongchao Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Lian Duan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jin Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xinren Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanbo Teng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongshuai Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China.
| | - Ruijie Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China.
| |
Collapse
|
54
|
Lill CM. Genetics of Parkinson's disease. Mol Cell Probes 2016; 30:386-396. [PMID: 27818248 DOI: 10.1016/j.mcp.2016.11.001] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 11/02/2016] [Accepted: 11/02/2016] [Indexed: 12/30/2022]
Abstract
Almost two decades after the identification of SNCA as the first causative gene in Parkinson's disease (PD) and the subsequent understanding that genetic factors play a substantial role in PD development, our knowledge of the genetic architecture underlying this disease has vastly improved. Approximately 5-10% of patients suffer from a monogenic form of PD where autosomal dominant mutations in SNCA, LRRK2, and VPS35 and autosomal recessive mutations in PINK1, DJ-1, and Parkin cause the disease with high penetrance. Furthermore, recent whole-exome sequencing have described autosomal recessive DNAJC6 mutations in predominately atypical, but also cases with typical PD. In addition, several other genes have been linked to atypical Parkinsonian phenotypes. However, the vast majority of PD is genetically complex, i.e. it is caused by the combined action of common genetic variants in concert with environmental factors. By the application of genome-wide association studies, 26 PD risk loci have been established to date. Similar to other genetically complex diseases, these show only moderate effects on PD risk. Increasing this etiologic complexity, many of the involved genetic and environmental risk factors likely interact in an intricate fashion. This article aims to provide a comprehensive overview of the current knowledge in PD genetics.
Collapse
Affiliation(s)
- Christina M Lill
- Genetic and Molecular Epidemiology Group, Institute of Neurogenetics, University of Lübeck, Maria-Goeppert-Str. 1, 23562, Lübeck, Germany.
| |
Collapse
|
55
|
Roppongi RT, Karimi B, Siddiqui TJ. Role of LRRTMs in synapse development and plasticity. Neurosci Res 2016; 116:18-28. [PMID: 27810425 DOI: 10.1016/j.neures.2016.10.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/10/2016] [Accepted: 10/14/2016] [Indexed: 12/19/2022]
Abstract
Leucine-rich-repeat transmembrane neuronal proteins (LRRTMs) are a family of four synapse organizing proteins critical for the development and function of excitatory synapses. The genes encoding LRRTMs and their binding partners, neurexins and HSPGs, are strongly associated with multiple psychiatric disorders. Here, we review the literature covering their structural features, expression patterns in the developing and adult brains, evolutionary origins, and discovery as synaptogenic proteins. We also discuss their role in the development and plasticity of excitatory synapses as well as their disease associations.
Collapse
Affiliation(s)
- Reiko T Roppongi
- Department of Physiology and Pathophysiology, College of Medicine, University of Manitoba, Winnipeg, MB, Canada; Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, 710 William Avenue, Winnipeg R3Y 0Z3, MB, Canada
| | - Benyamin Karimi
- Department of Physiology and Pathophysiology, College of Medicine, University of Manitoba, Winnipeg, MB, Canada; Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, 710 William Avenue, Winnipeg R3Y 0Z3, MB, Canada
| | - Tabrez J Siddiqui
- Department of Physiology and Pathophysiology, College of Medicine, University of Manitoba, Winnipeg, MB, Canada; Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, 710 William Avenue, Winnipeg R3Y 0Z3, MB, Canada.
| |
Collapse
|
56
|
Yang X, Zhao Q, An R, Zhou H, Lin Z, Xu Y. SNP rs1805874 of the Calbindin1 Gene Is Associated with Parkinson's Disease in Han Chinese. Genet Test Mol Biomarkers 2016; 20:753-757. [PMID: 27611799 DOI: 10.1089/gtmb.2016.0149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
AIMS The single-nucleotide polymorphism, rs1805874, in the Calbindin1 gene has been associated with Parkinson's disease among the Japanese people, but not among Europeans or Americans. To help clarify these contrasting results, we conducted a case-control study to explore whether such an association exists among the Han Chinese. METHODS We used the ligase detection reaction to genotype the rs1805874 SNP in 514 Han Chinese with Parkinson's disease as well as in 475 healthy Han Chinese controls. RESULTS We identified a significant association between the A allele of rs1805874 and the risk of Parkinson's disease (OR 1.257, 95% CI 1.036-1.524, p = 0.020). Subgroup analysis revealed a significant association of the A allele with male gender (OR 1.306, 95% CI 1.009-1.691, p = 0.042) and late onset of disease (OR 1.247, 95% CI 1.010-1.540, p = 0.040). CONCLUSION Our findings suggest that the A allele of rs1805874 is associated with risk of Parkinson's disease among the Han Chinese. Our results, combined with previous studies, suggest that rs1805874 is associated with Parkinson's disease in East Asians, but not Caucasians.
Collapse
Affiliation(s)
- Xinglong Yang
- Department of Neurology, West China Hospital, Sichuan University , Chengdu, People's Republic of China
| | - Quanzhen Zhao
- Department of Neurology, West China Hospital, Sichuan University , Chengdu, People's Republic of China
| | - Ran An
- Department of Neurology, West China Hospital, Sichuan University , Chengdu, People's Republic of China
| | - Huayong Zhou
- Department of Neurology, West China Hospital, Sichuan University , Chengdu, People's Republic of China
| | - Zhenfang Lin
- Department of Neurology, West China Hospital, Sichuan University , Chengdu, People's Republic of China
| | - Yanming Xu
- Department of Neurology, West China Hospital, Sichuan University , Chengdu, People's Republic of China
| |
Collapse
|
57
|
Analysis of LRRK2, SNCA, and ITGA8 Gene Variants with Sporadic Parkinson's Disease Susceptibility in Chinese Han Population. PARKINSONS DISEASE 2016; 2016:3474751. [PMID: 27668119 PMCID: PMC5030423 DOI: 10.1155/2016/3474751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 07/01/2016] [Accepted: 08/08/2016] [Indexed: 11/17/2022]
Abstract
Background. Parkinson's disease (PD) is an age-related neurodegenerative disease affected by multiple genetic and environmental factors. We performed a case-control study on candidate gene to scrutinize whether genetic variants in LRRK2, SNCA, and ITGA8 genes could be associated with sporadic PD in Chinese Han population. Methods. Five single-nucleotide polymorphisms (SNPs) of LRRK2 (rs1491942), SNCA (rs2301134, rs2301135, and rs356221), and ITGA8 (rs7077361) were selected and genotyped among 583 unrelated PD patients and 558 healthy controls. Results. Rs1491942 of LRRK2 gene had a significantly higher genotype frequency (P = 3.543E − 09) and allelic G/C frequencies (P = 2.601E − 10) in PD patients than controls. Rs2301135 of SNCA gene also showed an obvious difference in genotype frequency (P = 4.394E − 07) and allelic G/C frequencies (P = 9.116E − 13) between PD patients and controls. SNPs rs2301134 and rs356221 of SNCA gene and rs7077361 of ITGA8 gene lacked the significant association with the susceptibility of PD in Chinese Han population. Conclusions. Our study firstly expresses that rs1491942 of LRRK2 and rs2301135 of SNCA gene are substantially associated with sporadic Parkinson's disease in Chinese Han population.
Collapse
|
58
|
Biernacka JM, Chung SJ, Armasu SM, Anderson KS, Lill CM, Bertram L, Ahlskog JE, Brighina L, Frigerio R, Maraganore DM. Genome-wide gene-environment interaction analysis of pesticide exposure and risk of Parkinson's disease. Parkinsonism Relat Disord 2016; 32:25-30. [PMID: 27545685 DOI: 10.1016/j.parkreldis.2016.08.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 05/24/2016] [Accepted: 08/01/2016] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Genetic factors and environmental exposures, including pesticides, contribute to the risk of Parkinson's disease (PD). There have been few studies of gene and pesticide exposure interactions in PD, and all of the prior studies used a candidate gene approach. METHODS We performed the first genome-wide gene-environment interaction analysis of pesticide exposure and risk of Parkinson's disease. Analyses were performed using data on >700,000 single nucleotide polymorphisms (SNPs) in 364 discordant sibling pairs. In addition to testing for SNP-pesticide interaction effects, we also performed exploratory analyses of gene-pesticide interactions at the gene level. RESULTS None of the gene-environment interaction results were significant after genome-wide correction for multiple testing (α = 1.5E-07 for SNP-level tests; α = 2.1E-06 for gene-level tests). Top results in the SNP-level tests provided suggestive evidence (P < 5.0E-06) that the effect of pesticide exposure on PD risk may be modified by SNPs in the ERCC6L2 gene (P = 2.4E-06), which was also supported by suggestive evidence in the gene-level analysis (P = 4.7E-05). None of the candidate genes assessed in prior studies of gene-pesticide interactions reached statistical support in this genome-wide screen. CONCLUSION Although no significant interactions were identified, several of the genes with suggestive evidence of gene-environment interaction effects have biological plausibility for PD risk. Further investigation of the role of those genes in PD risk, particularly in the context of pesticide exposure, in large and carefully recruited samples is warranted.
Collapse
Affiliation(s)
- Joanna M Biernacka
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA; Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Sun Ju Chung
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | | | - Kari S Anderson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Christina M Lill
- Platform for Genome Analytics, Institutes of Neurogenetics & Integrative and Experimental Genomics, University of Lübeck, Lübeck, Germany
| | - Lars Bertram
- Platform for Genome Analytics, Institutes of Neurogenetics & Integrative and Experimental Genomics, University of Lübeck, Lübeck, Germany; School of Public Health, Faculty of Medicine, The Imperial College of Science, Technology, and Medicine, London, UK
| | - J E Ahlskog
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Laura Brighina
- Department of Neurology, San Gerardo Hospital, Milan Center for Neuroscience, Monza, Italy
| | - Roberta Frigerio
- Department of Neurology, NorthShore University HealthSystem, Evanston, IL, USA
| | | |
Collapse
|
59
|
George S, Brundin P. Immunotherapy in Parkinson's Disease: Micromanaging Alpha-Synuclein Aggregation. JOURNAL OF PARKINSONS DISEASE 2016; 5:413-24. [PMID: 26406122 PMCID: PMC4923719 DOI: 10.3233/jpd-150630] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Currently, several α-synuclein immunotherapies are being tested in experimental Parkinson’s disease models and in clinical trials. Recent research has revealed that α-synuclein is not just an intracellular synaptic protein but also exists extracellularly. Moreover, the transfer of misfolded α-synuclein between cells might be a crucial step in the process leading to a progressive increase in deposition of α-synuclein aggregates throughout the Parkinson’s disease brain. The revelation that α-synuclein is present outside cells has increased the interest in antibody-based therapies and opens up for the notion that microglia might play a key role in retarding Parkinson’s disease progression. The objectives of this review are to describe and contrast the use of active and passive immunotherapy in treating α-synucleinopathies and highlight the likely important role of microglia in clearing misfolded α-synuclein from the extracellular space.
Collapse
Affiliation(s)
| | - Patrik Brundin
- Correspondence to: Patrik Brundin, Van Andel Research Insti-tute, Center for Neurodegenerative Science, 333 Bostwick AvenueNE, Grand Rapids, MI 49503, USA.
Tel.: 616 234 5684; Fax: 616 234 5129.
| |
Collapse
|
60
|
Kim M, Semple I, Kim B, Kiers A, Nam S, Park HW, Park H, Ro SH, Kim JS, Juhász G, Lee JH. Drosophila Gyf/GRB10 interacting GYF protein is an autophagy regulator that controls neuron and muscle homeostasis. Autophagy 2016; 11:1358-72. [PMID: 26086452 DOI: 10.1080/15548627.2015.1063766] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Autophagy is an essential process for eliminating ubiquitinated protein aggregates and dysfunctional organelles. Defective autophagy is associated with various degenerative diseases such as Parkinson disease. Through a genetic screening in Drosophila, we identified CG11148, whose product is orthologous to GIGYF1 (GRB10-interacting GYF protein 1) and GIGYF2 in mammals, as a new autophagy regulator; we hereafter refer to this gene as Gyf. Silencing of Gyf completely suppressed the effect of Atg1-Atg13 activation in stimulating autophagic flux and inducing autophagic eye degeneration. Although Gyf silencing did not affect Atg1-induced Atg13 phosphorylation or Atg6-Pi3K59F (class III PtdIns3K)-dependent Fyve puncta formation, it inhibited formation of Atg13 puncta, suggesting that Gyf controls autophagy through regulating subcellular localization of the Atg1-Atg13 complex. Gyf silencing also inhibited Atg1-Atg13-induced formation of Atg9 puncta, which is accumulated upon active membrane trafficking into autophagosomes. Gyf-null mutants also exhibited substantial defects in developmental or starvation-induced accumulation of autophagosomes and autolysosomes in the larval fat body. Furthermore, heads and thoraxes from Gyf-null adults exhibited strongly reduced expression of autophagosome-associated Atg8a-II compared to wild-type (WT) tissues. The decrease in Atg8a-II was directly correlated with an increased accumulation of ubiquitinated proteins and dysfunctional mitochondria in neuron and muscle, which together led to severe locomotor defects and early mortality. These results suggest that Gyf-mediated autophagy regulation is important for maintaining neuromuscular homeostasis and preventing degenerative pathologies of the tissues. Since human mutations in the GIGYF2 locus were reported to be associated with a type of familial Parkinson disease, the homeostatic role of Gyf-family proteins is likely to be evolutionarily conserved.
Collapse
Affiliation(s)
- Myungjin Kim
- a Department of Molecular and Integrative Physiology ; University of Michigan ; Ann Arbor , MI USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Gallek MJ, Skoch J, Ansay T, Behbahani M, Mount D, Manziello A, Witte M, Bernas M, Labiner DM, Weinand ME. Cortical gene expression: prognostic value for seizure outcome following temporal lobectomy and amygdalohippocampectomy. Neurogenetics 2016; 17:211-218. [PMID: 27251580 DOI: 10.1007/s10048-016-0484-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 04/27/2016] [Indexed: 11/27/2022]
Abstract
Whole genome analyses were performed to test the hypothesis that temporal cortical gene expression differs between epilepsy patients rendered seizure-free versus non-seizure-free following anterior temporal lobectomy with amygdalohippocampectomy (ATL/AH). Twenty four patients underwent ATL/AH to treat medically intractable seizures of temporal lobe origin (mean age 35.5 years, mean follow-up 42.2 months); they were then dichotomized into seizure-free and non-seizure-free groups. Tissue RNA was isolated from the lateral temporal cortex and gene expression analysis was performed. Whole genome data were analyzed for prognostic value for seizure-free outcome following ATL/AH by logistic regression. Genes that could distinguish seizure outcome groups were identified based on providing an accuracy of >0.90 judging by area under the receiver operating characteristic curve, AUC, with a P value of the slope coefficient of <0.05. Four genes and seven RNA probes were with prognostic value for post-operative seizure-free outcome. Gene expression associated with seizure-free outcome included relative down-regulation of zinc finger protein 852 (ZNF852), CUB domain-containing protein 2 (CDCP2), proline-rich transmembrane protein 1 (PRRT1), hypothetical LOC440200 (FLJ41170), RNA probe 8047763, RNA probe 8126238, RNA probe 8113489, RNA probe 8092883, RNA probe 7935228, RNA probe 806293, and RNA probe 8104131. This study describes the predictive value of temporal cortical gene expression for seizure-free outcome after ATL/AH. Four genes and seven RNA probes were found to predict post-operative seizure-free outcome. Future prospective investigation of these genes and probes in human brain tissue and blood could establish new biomarkers predictive of seizure outcome following ATL/AH.
Collapse
Affiliation(s)
- Matthew J Gallek
- College of Nursing, University of Arizona, 1305 N Martin Ave, PO Box 210203, Tucson, AZ, 85721, USA.
| | - Jesse Skoch
- College of Medicine, Department of Surgery, Division of Neurosurgery, University of Arizona, Tucson, AZ, USA
| | - Tracy Ansay
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Mandana Behbahani
- College of Medicine, Department of Neurosurgery, University of Illinois at Chicago, Chicago, IL, USA
| | - David Mount
- Informatics/Bioinformatics Shared Services, Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Ann Manziello
- Informatics/Bioinformatics Shared Services, Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | - Marlys Witte
- College of Medicine, Department of Surgery, University of Arizona, Tucson, AZ, USA
| | - Michael Bernas
- College of Medicine, Department of Surgery, University of Arizona, Tucson, AZ, USA
| | - David M Labiner
- Department of Neurology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Martin E Weinand
- College of Medicine, Department of Surgery, Division of Neurosurgery, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
62
|
Korecka J, Levy S, Isacson O. In vivo modeling of neuronal function, axonal impairment and connectivity in neurodegenerative and neuropsychiatric disorders using induced pluripotent stem cells. Mol Cell Neurosci 2016; 73:3-12. [DOI: 10.1016/j.mcn.2015.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 12/04/2015] [Accepted: 12/08/2015] [Indexed: 02/07/2023] Open
|
63
|
Halbgebauer S, Öckl P, Wirth K, Steinacker P, Otto M. Protein biomarkers in Parkinson's disease: Focus on cerebrospinal fluid markers and synaptic proteins. Mov Disord 2016; 31:848-60. [PMID: 27134134 DOI: 10.1002/mds.26635] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/06/2016] [Accepted: 03/09/2016] [Indexed: 01/06/2023] Open
Abstract
Despite extensive research, to date, no validated biomarkers for PD have been found. This review seeks to summarize studies approaching the detection of biomarker candidates for PD and introduce promising ones in more detail, with special attention to synaptic proteins. To this end, we performed a PubMed search and included studies using proteomic tools (2-dimensional difference in gel electrophoresis and/or mass spectrometry) for the comparison of samples from PD and control patients. We found 27 studies reporting more than 500 differentially expressed proteins in which a total of 28 were detected in 2 and 17 in 3 or more independent studies, including posttranslationally modified proteins. In addition, of these 500 proteins, 25 were found to be brain specific, and 14 were enriched in synapses. Special attention was given to the applicability of the biomarker regarding sampling procedures, that is, using CSF/serum material for diagnosis. Furthermore, presynaptic proteins involved in vesicle membrane fusion seem to be interesting candidates for future analyses. Nonetheless, even though such promising biomarker candidates for PD exist, validation of these biomarkers in large-scale clinical studies is necessary to evaluate the diagnostic potential. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Patrick Öckl
- Department of Neurology, University of Ulm, Ulm, Germany
| | | | | | - Markus Otto
- Department of Neurology, University of Ulm, Ulm, Germany
| |
Collapse
|
64
|
Hernandez DG, Reed X, Singleton AB. Genetics in Parkinson disease: Mendelian versus non-Mendelian inheritance. J Neurochem 2016; 139 Suppl 1:59-74. [PMID: 27090875 DOI: 10.1111/jnc.13593] [Citation(s) in RCA: 320] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/25/2016] [Accepted: 02/09/2016] [Indexed: 12/12/2022]
Abstract
Parkinson's disease is a common, progressive neurodegenerative disorder, affecting 3% of those older than 75 years of age. Clinically, Parkinson's disease (PD) is associated with resting tremor, postural instability, rigidity, bradykinesia, and a good response to levodopa therapy. Over the last 15 years, numerous studies have confirmed that genetic factors contribute to the complex pathogenesis of PD. Highly penetrant mutations producing rare, monogenic forms of the disease have been discovered in singular genes such as SNCA, Parkin, DJ-1, PINK 1, LRRK2, and VPS35. Unique variants with incomplete penetrance in LRRK2 and GBA have been shown to be strong risk factors for PD in certain populations. Additionally, over 20 common variants with small effect sizes are now recognized to modulate the risk for PD. Investigating Mendelian forms of PD has provided precious insight into the pathophysiology that underlies the more common idiopathic form of disease; however, no treatment methodologies have developed. Furthermore, for identified common risk alleles, the functional basis underlying risk principally remains unknown. The challenge over the next decade will be to strengthen the findings delivered through genetic discovery by assessing the direct, biological consequences of risk variants in tandem with additional high-content, integrated datasets. This review discusses monogenic risk factors and mechanisms of Mendelian inheritance of Parkinson disease. Highly penetrant mutations in SNCA, Parkin, DJ-1, PINK 1, LRRK2 and VPS35 produce rare, monogenic forms of the disease, while unique variants within LRRK2 and GBA show incomplete penetrance and are strong risk factors for PD. Additionally, over 20 common variants with small effect sizes modulate disease risk. The challenge over the next decade is to strengthen genetic findings by assessing direct, biological consequences of risk variants in tandem with high-content, integrated datasets. This article is part of a special issue on Parkinson disease.
Collapse
Affiliation(s)
- Dena G Hernandez
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA.,German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Tübingen, Germany
| | - Xylena Reed
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA
| | - Andrew B Singleton
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, Maryland, USA.
| |
Collapse
|
65
|
Gómez-López S, Martínez-Silva AV, Montiel T, Osorio-Gómez D, Bermúdez-Rattoni F, Massieu L, Escalante-Alcalde D. Neural ablation of the PARK10 candidate Plpp3 leads to dopaminergic transmission deficits without neurodegeneration. Sci Rep 2016; 6:24028. [PMID: 27063549 PMCID: PMC4827058 DOI: 10.1038/srep24028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/18/2016] [Indexed: 01/02/2023] Open
Abstract
Parkinson’s disease (PD) is a multifactorial neurodegenerative disorder, characterised by the progressive loss of midbrain dopaminergic neurons and a variety of motor symptoms. The gene coding for the phospholipid phosphatase 3, PLPP3 (formerly PPAP2B or LPP3), maps within the PARK10 locus, a region that has been linked with increased risk to late-onset PD. PLPP3 modulates the levels of a range of bioactive lipids controlling fundamental cellular processes within the central nervous system. Here we show that PLPP3 is enriched in astroglial cells of the adult murine ventral midbrain. Conditional inactivation of Plpp3 using a Nestin::Cre driver results in reduced mesencephalic levels of sphingosine-1-phosphate receptor 1 (S1P1), a well-known mediator of pro-survival responses. Yet, adult PLPP3-deficient mice exhibited no alterations in the number of dopaminergic neurons or in the basal levels of striatal extracellular dopamine (DA). Potassium-evoked DA overflow in the striatum, however, was significantly decreased in mutant mice. Locomotor evaluation revealed that, although PLPP3-deficient mice exhibit motor impairment, this is not progressive or responsive to acute L-DOPA therapy. These findings suggest that disruption of Plpp3 during early neural development leads to dopaminergic transmission deficits in the absence of nigrostriatal degeneration, and without causing an age-related locomotor decline consistent with PD.
Collapse
Affiliation(s)
- Sandra Gómez-López
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, Mexico City, 04510, Mexico
| | - Ana Valeria Martínez-Silva
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, Mexico City, 04510, Mexico
| | - Teresa Montiel
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, Mexico City, 04510, Mexico
| | - Daniel Osorio-Gómez
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, Mexico City, 04510, Mexico
| | - Federico Bermúdez-Rattoni
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, Mexico City, 04510, Mexico
| | - Lourdes Massieu
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, Mexico City, 04510, Mexico
| | - Diana Escalante-Alcalde
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, Circuito Exterior s/n, Ciudad Universitaria, Mexico City, 04510, Mexico
| |
Collapse
|
66
|
Genetic Variants of Microtubule Actin Cross-linking Factor 1 (MACF1) Confer Risk for Parkinson’s Disease. Mol Neurobiol 2016; 54:2878-2888. [DOI: 10.1007/s12035-016-9861-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 03/17/2016] [Indexed: 01/12/2023]
|
67
|
Sesar A, Cacheiro P, López-López M, Camiña-Tato M, Quintáns B, Monroy-Jaramillo N, Alonso-Vilatela ME, Cebrián E, Yescas-Gómez P, Ares B, Rivas MT, Castro A, Carracedo A, Sobrido MJ. Synaptotagmin XI in Parkinson's disease: New evidence from an association study in Spain and Mexico. J Neurol Sci 2016; 362:321-5. [PMID: 26944171 DOI: 10.1016/j.jns.2016.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 01/10/2016] [Accepted: 02/07/2016] [Indexed: 01/29/2023]
Abstract
INTRODUCTION The pathophysiology of PD (Parkinson's disease) has been related to the ubiquitin proteasome system and oxidative stress. Parkin acts as ubiquitin ligase on several substrates. Because genetic variants often have different frequencies across populations, population specific analyses are necessary to complement and validate results from genome-wide association studies. METHODS We carried out an association study with genes coding for parkin substrates and cellular stress components in the Galician population (Northern Spain). SNCA and MAPT SNPs were also analyzed. We studied 75 SNPs in a discovery sample of 268 PD patients and 265 controls from Galicia. A replication sample of 271 patients and 260 controls was recruited from Mexico City. RESULTS We observed significant association between PD and SNPs in MAPT. Nominal p-values<0.05 were obtained in the Galician cohort for SNPs in SYT11, coding for synaptotagmin XI. These results were replicated in the Mexican sample. DISCUSSION The associated markers lie within a ~140kb strong linkage disequilibrium segment that harbors several candidate genes, including SYT11. SNPs from the GBA-SYT11-RAB25 region have been previously associated with PD, however the functionally relevant variants remain unknown. Our data support a likely role of genetic factors within 1q22 in PD susceptibility.
Collapse
Affiliation(s)
- Angel Sesar
- Department of Neurology, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain.
| | - Pilar Cacheiro
- Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain; Grupo de Medicina Xenómica, Fundación Pública Galega de Medicina Xenómica-SERGAS, Santiago de Compostela, Spain
| | - Marisol López-López
- Biological Systems Department, Universidad Autónoma Metropolitana, Campus Xochimilco, Mexico City, Mexico
| | - Montserrat Camiña-Tato
- Grupo de Medicina Xenómica, Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Beatriz Quintáns
- Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain; Grupo de Medicina Xenómica, Fundación Pública Galega de Medicina Xenómica-SERGAS, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III, Spain
| | - Nancy Monroy-Jaramillo
- Neurogenetics Department, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Mexico City, Mexico
| | - María-Elisa Alonso-Vilatela
- Neurogenetics Department, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Mexico City, Mexico
| | - Ernesto Cebrián
- Department of Neurology, Complejo Hospitalario Universitario de Pontevedra, Pontevedra, Spain
| | - Petra Yescas-Gómez
- Neurogenetics Department, Instituto Nacional de Neurología y Neurocirugía "Manuel Velasco Suárez", Mexico City, Mexico
| | - Begoña Ares
- Department of Neurology, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - María-Teresa Rivas
- Department of Neurology, Complejo Hospitalario Universitario de Coruña, La Coruña, Spain
| | - Alfonso Castro
- Department of Neurology, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain; Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Angel Carracedo
- Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain; Grupo de Medicina Xenómica, Fundación Pública Galega de Medicina Xenómica-SERGAS, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III, Spain
| | - María-Jesús Sobrido
- Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain; Grupo de Medicina Xenómica, Fundación Pública Galega de Medicina Xenómica-SERGAS, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III, Spain
| |
Collapse
|
68
|
Davis AA, Andruska KM, Benitez BA, Racette BA, Perlmutter JS, Cruchaga C. Variants in GBA, SNCA, and MAPT influence Parkinson disease risk, age at onset, and progression. Neurobiol Aging 2016; 37:209.e1-209.e7. [PMID: 26601739 PMCID: PMC4688052 DOI: 10.1016/j.neurobiolaging.2015.09.014] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/31/2015] [Accepted: 09/19/2015] [Indexed: 11/18/2022]
Abstract
Multiple genetic variants have been linked to risk of Parkinson disease (PD), but known mutations do not explain a large proportion of the total PD cases. Similarly, multiple loci have been associated with PD risk by genome-wide association studies (GWAS). The influence that genetic factors confer on phenotypic diversity remains unclear. Few studies have been performed to determine whether the GWAS loci are also associated with age at onset (AAO) or motor progression. We used 2 PD case-control data sets (Washington University and the Parkinson's Progression Markers Initiative) to determine whether polymorphisms located at the GWAS top hits (GBA, ACMSD/TMEM163, STK39, MCCC1/LAMP3, GAK/TMEM175, SNCA, and MAPT) show association with AAO or motor progression. We found associations between single nucleotide polymorphisms at the GBA and MAPT loci and PD AAO and progression. These findings reinforce the complex genetic basis of PD and suggest that distinct genes and variants explain the genetic architecture of PD risk, onset, and progression.
Collapse
Affiliation(s)
- Albert A Davis
- Department of Neurology, Washington University, St. Louis, MO, USA
| | | | - Bruno A Benitez
- Department of Psychiatry, Washington University, School of Medicine, St. Louis, MO, USA
| | - Brad A Racette
- Department of Neurology, Washington University, St. Louis, MO, USA; Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University, St. Louis, MO, USA; School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Parktown, South Africa
| | - Joel S Perlmutter
- Department of Neurology, Washington University, St. Louis, MO, USA; Department of Radiology, Washington University, St. Louis, MO, USA; Department of Anatomy and Neurobiology, Washington University, St. Louis, MO, USA; Programs in Physical Therapy and Occupational Therapy, Washington University, St. Louis, MO, USA; Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University, St. Louis, MO, USA
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, School of Medicine, St. Louis, MO, USA; Hope Center Program on Protein Aggregation and Neurodegeneration, Washington University, St. Louis, MO, USA.
| |
Collapse
|
69
|
Simón-Sánchez J, Heutink P, Gasser T. Variation in PARK10 is not associated with risk and age at onset of Parkinson's disease in large clinical cohorts. Neurobiol Aging 2015; 36:2907.e13-7. [PMID: 26260214 DOI: 10.1016/j.neurobiolaging.2015.07.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/01/2015] [Accepted: 07/03/2015] [Indexed: 11/17/2022]
Abstract
A recent study in autopsy-confirmed Parkinson's disease (PD) patients and controls revived the debate about the role of PARK10 in this disorder. In an attempt to replicate these results and further understand the role of this locus in the risk and age at onset of PD, we decided to explore NeuroX genotyping and whole exome sequencing data from 2 large independent cohorts of clinical patients and controls from the International Parkinson's Disease Genomic Consortium. A series of single-variant and gene-based aggregation (sequence kernel association test and combined multivariate and collapsing test) statistical tests suggested that common and rare genetic variation in this locus do not influence the risk or age at onset of clinical PD.
Collapse
Affiliation(s)
- Javier Simón-Sánchez
- Genetics and Epigenetics of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany; Genetics and Epigenetics of Neurodegeneration, German Center for Neurodegenerative diseases (DZNE)-Tübingen, Tübingen, Germany.
| | - Peter Heutink
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany; Genome Biology of Neurodegenerative Diseases, German Center for Neurodegenerative diseases (DZNE)-Tübingen, Tübingen, Germany
| | - Thomas Gasser
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany; Genome Biology of Neurodegenerative Diseases, German Center for Neurodegenerative diseases (DZNE)-Tübingen, Tübingen, Germany
| |
Collapse
|
70
|
Polymorphism in the Vesicular Monoamine Transporter 2 Gene Decreases the Risk of Parkinson's Disease in Han Chinese Men. PARKINSONS DISEASE 2015; 2015:903164. [PMID: 26246935 PMCID: PMC4515295 DOI: 10.1155/2015/903164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/19/2015] [Accepted: 06/24/2015] [Indexed: 02/05/2023]
Abstract
Background. Polymorphisms rs363371 and rs363324 in the vesicular monoamine transporter 2 (VMAT2) gene have been associated with risk of PD in an Italian population, and our aim is to investigate the association between the two single-nucleotide polymorphisms and PD in Han Chinese. Methods. 561 Han Chinese PD patients and 491 healthy age- and gender-matched controls were genotyped using Ligase detection reaction (LDR) method. Result. Both of patient and control groups showed similar genotype frequencies between patients and controls at both rs363371 and rs363324, as well as similar minor A allele frequencies at rs363371 (P = 0.452) and rs363324 (P = 0.413). None of the observed haplotypes showed a significant association with PD. Subgroup analysis by gender and age at onset revealed a significant association between the A allele of rs363371 and PD in Han Chinese males relative to healthy controls (OR 0.799, 95% CI 0.665 to 0.959, P = 0.016), and this association remained significant after adjusting for age (OR 0.785, 95% CI 0.652 to 0.945, P = 0.011). Conclusion. These results suggest that polymorphism of VMAT2 locus is associated with risk of PD in Han Chinese overall but that the A allele at rs363371 may protect against PD in Han Chinese males.
Collapse
|
71
|
Yu RL, Guo JF, Wang YQ, Liu ZH, Sun ZF, Su L, Zhang Y, Yan XX, Tang BS. The single nucleotide polymorphism Rs12817488 is associated with Parkinson’s disease in the Chinese population. J Clin Neurosci 2015; 22:1002-4. [DOI: 10.1016/j.jocn.2014.11.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 11/19/2014] [Accepted: 11/25/2014] [Indexed: 11/25/2022]
|
72
|
Han S, Andrei AC, Tsui KW. Efficient Stratified Testing Procedure for a False Discovery Rate. COMMUN STAT-SIMUL C 2015. [DOI: 10.1080/03610918.2013.809097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
73
|
Smith JG, Newton-Cheh C. Genome-wide association studies of late-onset cardiovascular disease. J Mol Cell Cardiol 2015; 83:131-41. [PMID: 25870159 DOI: 10.1016/j.yjmcc.2015.04.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/20/2015] [Accepted: 04/03/2015] [Indexed: 11/26/2022]
Abstract
Human genetics is a powerful tool for discovering causal mediators of human disease and physiology. Cardiovascular diseases with late onset in the lifecourse have historically not been considered genetic diseases, but in recent years the contribution of a heritable factor has been established. More importantly, over the last decade genome-wide association studies (GWASs) have identified many loci associated with late-onset cardiovascular diseases including coronary artery disease, carotid artery disease, ischemic stroke, aortic aneurysm, peripheral vascular disease, atrial fibrillation, valvular disease and correlates of vascular and myocardial function. Here we review findings from GWASs considered statistically robust with regard to multiple testing (p<5×10(-8)) for late-onset cardiovascular diseases and traits. Although for only a handful of the 92 genetic loci described here have the mechanisms underlying disease association been established, new and previously unsuspected pathways have been implicated for several conditions. Examples include a role for NO signaling in myocardial repolarization and sudden cardiac death and a role for the protein sortilin in lipid metabolism and coronary artery disease. Genetic loci with multiple trait associations have also provided novel biological insights. For example, of the 46 genetic loci associated with coronary artery disease, only 16 are also associated with conventional risk factors for cardiovascular disease whereas the remaining two thirds may reflect novel pathways. Much work remains to functionally characterize genetic loci and for clinical utility, but accruing insights into the biological basis of cardiovascular aging in human populations promise to point to novel therapeutic and preventive strategies. This article is part of a Special Issue entitled 'SI:CV Aging'.
Collapse
Affiliation(s)
- J Gustav Smith
- Center for Human Genetic Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Department of Cardiology, Clinical Sciences, Lund University and Skåne University Hospital, Lund, Sweden.
| | - Christopher Newton-Cheh
- Center for Human Genetic Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
74
|
Escott-Price V, Nalls MA, Morris HR, Lubbe S, Brice A, Gasser T, Heutink P, Wood NW, Hardy J, Singleton AB, Williams NM. Polygenic risk of Parkinson disease is correlated with disease age at onset. Ann Neurol 2015; 77:582-91. [PMID: 25773351 PMCID: PMC4737223 DOI: 10.1002/ana.24335] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 11/28/2014] [Accepted: 12/07/2014] [Indexed: 02/01/2023]
Abstract
Objective We have investigated the polygenic architecture of Parkinson disease (PD) and have also explored the potential relationship between an individual's polygenic risk score and their disease age at onset. Methods This study used genotypic data from 4,294 cases and 10,340 controls obtained from the meta‐analysis of PD genome‐wide association studies. Polygenic score analysis was performed as previously described by the International Schizophrenia Consortium, testing whether the polygenic score alleles identified in 1 association study were significantly enriched in the cases relative to the controls of 3 independent studies. Linear regression was used to investigate the relationship between an individual's polygenic score for PD risk alleles and disease age at onset. Results Our polygenic score analysis has identified significant evidence for a polygenic component enriched in the cases of each of 3 independent PD genome‐wide association cohorts (minimum p = 3.76 × 10−6). Further analysis identified compelling evidence that the average polygenic score in patients with an early disease age at onset was significantly higher than in those with a late age at onset (p = 0.00014). Interpretation This provides strong support for a large polygenic contribution to the overall heritable risk of PD and also suggests that early onset forms of the illness are not exclusively caused by highly penetrant Mendelian mutations, but can also be contributed to by an accumulation of common polygenic alleles with relatively low effect sizes. Ann Neurol 2015;77:582–591
Collapse
Affiliation(s)
- Valentina Escott-Price
- Institute of Psychological Medicine and Clinical Neurosciences, Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, Cardiff University School of Medicine, Cardiff, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Abstract
Many neurological disorders are characterised by structural changes in neuronal connections, ranging from presymptomatic synaptic changes to the loss or rewiring of entire axon bundles. The molecular mechanisms that underlie this perturbed connectivity are poorly understood, but recent studies suggest a role for axon guidance proteins. Axon guidance proteins guide growing axons during development and control structural plasticity of synaptic connections in adults. Changes in expression or function of these proteins might induce pathological changes in neural circuits that predispose to, or cause, neurological diseases. For some neurological disorders, such as midline crossing disorders, investigators have identified causative mutations in genes for axon guidance. However, for most other disorders, evidence is correlative and further studies are needed to confirm the pathological role of defects in proteins for axon guidance. Importantly, further insight into how dysregulation of axon guidance proteins causes disease will help the development of therapeutic strategies for neurological disorders.
Collapse
Affiliation(s)
- Eljo Y Van Battum
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sara Brignani
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
76
|
von Otter M, Bergström P, Quattrone A, De Marco EV, Annesi G, Söderkvist P, Wettinger SB, Drozdzik M, Bialecka M, Nissbrandt H, Klein C, Nilsson M, Hammarsten O, Nilsson S, Zetterberg H. Genetic associations of Nrf2-encoding NFE2L2 variants with Parkinson's disease - a multicenter study. BMC MEDICAL GENETICS 2014; 15:131. [PMID: 25496089 PMCID: PMC4335439 DOI: 10.1186/s12881-014-0131-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/01/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND The transcription factor Nrf2, encoded by the NFE2L2 gene, is an important regulator of the cellular protection against oxidative stress. Parkinson's disease is a neurodegenerative disease highly associated with oxidative stress. In a previously published study, we reported associations of NFE2L2 haplotypes with risk and age at onset of idiopathic Parkinson's disease in a Swedish discovery material and a Polish replication material. Here, we have extended the replication study and performed meta-analyses including the Polish material and four new independent European patient-control materials. Furthermore, all SNPs included in the haplotype windows were investigated individually for associations with Parkinson's disease in meta-analyses including all six materials. METHODS Totally 1038 patients and 1600 control subjects were studied. Based on previous NFE2L2 haplotype associations with Parkinson's disease, five NFE2L2 tag SNPs were genotyped by allelic discrimination and three functional NFE2L2 promoter SNPs were genotyped by sequencing. The impact of individual SNPs and haplotypes on risk and age at onset of Parkinson's disease were investigated in each material individually and in meta-analyses of the obtained results. RESULTS Meta-analyses of NFE2L2 haplotypes showed association of haplotype GAGCAAAA, including the fully functional promoter haplotype AGC, with decreased risk (OR = 0.8 per allele, p = 0.012) and delayed onset (+1.1 years per allele, p = 0.048) of Parkinson's disease. These results support the previously observed protective effect of this haplotype in the first study. Further, meta-analyses of the SNPs included in the haplotypes revealed four NFE2L2 SNPs associated with age at onset of Parkinson's disease (rs7557529 G > A, -1.0 years per allele, p = 0.042; rs35652124 A > G, -1.1 years per allele, p = 0.045; rs2886161 A > G, -1.2 years per allele, p = 0.021; rs1806649 G > A, +1.2 years per allele, p = 0.029). One of these (rs35652124) is a functional SNP located in the NFE2L2 promoter. No individual SNP was associated with risk of Parkinson's disease. CONCLUSION Our results support the hypothesis that variation in the NFE2L2 gene, encoding a central protein in the cellular protection against oxidative stress, may contribute to the pathogenesis of Parkinson's disease. Functional studies are now needed to explore these results further.
Collapse
|
77
|
Spataro N, Calafell F, Cervera-Carles L, Casals F, Pagonabarraga J, Pascual-Sedano B, Campolongo A, Kulisevsky J, Lleó A, Navarro A, Clarimón J, Bosch E. Mendelian genes for Parkinson's disease contribute to the sporadic forms of the disease†. Hum Mol Genet 2014; 24:2023-34. [PMID: 25504046 DOI: 10.1093/hmg/ddu616] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Nino Spataro
- Institute of Evolutionary Biology (CSIC-UPF), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Francesc Calafell
- Institute of Evolutionary Biology (CSIC-UPF), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Laura Cervera-Carles
- Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau-Hospital de Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain, Center for Networking Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ferran Casals
- Genomics Core Facility, Universitat Pompeu Fabra, Barcelona Biomedical Research Park (PRBB), 08003 Barcelona, Spain
| | - Javier Pagonabarraga
- Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau-Hospital de Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain, Center for Networking Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Berta Pascual-Sedano
- Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau-Hospital de Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain, Center for Networking Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Antònia Campolongo
- Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau-Hospital de Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain, Center for Networking Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jaime Kulisevsky
- Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau-Hospital de Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain, Center for Networking Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain, Health Sciences Department, Universitat Oberta de Catalunya, Catalonia, Spain
| | - Alberto Lleó
- Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau-Hospital de Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain, Center for Networking Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Arcadi Navarro
- Institute of Evolutionary Biology (CSIC-UPF), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain, National Institute for Bioinformatics (INB), Barcelona Biomedical Research Park (PRBB), 08003 Barcelona, Spain, Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona Biomedical Research Park (PRBB), 08003 Barcelona, Spain and Center for Genomic Regulation (CRG), Barcelona Biomedical Research Park (PRBB), 08003 Barcelona, Spain
| | - Jordi Clarimón
- Department of Neurology, Institut d'Investigacions Biomèdiques Sant Pau-Hospital de Sant Pau, Universitat Autònoma de Barcelona, 08025 Barcelona, Spain, Center for Networking Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Elena Bosch
- Institute of Evolutionary Biology (CSIC-UPF), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain,
| |
Collapse
|
78
|
Vaidyanathan U, Malone SM, Miller MB, McGue M, Iacono WG. Heritability and molecular genetic basis of acoustic startle eye blink and affectively modulated startle response: a genome-wide association study. Psychophysiology 2014; 51:1285-99. [PMID: 25387708 PMCID: PMC4231542 DOI: 10.1111/psyp.12348] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Acoustic startle responses have been studied extensively in relation to individual differences and psychopathology. We examined three indices of the blink response in a picture-viewing paradigm-overall startle magnitude across all picture types, and aversive and pleasant modulation scores-in 3,323 twins and parents. Biometric models and molecular genetic analyses showed that half the variance in overall startle was due to additive genetic effects. No single nucleotide polymorphism was genome-wide significant, but GRIK3 produced a significant effect when examined as part of a candidate gene set. In contrast, emotion modulation scores showed little evidence of heritability in either biometric or molecular genetic analyses. However, in a genome-wide scan, PARP14 produced a significant effect for aversive modulation. We conclude that, although overall startle retains potential as an endophenotype, emotion-modulated startle does not.
Collapse
Affiliation(s)
- Uma Vaidyanathan
- Department of Psychology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | | | | |
Collapse
|
79
|
Ahmed I, Lee PC, Lill CM, Searles Nielsen S, Artaud F, Gallagher LG, Loriot MA, Mulot C, Nacfer M, Liu T, Biernacka JM, Armasu S, Anderson K, Farin FM, Funch Lassen C, Hansen J, Olsen JH, Bertram L, Maraganore DM, Checkoway H, Ritz B, Elbaz A. Lack of replication of the GRIN2A-by-coffee interaction in Parkinson disease. PLoS Genet 2014; 10:e1004788. [PMID: 25412286 PMCID: PMC4238979 DOI: 10.1371/journal.pgen.1004788] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Ismaïl Ahmed
- INSERM, Centre for research in Epidemiology and Population Health, U1018, Biostatistics team, Villejuif, France
- Univ Paris-Sud, UMRS 1018, Paris, Villejuif, France
| | - Pei-Chen Lee
- Department of Health Care Management, College of Healthcare Administration and Management, National Taipei University of Nursing Health Sciences, Taipei, Taiwan
| | - Christina M. Lill
- Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Department of Neurology, Focus Program Translational Neuroscience, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Susan Searles Nielsen
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Fanny Artaud
- INSERM, Centre for research in Epidemiology and Population Health, U1018, Social and occupational determinants of health, Villejuif, France
- Univ de Versailles St-Quentin, UMRS 1018, Versailles, France
| | - Lisa G. Gallagher
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Marie-Anne Loriot
- Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Biochimie, Paris, France
- Université Paris Descartes, Inserm, UMR-S1147, Médecine personnalisée, Pharmacogénomique et optimisation thérapeutique, Paris, France
| | - Claire Mulot
- Université Paris Descartes, Inserm, UMR-S1147, Médecine personnalisée, Pharmacogénomique et optimisation thérapeutique, Paris, France
- Centre de ressources biologiques (CRB) Epigenetec, Paris, France
| | - Magali Nacfer
- Université Paris Descartes, Inserm, UMR-S1147, Médecine personnalisée, Pharmacogénomique et optimisation thérapeutique, Paris, France
- Centre de ressources biologiques (CRB) Epigenetec, Paris, France
| | - Tian Liu
- Max Planck Institute for Human Development, Berlin, Germany
| | - Joanna M. Biernacka
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Sebastian Armasu
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kari Anderson
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Federico M. Farin
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | | | - Johnni Hansen
- Danish Cancer Society Research Center, Danish Cancer Society, Copenhagen, Denmark
| | - Jørgen H. Olsen
- Danish Cancer Society Research Center, Danish Cancer Society, Copenhagen, Denmark
| | - Lars Bertram
- Department of Vertebrate Genomics, Neuropsychiatric Genetics Group, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Department of Medicine, School of Public Health, Imperial College London, London, United Kingdom
| | - Demetrius M. Maraganore
- Department of Neurology, NorthShore University HealthSystem, Evanston, Illinois, United States of America
| | - Harvey Checkoway
- Department of Family and Preventive Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - Beate Ritz
- Department of Epidemiology, Fielding School of Public Health, University of California Los Angeles, Los Angeles, California, United States of America
| | - Alexis Elbaz
- INSERM, Centre for research in Epidemiology and Population Health, U1018, Social and occupational determinants of health, Villejuif, France
- Univ de Versailles St-Quentin, UMRS 1018, Versailles, France
- * E-mail:
| |
Collapse
|
80
|
Dashtipour K. Do genetic factors protect against Parkinson's disease? What I can learn from my healthy grandma. Med Hypotheses 2014; 83:637-9. [PMID: 25459129 DOI: 10.1016/j.mehy.2014.09.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 09/16/2014] [Accepted: 09/30/2014] [Indexed: 11/29/2022]
Abstract
Parkinson's disease (PD) is a progressive, neurodegenerative disorder and it affects 4-5% of people age 85 years or older. The etiopathogenesis of PD is a consequence of interaction between two factors, environmental pathogens and genetic susceptibility. If an environmental agent such as a toxin or pathogen were to play a major role in the causality of PD, it would need to be something relatively ubiquitous in our environment since we cannot find a specific population at risk. On the other hand, all efforts to implicate specific genetic sequences in risk of PD were futile since the great majority of PD cases are sporadic; however, if the majority of the population is exposed to a culpable environmental factor and only 5% of the population 85 years or older manifest the disorder, this raises an important question: Why and how does vast majority of the population not manifest with PD? It seems that we should investigate the certain genome or epigenetic alterations of the unaffected 95%. This large non affected population might have PD but they are not yet symptomatic and some may not be so for at least another 10 or 20 years. To further address this issue, we should screen and study the population that have been exposed to the environmental factor but with high certainty are not yet affected. Therefore the perfect population would be non-PD subjects who are 90 years or older. We believe the following are the unmet research needs that deserve more attention in PD. (1) More genetic studies. Comparison should be between PD subjects and non-PD control subjects who are 90 years old and above. (2) Study the mechanism of action of the candidate genes, as a subsequent examination of their gene products may lead to the discovery of neuroprotective agents in the disease.
Collapse
Affiliation(s)
- Khashayar Dashtipour
- Loma Linda University School of Medicine, Department of Neurology, 11370 Anderson, Suite B-100, Loma Linda, CA 92354, USA.
| |
Collapse
|
81
|
O'Malley AM, Shanley DK, Kelly AT, Barry DS. Towards an understanding of semaphorin signalling in the spinal cord. Gene 2014; 553:69-74. [PMID: 25300255 DOI: 10.1016/j.gene.2014.10.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/05/2014] [Indexed: 01/11/2023]
Abstract
Semaphorins are a large family of proteins that are classically associated with axon guidance. These proteins and their interacting partners, the neuropilins and plexins are now known to be key mediators in a variety of processes throughout the nervous system ranging from synaptic refinement to the correct positioning of neuronal and glial cell bodies. Recently, much attention has been given to the roles semaphorins play in other body tissues including the immune and vascular systems. This review wishes to draw attention back to the nervous system, specifically focusing on the role of semaphorins in the development of the spinal cord and their proposed roles in the adult. In addition, their functions in spinal cord injury at the glial scar are also discussed.
Collapse
Affiliation(s)
- Aisling M O'Malley
- Department of Anatomy, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Daniel K Shanley
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | - Albert T Kelly
- Department of Anatomy, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Denis S Barry
- Department of Anatomy, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
82
|
Wang Z, Zhang H, Yang H, Wang S, Rong E, Pei W, Li H, Wang N. Genome-wide association study for wool production traits in a Chinese Merino sheep population. PLoS One 2014; 9:e107101. [PMID: 25268383 PMCID: PMC4182092 DOI: 10.1371/journal.pone.0107101] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 08/14/2014] [Indexed: 01/01/2023] Open
Abstract
Genome-wide association studies (GWAS) provide a powerful approach for identifying quantitative trait loci without prior knowledge of location or function. To identify loci associated with wool production traits, we performed a genome-wide association study on a total of 765 Chinese Merino sheep (JunKen type) genotyped with 50 K single nucleotide polymorphisms (SNPs). In the present study, five wool production traits were examined: fiber diameter, fiber diameter coefficient of variation, fineness dispersion, staple length and crimp. We detected 28 genome-wide significant SNPs for fiber diameter, fiber diameter coefficient of variation, fineness dispersion, and crimp trait in the Chinese Merino sheep. About 43% of the significant SNP markers were located within known or predicted genes, including YWHAZ, KRTCAP3, TSPEAR, PIK3R4, KIF16B, PTPN3, GPRC5A, DDX47, TCF9, TPTE2, EPHA5 and NBEA genes. Our results not only confirm the results of previous reports, but also provide a suite of novel SNP markers and candidate genes associated with wool traits. Our findings will be useful for exploring the genetic control of wool traits in sheep.
Collapse
Affiliation(s)
- Zhipeng Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin, P. R. China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Hui Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin, P. R. China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Hua Yang
- Institute of Animal Husbandry and Veterinary, Xinjiang Academy of Agricultural and Reclamation Science, Shihezi, P.R. China
| | - Shouzhi Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin, P. R. China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Enguang Rong
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin, P. R. China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Wenyu Pei
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin, P. R. China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
| | - Hui Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin, P. R. China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
- * E-mail: (NW); (HL)
| | - Ning Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang province, Harbin, P. R. China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, P. R. China
- * E-mail: (NW); (HL)
| |
Collapse
|
83
|
Abstract
Parkinson’s disease (PD) is a progressively debilitating neurodegenerative syndrome. Although best described as a movement disorder, the condition has prominent autonomic, cognitive, psychiatric, sensory and sleep components. Striatal dopaminergic innervation and nigral neurons are progressively lost, with associated Lewy pathology readily apparent on autopsy. Nevertheless, knowledge of the molecular events leading to this pathophysiology is limited. Current therapies offer symptomatic benefit but they fail to slow progression and patients continue to deteriorate. Recent discoveries in sporadic, Mendelian and more complex forms of parkinsonism provide novel insight into disease etiology; 28 genes, including those encoding alpha-synuclein (SNCA), leucine-rich repeat kinase 2 (LRRK2) and microtubule-associated protein tau (MAPT), have been linked and/or associated with PD. A consensus regarding the affected biological pathways and molecular processes has also started to emerge. In early-onset and more a typical PD, deficits in mitophagy pathways and lysosomal function appear to be prominent. By contrast, in more typical late-onset PD, chronic, albeit subtle, dysfunction in synaptic transmission, early endosomal trafficking and receptor recycling, as well as chaperone-mediated autophagy, provide a unifying synthesis of the molecular pathways involved. Disease-modification (neuroprotection) is no longer such an elusive goal given the unparalleled opportunity for diagnosis, translational neuroscience and therapeutic development provided by genetic discovery.
Collapse
Affiliation(s)
- Michelle K Lin
- Djavad Mowafaghian Centre for Brain Health, Centre for Applied Neurogenetics, Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Matthew J Farrer
- Djavad Mowafaghian Centre for Brain Health, Centre for Applied Neurogenetics, Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
84
|
Sekiyama K, Takamatsu Y, Waragai M, Hashimoto M. Role of genomics in translational research for Parkinson's disease. Biochem Biophys Res Commun 2014; 452:226-35. [PMID: 24950403 DOI: 10.1016/j.bbrc.2014.06.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 06/06/2014] [Indexed: 01/07/2023]
Abstract
Research on Parkinson's disease (PD) has made remarkable progress in recent decades, due largely to new genomic technologies, such as high throughput sequencing and microarray analyses. Since the discovery of a linkage of a missense mutation of the α-synuclein (αS) gene to a rare familial dominant form of PD in 1996, positional cloning and characterization of a number of familial PD risk factors have established a hypothesis that aggregation of αS may play a major role in the pathogenesis of PD. Furthermore, dozens of sensitizing alleles related to the disease have been identified by genome wide association studies (GWAS) and meta-GWAS, contributing to a better understanding of the pathological mechanisms of sporadic PD. Thus, the knowledge obtained from the association studies will be valuable for "the personal genome" of PD. Besides summarizing such progress, this paper focuses on the role of microRNAs in the field of PD research, since microRNAs might be promising as a biomarker and as a therapeutic reagent for PD. We further refer to a recent view that neurodegenerative diseases, including PD, coexist with metabolic disorders and are stimulated by type II diabetes, the most common disease among elderly populations. The development of genomic approaches may potentially contribute to therapeutic intervention for PD.
Collapse
Affiliation(s)
- Kazunari Sekiyama
- Tokyo Metropolitan Institute of Medical Sciences, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-0057, Japan
| | - Yoshiki Takamatsu
- Tokyo Metropolitan Institute of Medical Sciences, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-0057, Japan
| | - Masaaki Waragai
- Tokyo Metropolitan Institute of Medical Sciences, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-0057, Japan
| | - Makoto Hashimoto
- Tokyo Metropolitan Institute of Medical Sciences, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-0057, Japan.
| |
Collapse
|
85
|
Richter F, Gao F, Medvedeva V, Lee P, Bove N, Fleming SM, Michaud M, Lemesre V, Patassini S, De La Rosa K, Mulligan CK, Sioshansi PC, Zhu C, Coppola G, Bordet T, Pruss RM, Chesselet MF. Chronic administration of cholesterol oximes in mice increases transcription of cytoprotective genes and improves transcriptome alterations induced by alpha-synuclein overexpression in nigrostriatal dopaminergic neurons. Neurobiol Dis 2014; 69:263-75. [PMID: 24844147 DOI: 10.1016/j.nbd.2014.05.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 05/02/2014] [Accepted: 05/06/2014] [Indexed: 12/14/2022] Open
Abstract
Cholesterol-oximes TRO19622 and TRO40303 target outer mitochondrial membrane proteins and have beneficial effects in preclinical models of neurodegenerative diseases leading to their advancement to clinical trials. Dopaminergic neurons degenerate in Parkinson's disease (PD) and are prone to oxidative stress and mitochondrial dysfunction. In order to provide insights into the neuroprotective potential of TRO19622 and TRO40303 for dopaminergic neurons in vivo, we assessed their effects on gene expression in laser captured nigrostriatal dopaminergic neurons of wildtype mice and of mice that over-express alpha-synuclein, a protein involved in both familial and sporadic forms of PD (Thy1-aSyn mice). Young mice were fed the drugs in food pellets or a control diet from 1 to 4months of age, approximately 10months before the appearance of striatal dopamine loss in this model. Unbiased weighted gene co-expression network analysis (WGCNA) of transcriptional changes revealed effects of cholesterol oximes on transcripts related to mitochondria, cytoprotection and anti-oxidant response in wild-type and transgenic mice, including increased transcription of stress defense (e.g. Prdx1, Prdx2, Glrx2, Hspa9, Pink1, Drp1, Trak1) and dopamine-related (Th, Ddc, Gch1, Dat, Vmat2, Drd2, Chnr6a) genes. Even at this young age transgenic mice showed alterations in transcripts implicated in mitochondrial function and oxidative stress (e.g. Bcl-2, Bax, Casp3, Nos2), and both drugs normalized about 20% of these alterations. Young Thy1-aSyn mice exhibit motor deficits that differ from parkinsonism and are established before the onset of treatment; these deficits were not improved by cholesterol oximes. However, high doses of TRO40303 improved olfaction and produced the same effects as dopamine agonists on a challenging beam test, specifically an increase in footslips, an observation congruent with its effects on transcripts involved in dopamine synthesis. High doses of TRO19622 increased alpha-synuclein aggregates in the substantia nigra; this effect, not seen with TRO40303 was inconsistent and may represent a protective mechanism as in other neurodegenerative diseases. Overall, the results suggest that cholesterol oximes, while not improving early effects of alpha-synuclein overexpression on motor behavior or pathology, may ameliorate the function and resilience of dopaminergic neurons in vivo and support further studies of neuroprotection in models with dopaminergic cell loss.
Collapse
Affiliation(s)
- Franziska Richter
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Fuying Gao
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Vera Medvedeva
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Patrick Lee
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Nicholas Bove
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Sheila M Fleming
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Magali Michaud
- Trophos S.A. Parc Scientifique de Luminy, Case 931, 13288 Marseille Cedex 9, France
| | - Vincent Lemesre
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Stefano Patassini
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Krystal De La Rosa
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Caitlin K Mulligan
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Pedrom C Sioshansi
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Chunni Zhu
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Giovanni Coppola
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA; Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
| | - Thierry Bordet
- Trophos S.A. Parc Scientifique de Luminy, Case 931, 13288 Marseille Cedex 9, France
| | - Rebecca M Pruss
- Trophos S.A. Parc Scientifique de Luminy, Case 931, 13288 Marseille Cedex 9, France
| | - Marie-Françoise Chesselet
- Department of Neurology, The David Geffen School of Medicine at UCLA, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA.
| |
Collapse
|
86
|
Goldman SM, Kamel F, Ross GW, Jewell SA, Marras C, Hoppin JA, Umbach DM, Bhudhikanok GS, Meng C, Korell M, Comyns K, Hauser RA, Jankovic J, Factor SA, Bressman S, Lyons KE, Sandler DP, Langston JW, Tanner CM. Peptidoglycan recognition protein genes and risk of Parkinson's disease. Mov Disord 2014; 29:1171-80. [PMID: 24838182 DOI: 10.1002/mds.25895] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 03/12/2014] [Accepted: 03/25/2014] [Indexed: 12/26/2022] Open
Abstract
Increased gut permeability, inflammation, and colonic α-synuclein pathology are present in early Parkinson's disease (PD) and have been proposed to contribute to PD pathogenesis. Peptidoglycan is a structural component of the bacterial cell wall. Peptidoglycan recognition proteins (PGRPs) maintain healthy gut microbial flora by regulating the immune response to both commensal and harmful bacteria. We tested the hypothesis that variants in genes that encode PGRPs are associated with PD risk. Participants in two independent case-control studies were genotyped for 30 single-nucleotide polymorphisms (SNPs) in the four PGLYRP genes. Using logistic regression to estimate odds ratios (OR) and 95% confidence intervals (CI) adjusted for potential confounding variables, we conducted analyses in each study, separately and pooled. One SNP failed the assay, and three had little to no variation. The ORs were similar in both study populations. In pooled analyses, three of seven PGLYRP2 SNPs (rs3813135, rs733731, rs892145), one of five PGLYRP3 SNPs (rs2987763), and six of nine PGLYRP4 SNPs (rs10888557, rs12063091, rs3006440, rs3006448, rs3006458, and rs3014864) were significantly associated with PD risk. Association was strongest for PGLYRP4 5'untranslated region (UTR) SNP rs10888557 (GG reference, CG OR 0.6 [95%CI 0.4-0.9], CC OR 0.15 [95%CI 0.04-0.6]; log-additive P-trend, 0.0004). Common variants in PGLYRP genes are associated with PD risk in two independent studies. These results require replication, but they are consistent with hypotheses of a causative role for the gut microbiota and gastrointestinal immune response in PD.
Collapse
Affiliation(s)
- Samuel M Goldman
- San Francisco Veterans Affairs Medical Center, San Francisco, California, USA; University of California, San Francisco, San Francisco, California, USA; The Parkinson's Institute, Sunnyvale, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Na SJ, Lee JH, Kim SW, Kim DS, Shon EH, Park HJ, Shin HY, Kim SM, Choi YC. Whole-genome analysis in Korean patients with autoimmune myasthenia gravis. Yonsei Med J 2014; 55:660-8. [PMID: 24719132 PMCID: PMC3990093 DOI: 10.3349/ymj.2014.55.3.660] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/24/2013] [Accepted: 08/23/2013] [Indexed: 11/27/2022] Open
Abstract
PURPOSE The underlying cause of myasthenia gravis (MG) is unknown, although it likely involves a genetic component. However, no common genetic variants have been unequivocally linked to autoimmune MG. We sought to identify the genetic variants associated with an increased or decreased risk of developing MG in samples from a Korean Multicenter MG Cohort. MATERIALS AND METHODS To determine new genetic targets related to autoimmune MG, a whole genome-based single nucleotide polymorphisms (SNP) analysis was conducted using an Axiom™ Genome-Wide ASI 1 Array, comprising 598375 SNPs and samples from 109 MG patients and 150 neurologically normal controls. RESULTS In total, 641 SNPs from five case-control associations showed p-values of less than 10⁻⁵. From regional analysis, we selected seven candidate genes (RYR3, CACNA1S, SLAMF1, SOX5, FHOD3, GABRB1, and SACS) for further analysis. CONCLUSION The present study suggests that a few genetic polymorphisms, such as in RYR3, CACNA1S, and SLAMF1, might be related to autoimmune MG. Our findings also encourage further studies, particularly confirmatory studies with larger samples, to validate and analyze the association between these SNPs and autoimmune MG.
Collapse
Affiliation(s)
- Sang-Jun Na
- Department of Neurology, Konyang University College of Medicine, Daejeon, Korea
| | - Ji Hyun Lee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
| | - So Won Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
| | - Dae-Seong Kim
- Department of Neurology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Eun Hee Shon
- Department of Neurology, Chungnam University Hospital, Daejeon, Korea
| | - Hyung Jun Park
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Ha Young Shin
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Min Kim
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| | - Young-Chul Choi
- Department of Neurology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
88
|
Multivariate analysis reveals genetic associations of the resting default mode network in psychotic bipolar disorder and schizophrenia. Proc Natl Acad Sci U S A 2014; 111:E2066-75. [PMID: 24778245 DOI: 10.1073/pnas.1313093111] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The brain's default mode network (DMN) is highly heritable and is compromised in a variety of psychiatric disorders. However, genetic control over the DMN in schizophrenia (SZ) and psychotic bipolar disorder (PBP) is largely unknown. Study subjects (n = 1,305) underwent a resting-state functional MRI scan and were analyzed by a two-stage approach. The initial analysis used independent component analysis (ICA) in 324 healthy controls, 296 SZ probands, 300 PBP probands, 179 unaffected first-degree relatives of SZ probands (SZREL), and 206 unaffected first-degree relatives of PBP probands to identify DMNs and to test their biomarker and/or endophenotype status. A subset of controls and probands (n = 549) then was subjected to a parallel ICA (para-ICA) to identify imaging-genetic relationships. ICA identified three DMNs. Hypo-connectivity was observed in both patient groups in all DMNs. Similar patterns observed in SZREL were restricted to only one network. DMN connectivity also correlated with several symptom measures. Para-ICA identified five sub-DMNs that were significantly associated with five different genetic networks. Several top-ranking SNPs across these networks belonged to previously identified, well-known psychosis/mood disorder genes. Global enrichment analyses revealed processes including NMDA-related long-term potentiation, PKA, immune response signaling, axon guidance, and synaptogenesis that significantly influenced DMN modulation in psychoses. In summary, we observed both unique and shared impairments in functional connectivity across the SZ and PBP cohorts; these impairments were selectively familial only for SZREL. Genes regulating specific neurodevelopment/transmission processes primarily mediated DMN disconnectivity. The study thus identifies biological pathways related to a widely researched quantitative trait that might suggest novel, targeted drug treatments for these diseases.
Collapse
|
89
|
Várady G, Cserepes J, Németh A, Szabó E, Sarkadi B. Cell surface membrane proteins as personalized biomarkers: where we stand and where we are headed. Biomark Med 2014; 7:803-19. [PMID: 24044572 DOI: 10.2217/bmm.13.90] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Personalized medicine requires the development of a wide array of biomarker diagnostic assays, reflecting individual variations and thus allowing tailored therapeutic interventions. Membrane proteins comprise approximately 30% of total human proteins; they play a key role in various physiological functions and pathological conditions, although, currently, only a limited number of membrane proteins are applied as biomarkers. In many normal tissues, cell surface membrane proteins are not easily accessible for diagnostic sampling, and tumor-derived membrane preparations - while serving as potential tumor biomarkers - may not reflect physiological protein expression. In addition to post-translational modifications, which may include glycosylation, phosphorylation and lipid modifications, the trafficking of membrane proteins is also regulated. Moreover, a tight cellular quality control monitors membrane protein maturation, and continuous removal and reinsertion, involving special signaling systems, occurs in many cases. However, cell surface membrane proteins already serve as valuable prognostic and predicative biomarkers, for example, in hematological and immunological diseases, by the determination of the cluster of differentiation markers. In this review, we demonstrate the relevance of cell surface membrane biomarkers in various diseases and call attention to the potential application of red blood cell (erythrocyte) membrane proteins in this regard. Surprisingly, red blood cells express hundreds of membrane proteins, which seem to reflect a general genetic and regulatory background, and may serve as relatively stable and easily accessible personalized membrane biomarkers. Quantitative membrane protein detection in red blood cells by flow cytometry may bring a breakthrough in this regard.
Collapse
Affiliation(s)
- György Várady
- MTA-SE Molecular Biophysics Research Group, Semmelweis University, Budapest, Hungary
| | | | | | | | | |
Collapse
|
90
|
Sheerin UM, Houlden H, Wood NW. Advances in the Genetics of Parkinson's Disease: A Guide for the Clinician. Mov Disord Clin Pract 2014; 1:3-13. [PMID: 30363913 DOI: 10.1002/mdc3.12000] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 12/19/2013] [Accepted: 12/19/2013] [Indexed: 12/13/2022] Open
Abstract
Over the last 16 years, insights in clinical and genetic characteristics of Parkinson's disease (PD) have increased substantially. We summarize the clinical, genetic, and pathological findings of autosomal dominant PD linked to mutations in SNCA, leucine-rich repeat kinase 2, vacuolar protein sorting-35, and eukaryotic translation initiation factor 4 gamma 1 and autosomal recessive PD linked to parkin,PINK1, and DJ-1, as well as autosomal recessive complicated parkinsonian syndromes caused by mutations in ATP13A2,FBXO7,PLA2G6,SYNJ1, and DNAJC6. We also review the advances in high- and low-risk genetic susceptibility factors and present multisystem disorders that may present with parkinsonism as the major clinical feature and provide recommendations for prioritization of genetic testing. Finally, we consider the challenges of future genetic research in PD.
Collapse
Affiliation(s)
- Una-Marie Sheerin
- Department of Molecular Neuroscience UCL Institute of Neurology University College London London United Kingdom
| | - Henry Houlden
- Department of Molecular Neuroscience UCL Institute of Neurology University College London London United Kingdom
| | - Nicholas W Wood
- UCL Department of Molecular Neuroscience and UCL Genetics Institute University College London London United Kingdom
| |
Collapse
|
91
|
Labbé C, Ross OA. Association studies of sporadic Parkinson's disease in the genomic era. Curr Genomics 2014; 15:2-10. [PMID: 24653658 PMCID: PMC3958956 DOI: 10.2174/1389202914666131210212745] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 09/09/2013] [Accepted: 10/23/2013] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease is a common age-related progressive neurodegenerative disorder. Over the last 10 years, advances have been made in our understanding of the etiology of the disease with the greatest insights perhaps coming from genetic studies, including genome-wide association approaches. These large scale studies allow the identification of genomic regions harboring common variants associated to disease risk. Since the first genome-wide association study on sporadic Parkinson's disease performed in 2005, improvements in study design, including the advent of meta-analyses, have allowed the identification of ~21 susceptibility loci. The first loci to be nominated were previously associated to familial PD (SNCA, MAPT, LRRK2) and these have been extensively replicated. For other more recently identified loci (SREBF1, SCARB2, RIT2) independent replication is still warranted. Cumulative risk estimates of associated variants suggest that more loci are still to be discovered. Additional association studies combined with deep re-sequencing of known genome-wide association study loci are necessary to identify the functional variants that drive disease risk. As each of these associated genes and variants are identified they will give insight into the biological pathways involved the etiology of Parkinson's disease. This will ultimately lead to the identification of molecules that can be used as biomarkers for diagnosis and as targets for the development of better, personalized treatment.
Collapse
Affiliation(s)
- Catherine Labbé
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Owen A Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| |
Collapse
|
92
|
Clarimón J, Kulisevsky J. Parkinson's disease: from genetics to clinical practice. Curr Genomics 2014; 14:560-7. [PMID: 24532987 PMCID: PMC3924250 DOI: 10.2174/1389202914666131210212305] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 09/14/2013] [Accepted: 10/31/2013] [Indexed: 12/13/2022] Open
Abstract
Breakthroughs in genetics over the last decade have radically advanced our understanding of the etiological basis
of Parkinson's disease (PD). Although much research remains to be done, the main genetic causes of this neurodegenerative
disorder are now partially unraveled, allowing us to feel more confident that our knowledge about the genetic architecture
of PD will continue to increase exponentially. How and when these discoveries will be introduced into general
clinical practice, however, remains uncertain. In this review, we provide a general summary of the progress in the genetics
of PD and discuss how this knowledge will contribute to the diagnosis and clinical management of patients with, or at risk
of this disorder.
Collapse
Affiliation(s)
- Jordi Clarimón
- Neurology Department, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Jaime Kulisevsky
- Neurology Department, Institut d'Investigacions Biomèdiques Sant Pau, Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
93
|
Mellick GD, Silburn PA, Sutherland GT, Siebert GA. Exploiting the potential of molecular profiling in Parkinson’s disease: current practice and future probabilities. Expert Rev Mol Diagn 2014; 10:1035-50. [PMID: 21080820 DOI: 10.1586/erm.10.86] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- George D Mellick
- Eskitis Institute for Cell & Molecular Therapies, School of Biomolecular & Physical Sciences, Griffith University, Brisbane, QLD 4111, Australia.
| | | | | | | |
Collapse
|
94
|
Jiménez-Jiménez FJ, García-Martín E, Alonso-Navarro H, Agúndez JA. PITX3 and Risk for Parkinson's Disease: A Systematic Review and Meta-Analysis. Eur Neurol 2013; 71:49-56. [DOI: 10.1159/000353981] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 06/23/2013] [Indexed: 12/13/2022]
|
95
|
Soto-Ortolaza AI, Heckman MG, Labbé C, Serie DJ, Puschmann A, Rayaprolu S, Strongosky A, Boczarska-Jedynak M, Opala G, Krygowska-Wajs A, Barcikowska M, Czyzewski K, Lynch T, Uitti RJ, Wszolek ZK, Ross OA. GWAS risk factors in Parkinson's disease: LRRK2 coding variation and genetic interaction with PARK16. AMERICAN JOURNAL OF NEURODEGENERATIVE DISEASE 2013; 2:287-299. [PMID: 24319646 PMCID: PMC3852568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 10/15/2013] [Indexed: 06/03/2023]
Abstract
Parkinson's disease (PD) is a multifactorial movement disorder characterized by progressive neurodegeneration. Genome-wide association studies (GWAS) have nominated over fifteen distinct loci associated with risk of PD, however the biological mechanisms by which these loci influence disease risk are mostly unknown. GWAS are only the first step in the identification of disease genes: the specific causal variants responsible for the risk within the associated loci and the interactions between them must be identified to fully comprehend their impact on the development of PD. In the present study, we first attempted to replicate the association signals of 17 PD GWAS loci in our series of 1381 patients with PD and 1328 controls. BST1, SNCA, HLA-DRA, CCDC62/HIP1R and MAPT all showed a significant association with PD under different models of inheritance and LRRK2 showed a suggestive association. We then examined the role of coding LRRK2 variants in the GWAS association signal for that gene. The previously identified LRRK2 risk mutant p.M1646T and protective haplotype p.N551K-R1398H-K1423K did not explain the association signal of LRRK2 in our series. Finally, we investigated the gene-gene interaction between PARK16 and LRRK2 that has previously been proposed. We observed no interaction between PARK16 and LRRK2 GWAS variants, but did observe a non-significant trend toward interaction between PARK16 and LRRK2 variants within the protective haplotype. Identification of causal variants and the interactions between them is the crucial next step in making biological sense of the massive amount of data generated by GWAS studies. Future studies combining larger sample sizes will undoubtedly shed light on the complex molecular interplay leading to the development of PD.
Collapse
Affiliation(s)
| | | | - Catherine Labbé
- Department of Neuroscience, Mayo ClinicJacksonville, Florida, USA
| | - Daniel J Serie
- Section of Biostatistics, Mayo ClinicJacksonville, Florida, USA
| | - Andreas Puschmann
- Department of Neuroscience, Mayo ClinicJacksonville, Florida, USA
- Department of Neurology, Lund UniversitySweden
| | - Sruti Rayaprolu
- Department of Neuroscience, Mayo ClinicJacksonville, Florida, USA
| | | | | | - Grzegorz Opala
- Department of Neurology, Medical University of SilesiaKatowice, Poland
| | | | - Maria Barcikowska
- Department of Neurodegenerative Disorders, Medical Research Centre, Polish Academy of SciencesWarsaw, Poland
| | - Krzysztof Czyzewski
- Department of Neurology, Central Hospital of The Ministry of Interior and AdministrationWarsaw, Poland
| | - Timothy Lynch
- Dublin Neurological Institute at The Mater Misericordiae University Hospital, Conway Institute of Biomolecular & Biomedical Research, University College DublinIreland
| | - Ryan J Uitti
- Department of Neurology, Mayo ClinicJacksonville, Florida, USA
| | | | - Owen A Ross
- Department of Neuroscience, Mayo ClinicJacksonville, Florida, USA
| |
Collapse
|
96
|
Zhu B, Chen C, Xue G, Moyzis RK, Dong Q, Chen C, Li J, He Q, Lei X, Wang Y, Lin C. The SEMA5A gene is associated with hippocampal volume, and their interaction is associated with performance on Raven's Progressive Matrices. Neuroimage 2013; 88:181-7. [PMID: 24291503 DOI: 10.1016/j.neuroimage.2013.11.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Revised: 11/13/2013] [Accepted: 11/17/2013] [Indexed: 12/11/2022] Open
Abstract
The Allen Brain Atlas shows that the semaphorin 5A (SEMA5A) gene, which encodes an important protein for neurogenesis and neuronal apoptosis, is predominantly expressed in the human hippocampus. Structural and functional neuroimaging studies have further shown that the hippocampus plays an important role in the performance on Raven's Progressive Matrices (RPM), a measure of reasoning ability and general fluid intelligence. Thus far, however, no study has examined the relationships between the SEMA5A gene polymorphism, hippocampal volume, and RPM performance. The current study collected both structural MRI, genetic, and behavioral data in 329 healthy Chinese adults, and examined associations between SEMA5A variants, hippocampal volume, and performance on RAPM (the advanced form of RPM). After controlling for intracranial volume (ICV), sex, and age, SEMA5A genetic polymorphism at the SNP rs42352 had the strongest association with hippocampal volume (p=0.00000552 and 0.000103 for right and left hippocampal volumes, respectively), with TT homozygotes having higher hippocampal volume than the other genotypes. Furthermore, there was a high correlation between right hippocampal volume and RAPM performance (r=0.42, p=0.0000509) for SEMA5A rs42352 TT homozygotes. This study provides the first evidence for the involvement of the SEMA5A gene in hippocampal structure and their interaction on RAPM performance. Future studies of the hippocampus-RPM associations should consider genetic factors as potential moderators.
Collapse
Affiliation(s)
- Bi Zhu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China; Center for Collaboration and Innovation in Brain and Learning Sciences, Beijing Normal University, Beijing, China
| | - Chuansheng Chen
- Department of Psychology and Social Behavior, University of California, Irvine, CA, USA.
| | - Gui Xue
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China; Center for Collaboration and Innovation in Brain and Learning Sciences, Beijing Normal University, Beijing, China
| | - Robert K Moyzis
- Department of Biological Chemistry and Institute of Genomics and Bioinformatics, University of California, Irvine, CA, USA
| | - Qi Dong
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China; Center for Collaboration and Innovation in Brain and Learning Sciences, Beijing Normal University, Beijing, China.
| | - Chunhui Chen
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Jin Li
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Qinghua He
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Xuemei Lei
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Yunxin Wang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Chongde Lin
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| |
Collapse
|
97
|
Li NN, Tan EK, Chang XL, Mao XY, Zhang JH, Zhao DM, Liao Q, Yu WJ, Peng R. Genetic association study between STK39 and CCDC62/HIP1R and Parkinson's disease. PLoS One 2013; 8:e79211. [PMID: 24312176 PMCID: PMC3842305 DOI: 10.1371/journal.pone.0079211] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2013] [Accepted: 09/20/2013] [Indexed: 02/05/2023] Open
Abstract
Background The first large-scale meta-analysis of published genome-wide association studies (GWAS) in Parkinson’s disease (PD) identified 5 new genetic loci (ACMSD, STK39, MCCC1/LAMP3, SYT11, and CCDC62/HIP1R). Very recently, a large-scale replication and heterogeneity study also reported that STK39 and CCDC62/HIP1R increased risk of PD in Asian and Caucasian populations. However, their roles still remain unclear in a Han Chinese population from mainland China. Methods We examined genetic associations of STK39 rs2102808 and CCDC62/HIP1R rs12817488 with PD susceptibility in a Han Chinese population of 783 PD patients and 725 controls. We also performed further stratified analyses by the age of onset and accomplished in-depth clinical characteristics analyses between the different genotypes for each locus. Results No significant differences were observed in the minor allele frequency (MAF) among cases and controls at the two loci (STK39 rs2102808: OR = 1.06, 95% CI = 0.91, 1.23, P = 0.467; CCDC62/HIP1R rs12817488: OR = 0.88, 95% CI = 0.76, 1.01, P = 0.072). Subgroup analyses by the age of onset also showed no significant differences among different subgroups of the two loci. In addition, minor allele carriers cannot be distinguished from non-carriers based on their clinical features at the two loci. Conclusions We are unable to demonstrate the association between STK39 and CCDC62/HIP1R and PD susceptibility in a Han Chinese population from mainland China. Additional replication studies in other populations and functional studies are warranted to better validate the role of the two new loci in PD risk.
Collapse
Affiliation(s)
- Nan-Nan Li
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Eng-King Tan
- Department of Neurology, Singapore General Hospital, National Neuroscience Institute, Singapore, Singapore
- Duke–NUS Graduate Medical School, Singapore, Singapore
| | - Xue-Li Chang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xue-Ye Mao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jin-Hong Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Internal Medicine, Wangjiang Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dong-Mei Zhao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiao Liao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wen-Juan Yu
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong Peng
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- * E-mail:
| |
Collapse
|
98
|
Schulte EC, Stahl I, Czamara D, Ellwanger DC, Eck S, Graf E, Mollenhauer B, Zimprich A, Lichtner P, Haubenberger D, Pirker W, Brücke T, Bereznai B, Molnar MJ, Peters A, Gieger C, Müller-Myhsok B, Trenkwalder C, Winkelmann J. Rare variants in PLXNA4 and Parkinson's disease. PLoS One 2013; 8:e79145. [PMID: 24244438 PMCID: PMC3823607 DOI: 10.1371/journal.pone.0079145] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 09/18/2013] [Indexed: 11/18/2022] Open
Abstract
Approximately 20% of individuals with Parkinson's disease (PD) report a positive family history. Yet, a large portion of causal and disease-modifying variants is still unknown. We used exome sequencing in two affected individuals from a family with late-onset familial PD followed by frequency assessment in 975 PD cases and 1014 ethnically-matched controls and linkage analysis to identify potentially causal variants. Based on the predicted penetrance and the frequencies, a variant in PLXNA4 proved to be the best candidate and PLXNA4 was screened for additional variants in 862 PD cases and 940 controls, revealing an excess of rare non-synonymous coding variants in PLXNA4 in individuals with PD. Although we cannot conclude that the variant in PLXNA4 is indeed the causative variant, these findings are interesting in the light of a surfacing role of axonal guidance mechanisms in neurodegenerative disorders but, at the same time, highlight the difficulties encountered in the study of rare variants identified by next-generation sequencing in diseases with autosomal dominant or complex patterns of inheritance.
Collapse
Affiliation(s)
- Eva C. Schulte
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität, München, Munich, Germany
- Institut für Humangenetik, Helmholtz Zentrum München, Munich, Germany
| | - Immanuel Stahl
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität, München, Munich, Germany
- Institut für Humangenetik, Helmholtz Zentrum München, Munich, Germany
| | - Darina Czamara
- Max-Planck Institut für Psychiatrie, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Daniel C. Ellwanger
- Chair for Genome-Oriented Bioinformatics, Technische Universität München, Life and Food Science Center Weihenstephan, Freising-Weihenstephan, Germany
| | - Sebastian Eck
- Institut für Humangenetik, Helmholtz Zentrum München, Munich, Germany
| | - Elisabeth Graf
- Institut für Humangenetik, Helmholtz Zentrum München, Munich, Germany
| | - Brit Mollenhauer
- Paracelsus Elena Klinik, Kassel, Germany
- Neurochirurgische Klinik, Georg August Universität, Göttingen, Germany
| | | | - Peter Lichtner
- Institut für Humangenetik, Helmholtz Zentrum München, Munich, Germany
- Institut für Humangenetik, Technische Universität München, Munich, Germany
| | | | - Walter Pirker
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Brücke
- Department of Neurology, Wilhelminenspital, Vienna, Austria
| | - Benjamin Bereznai
- Center for Molecular Neurology, Department of Neurology, Semmelweis University, Budapest, Hungary
| | - Maria J. Molnar
- Center for Molecular Neurology, Department of Neurology, Semmelweis University, Budapest, Hungary
| | - Annette Peters
- Institute for Epidemiology II, Helmholtz Zentrum München, Munich, Germany
| | - Christian Gieger
- Institute for Genetic Epidemiology, Helmholtz Zentrum München, Munich, Germany
| | - Bertram Müller-Myhsok
- Max-Planck Institut für Psychiatrie, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Claudia Trenkwalder
- Paracelsus Elena Klinik, Kassel, Germany
- Neurochirurgische Klinik, Georg August Universität, Göttingen, Germany
| | - Juliane Winkelmann
- Neurologische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität, München, Munich, Germany
- Institut für Humangenetik, Helmholtz Zentrum München, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institut für Humangenetik, Technische Universität München, Munich, Germany
- Department of Neurology and Neurosciences, Stanford University, Palo Alto, California, United States of America
- * E-mail:
| |
Collapse
|
99
|
Grewal A, Stephan DA. Diagnostics for personalized medicine: what will change in the era of large-scale genomics studies? Per Med 2013; 10:835-848. [DOI: 10.2217/pme.13.82] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The era of personalized medicine is upon us and it is being fueled by large available data sets of many types that are setting the foundation for the development of more precise diagnostic tools and targeted therapies, which are improving patient outcomes. Technology innovation and concomitant price decreases in molecular scanning technologies are at the heart of this change, both accelerating at a rate that has exceeded Moore’s law. This technology trend is enabling the research community to generate, and make publicly available, massive amounts of genomic data. These data come in the form not only of contextual information about the structure and function of the genome, but also in the form of variants that are correlated with human disease. Coupled with this molecular information, we are making dramatic inroads into capturing and making available high-resolution phenotypic and environmental exposure data through both incentives to physicians to migrate electronic medical records and to adoption of consumer-facing data collection and aggregation technologies. These large-scale genomic, environmental and phenotypic data together allow us to provide a multitude of new diagnostic correlations across the spectrum of possible clinical indications. To fully leverage the data foundation that will lead us to precise diagnostics and truly move the needle in outcome improvement, we need to achieve a culture shift as to how to apply this new personalized and probabilistic diagnostic information to better practice the art of medicine.
Collapse
Affiliation(s)
- Anoop Grewal
- Silicon Valley Biosystems, 950 Tower Lane, 11th Floor, Foster City, CA 94404, USA
| | - Dietrich A Stephan
- Silicon Valley Biosystems, 950 Tower Lane, 11th Floor, Foster City, CA 94404, USA
| |
Collapse
|
100
|
Davis MF, Cummings AC, D'Aoust LN, Jiang L, Velez Edwards DR, Laux R, Reinhart-Mercer L, Fuzzell D, Scott WK, Pericak-Vance MA, Lee SL, Haines JL. Parkinson disease loci in the mid-western Amish. Hum Genet 2013; 132:1213-21. [PMID: 23793441 PMCID: PMC3797866 DOI: 10.1007/s00439-013-1316-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 05/16/2013] [Indexed: 11/26/2022]
Abstract
Previous evidence has shown that Parkinson disease (PD) has a heritable component, but only a small proportion of the total genetic contribution to PD has been identified. Genetic heterogeneity complicates the verification of proposed PD genes and the identification of new PD susceptibility genes. Our approach to overcome the problem of heterogeneity is to study a population isolate, the mid-western Amish communities of Indiana and Ohio. We performed genome-wide association and linkage analyses on 798 individuals (31 with PD), who are part of a 4,998 member pedigree. Through these analyses, we identified a region on chromosome 5q31.3 that shows evidence of association (p value < 1 × 10(-4)) and linkage (multipoint HLOD = 3.77). We also found further evidence of linkage on chromosomes 6 and 10 (multipoint HLOD 4.02 and 4.35 respectively). These data suggest that locus heterogeneity, even within the Amish, may be more extensive than previously appreciated.
Collapse
Affiliation(s)
- M F Davis
- Center for Human Genetics Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|