1
|
Yang Y, Zhou ZD, Yi L, Tan BJW, Tan EK. Interaction between caffeine consumption & genetic susceptibility in Parkinson's disease: A systematic review. Ageing Res Rev 2024; 99:102381. [PMID: 38914264 DOI: 10.1016/j.arr.2024.102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/03/2024] [Accepted: 06/14/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND Caffeine is one of the most consumed psychoactive substances globally. Caffeine-gene interactions in Parkinson's disease (PD) has not been systematically examined. OBJECTIVES To conduct a systematic review on the interaction between caffeine consumption and genetic susceptibility to PD. METHODOLOGY We conducted PubMed and Embase search using terms "Genetic association studies", "Caffeine", "polymorphism" and "Parkinson's disease", from inception till 2023. Of the initial 2391 studies, 21 case-control studies were included. The demographic, genetic and clinical data were extracted and analyzed. RESULTS We identified 21 studies which involved a total of 607,074 study subjects and 17 gene loci (SNCA, MAPT, HLA-DRA, NOS1, NOS3, GBA, ApoE, BST1, ESR2, NAT2, SLC2A13, LRRK2, NOS2A, GRIN2A, CYP1A2, ESR1, ADORA2A) have been investigated for the effect of gene-caffeine interaction and PD risk. The genes were identified through PD GWAS or involved in caffeine or related metabolism pathways. Based on the genetic association and interaction studies, only MAPT, SLC2A13, LRRK2, ApoE, NOS2A, GRIN2A, CYP1A2, and ADORA2A have been shown by at least one study to have a positive caffeine-gene interaction influencing the risk of PD. CONCLUSION Studies have shown an interaction between caffeine with genetic variants of MAPT, SLC2A13, LRRK2, ApoE, NOS2A, GRIN2A, CYP1A2, and ADORA2A in modulating the risk of PD. Due to the potential limitations of these discovery/pilot studies, further independent replication studies are needed. Better designed genetic association studies in multi-ancestry and admixed cohorts to identify potential shared or unique multivariate gene-environmental interactions, as well as functional studies of gene-caffeine interactions will be useful.
Collapse
Affiliation(s)
- Yujuan Yang
- Department of Neurology, National Neuroscience Institute, Singapore; Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore.
| | - Zhi Dong Zhou
- Department of Neurology, National Neuroscience Institute, Singapore; Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore.
| | - Lingxiao Yi
- Department of Neurology, National Neuroscience Institute, Singapore.
| | | | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore; Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore.
| |
Collapse
|
2
|
Cherian A, K P D, Vijayaraghavan A. Parkinson's disease - genetic cause. Curr Opin Neurol 2023; Publish Ahead of Print:00019052-990000000-00070. [PMID: 37366140 DOI: 10.1097/wco.0000000000001167] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
PURPOSE OF REVIEW Our knowledge of the genetic architecture underlying Parkinson's disease has vastly improved in the past quarter century. About 5-10% of all patients suffer from a monogenic form of Parkinson's disease. RECENT FINDINGS Mutations in autosomal dominant genes (e.g. SNCA, LRRK2, VPS35) or autosomal recessive genes (e.g. PRKN, PINK1, DJ-1) can cause genetic Parkinson's disease. Recessive DNAJC6 mutations can present predominantly as atypical parkinsonism, but also rarely as typical Parkinson's disease. Majority of Parkinson's disease is genetically complex. Mutation in RIC3, a chaperone of neuronal nicotinic acetylcholine receptor subunit α-7 (CHRNA7), provides strong evidence for the role of cholinergic pathway, for the first time, in cause of Parkinson's disease. X-linked parkinsonism manifests at a young age accompanied by many (atypical) features such as intellectual disability, spasticity, seizures, myoclonus, dystonia, and have poor response to levodopa. SUMMARY This review article aims to provide a comprehensive overview on Parkinson's disease genetics. MAPT, which encodes the microtubule associated protein tau, TMEM230, LRP10, NUS1 and ARSA are the five new putative disease-causing genes in Parkinson's disease. The validation of novel genes and its association with Parkinson's disease remains extremely challenging, as genetically affected families are sparse and globally widespread. In the near future, genetic discoveries in Parkinson's disease will influence our ability to predict and prognosticate the disease, help in defining etiological subtypes that are critical in implementation of precision medicine.
Collapse
Affiliation(s)
- Ajith Cherian
- Department of Neurology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India
| | | | | |
Collapse
|
3
|
Cherian A, Divya KP. Genetics of Parkinson's disease. Acta Neurol Belg 2020; 120:1297-1305. [PMID: 32813147 DOI: 10.1007/s13760-020-01473-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/11/2020] [Indexed: 12/21/2022]
Abstract
Less than a quarter century after the discovery of SNCA as the first attributable gene in Parkinson's disease (PD), our knowledge of the genetic architecture underlying this disease has improved by leaps and bounds. About 5-10% of all patients suffer from a monogenic form of PD where mutations in autosomal-dominant (AD) genes-SNCA, LRRK2, and VPS35 and autosomal recessive (AR) genes-PINK1, DJ-1, and Parkin cause the disease. Whole-exome sequencing has described AR DNAJC6 mutations not only in predominantly atypical, but also in patients with typical PD. Majority of PD is genetically complex, caused by the combination of common genetic variants in concert with environmental factors. Genome-wide association studies have identified twenty six PD risk loci till date; however, these show only moderate effects on the risk for PD. The validation of novel genes and its association with PD remains extremely challenging as families harboring rare genetic variants are sparse and globally widespread. This review article aims to provide a comprehensive overview on PD genetics.
Collapse
Affiliation(s)
- Ajith Cherian
- Department of Neurology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India, 695011
| | - K P Divya
- Department of Neurology, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, Kerala, India, 695011.
| |
Collapse
|
4
|
Lill CM. WITHDRAWN: Genetics of Parkinson's disease. Mol Cell Probes 2020:101471. [PMID: 31978549 DOI: 10.1016/j.mcp.2019.101471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 10/17/2019] [Indexed: 11/25/2022]
Abstract
The Publisher regrets that this article is an accidental duplication of an article that has already been published, DOI of original article: https://doi.org/10.1016/j.mcp.2016.11.001. The duplicate article has therefore been withdrawn. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
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
|
5
|
Nepal G, Rehrig JH, Ojha R. Glutamate ionotropic receptor NMDA type subunit 2A ( GRIN2A) gene polymorphism (rs4998386) and Parkinson's disease susceptibility: A meta-analysis. Aging Med (Milton) 2019; 2:174-183. [PMID: 31942532 PMCID: PMC6880709 DOI: 10.1002/agm2.12075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE Dopaminergic neuronal degeneration seen in Parkinson's disease (PD) might result from a single nucleotide polymorphism (SNP) in the glutamate ionotropic receptor NMDA type subunit 2A (GRIN2A) gene. We thus performed a meta-analysis exploring the relationship between the rs4998386 SNP of the GRIN2A gene and PD susceptibility. METHODS We searched PubMed, EMBASE, Web of Science, Google Scholar, and China National Knowledge Infrastructure for studies published between January 2005 and January 2019. The association between the rs4998386 polymorphism and PD susceptibility was evaluated by calculating the pooled odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS Meta-analysis results did not show a significant association between the rs4998386 polymorphism of the GRIN2A gene and PD susceptibility when assuming an allelic model (OR, 0.90; 95% CI, 0.76-1.07; P = .22; I 2 = 53%), a dominant model (OR, 0.96; 95% CI, 0.82-1.12; P = .62; I 2 = 64%), or a recessive model (OR, 1.14; 95% CI, 0.93-1.38; P = .22; I 2 = 0%). CONCLUSION Our meta-analysis found that the rs4998386 polymorphism of the GRIN2A gene is not associated with risk of PD in either Europeans or white Americans. However, large sample studies with different ethnicities should be conducted to establish the role of the rs4998386 polymorphism in PD pathophysiology.
Collapse
Affiliation(s)
- Gaurav Nepal
- Tribhuvan University Institute of MedicineKathmanduNepal
| | | | - Rajeev Ojha
- Department of NeurologyTribhuvan University Institute of MedicineKathmanduNepal
| |
Collapse
|
6
|
Dunn AR, O'Connell KMS, Kaczorowski CC. Gene-by-environment interactions in Alzheimer's disease and Parkinson's disease. Neurosci Biobehav Rev 2019; 103:73-80. [PMID: 31207254 PMCID: PMC6700747 DOI: 10.1016/j.neubiorev.2019.06.018] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022]
Abstract
Diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD) arise from complex interactions of genetic and environmental factors, with genetic variants regulating individual responses to environmental exposures (i.e. gene-by-environment interactions). Identifying gene-by-environment interactions will be critical to fully understanding disease mechanisms and developing personalized therapeutics, though these interactions are still poorly understood and largely under-studied. Candidate gene approaches have shown that known disease risk variants often regulate response to environmental factors. However, recent improvements in exposome- and genome-wide association and interaction studies in humans and mice are enabling discovery of novel genetic variants and pathways that predict response to a variety of environmental factors. Here, we highlight recent approaches and ongoing developments in human and rodent studies to identify genetic modulators of environmental factors using AD and PD as exemplars. Identifying gene-by-environment interactions in disease will be critical to developing personalized intervention strategies and will pave the way for precision medicine.
Collapse
Affiliation(s)
- Amy R Dunn
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA.
| | | | | |
Collapse
|
7
|
Marras C, Canning CG, Goldman SM. Environment, lifestyle, and Parkinson's disease: Implications for prevention in the next decade. Mov Disord 2019; 34:801-811. [DOI: 10.1002/mds.27720] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/17/2019] [Accepted: 04/26/2019] [Indexed: 12/14/2022] Open
Affiliation(s)
- Connie Marras
- The Edmond J. Safra Program in Parkinson's DiseaseToronto Western Hospital Toronto Ontario Canada
| | - Colleen G. Canning
- Discipline of Physiotherapy, Faculty of Health SciencesThe University of Sydney Sydney Australia
| | - Samuel M. Goldman
- School of MedicineUniversity of California–San Francisco San Francisco California USA
- Division of Occupational and Environmental MedicineSan Francisco Veterans Affairs Health Care System San Francisco California USA
| |
Collapse
|
8
|
Nehlig A. Interindividual Differences in Caffeine Metabolism and Factors Driving Caffeine Consumption. Pharmacol Rev 2018. [PMID: 29514871 DOI: 10.1124/pr.117.014407] [Citation(s) in RCA: 299] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Most individuals adjust their caffeine intake according to the objective and subjective effects induced by the methylxanthine. However, to reach the desired effects, the quantity of caffeine consumed varies largely among individuals. It has been known for decades that the metabolism, clearance, and pharmacokinetics of caffeine is affected by many factors such as age, sex and hormones, liver disease, obesity, smoking, and diet. Caffeine also interacts with many medications. All these factors will be reviewed in the present document and discussed in light of the most recent data concerning the genetic variability affecting caffeine levels and effects at the pharmacokinetic and pharmacodynamic levels that both critically drive the level of caffeine consumption. The pharmacokinetics of caffeine are highly variable among individuals due to a polymorphism at the level of the CYP1A2 isoform of cytochrome P450, which metabolizes 95% of the caffeine ingested. Moreover there is a polymorphism at the level of another critical enzyme, N-acetyltransferase 2. At the pharmacodynamic level, there are several polymorphisms at the main brain target of caffeine, the adenosine A2A receptor or ADORA2. Genetic studies, including genome-wide association studies, identified several loci critically involved in caffeine consumption and its consequences on sleep, anxiety, and potentially in neurodegenerative and psychiatric diseases. We start reaching a better picture on how a multiplicity of biologic mechanisms seems to drive the levels of caffeine consumption, although much more knowledge is still required to understand caffeine consumption and effects on body functions.
Collapse
Affiliation(s)
- Astrid Nehlig
- INSERM U 1129, Pediatric Neurology, Necker-Enfants Malades Hospital, University of Paris Descartes, Inserm U1129, Paris, France
| |
Collapse
|
9
|
Pihlstrøm L, Wiethoff S, Houlden H. Genetics of neurodegenerative diseases: an overview. HANDBOOK OF CLINICAL NEUROLOGY 2018; 145:309-323. [PMID: 28987179 DOI: 10.1016/b978-0-12-802395-2.00022-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Genetic factors are central to the etiology of neurodegeneration, both as monogenic causes of heritable disease and as modifiers of susceptibility to complex, sporadic disorders. Over the last two decades, the identification of disease genes and risk loci has led to some of the greatest advances in medicine and invaluable insights into pathogenic mechanisms and disease pathways. Large-scale research efforts, novel study designs, and advances in methodology are rapidly expanding our understanding of the genome and the genetic architecture of neurodegenerative disease. Here, we review major developments in the field to date, highlighting overarching historic trends and general insights. Monogenic neurodegenerative diseases are discussed from the perspectives of both rare Mendelian forms of common disorders, such as Alzheimer disease and Parkinson disease, and heterogeneous heritable conditions, including ataxias and spastic paraplegias. Next, we summarize the experiences from investigations of complex neurodegenerative disorders, including genomewide association studies. In the final section, we reflect upon the limitations of current findings and outline important future directions. Genetics plays an essential role in translational research, ultimately aiming to develop novel disease-modifying therapies for neurodegenerative disorders. We anticipate that individual genetic profiling will also be increasingly relevant in a clinical context, with implications for patient care in line with the proposed ideal of personalized medicine.
Collapse
Affiliation(s)
- Lasse Pihlstrøm
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway; UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Sarah Wiethoff
- UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, United Kingdom; Center for Neurology and Hertie Institute for Clinical Brain Research, Eberhard-Karls-University, Tübingen, Germany
| | - Henry Houlden
- UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, United Kingdom.
| |
Collapse
|
10
|
Kim IY, O'Reilly ÉJ, Hughes KC, Gao X, Schwarzschild MA, McCullough ML, Hannan MT, Betensky RA, Ascherio A. Interaction between caffeine and polymorphisms of glutamate ionotropic receptor NMDA type subunit 2A (GRIN2A) and cytochrome P450 1A2 (CYP1A2) on Parkinson's disease risk. Mov Disord 2018; 33:414-420. [PMID: 29318639 DOI: 10.1002/mds.27279] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/20/2017] [Accepted: 11/26/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Caffeine intake has been inversely associated with Parkinson's disease (PD) risk. This relationship may be modified by polymorphisms of glutamate ionotropic receptor NMDA type subunit 2A (GRIN2A) and cytochrome P450 1A2 (CYP1A2), but the results of previous studies have been inconsistent. METHOD We examined the interaction of caffeine intake with GRIN2A-rs4998386 and CYP1A2-rs762551 polymorphisms in influencing PD risk among 829 incident cases of PD and 2,754 matched controls selected among participants in the following 3 large prospective ongoing cohorts: the Nurses' Health Study, the Health Professionals' Follow-up Study, and the Cancer Prevention Study II Nutrition Cohort. Matching factors included cohort, birth year, source of DNA, date of DNA collection, and race. Relative risks and 95% confidence intervals were estimated using conditional logistic models. Interactions were tested both on the multiplicative scale and on the additive scale. RESULTS Overall, caffeine intake was associated with a lower PD risk (adjusted relative risk for highest versus lowest tertile = 0.70; 95% confidence interval, 0.57-0.86; p < .001). In analyses stratified by the GRIN2A-rs4998386 genotype, the multivariable-adjusted relative risk of PD comparing the highest to the lowest tertile of caffeine was 0.69 (95% confidence interval, 0.55-0.88; p < .01) among individuals homozygous for the C allele, and 0.85 (95% confidence interval, 0.55-1.32; p = .47; pRERI = .43) among carriers for the T allele. Interactions between caffeine and GRIN2A were not significant in either the multiplicative or additive scales. We also did not observe significant interactions for CYP1A2-rs762551 and incident PD risk. CONCLUSION Our findings do not support the hypothesis of an interaction between the GRIN2A-rs4998386 or CYP1A2-rs762551 polymorphism and caffeine intake in determining PD risk. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Iris Y Kim
- Department of Epidemiology, Harvard T. H. School Chan School of Public Health, Boston, Massachusetts, USA
| | - Éilis J O'Reilly
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA.,School of Public Health, University College Cork, Cork, Ireland
| | - Katherine C Hughes
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Xiang Gao
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Michael A Schwarzschild
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marjorie L McCullough
- Department of Epidemiology, Harvard T. H. School Chan School of Public Health, Boston, Massachusetts, USA.,Epidemiology Research Program, American Cancer Society, Atlanta, Georgia, USA
| | - Marian T Hannan
- The Institute for Aging Research, Hebrew Senior Life, Boston, Massachusetts, USA.,Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca A Betensky
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Alberto Ascherio
- Department of Epidemiology, Harvard T. H. School Chan School of Public Health, Boston, Massachusetts, USA.,Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Chen H, Ritz B. The Search for Environmental Causes of Parkinson's Disease: Moving Forward. JOURNAL OF PARKINSON'S DISEASE 2018; 8:S9-S17. [PMID: 30584168 PMCID: PMC6311360 DOI: 10.3233/jpd-181493] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/15/2018] [Indexed: 12/12/2022]
Abstract
It is widely believed that environmental exposures contribute to the vast majority of late-onset sporadic Parkinson's disease (PD), alone or via interactions with genetic factors. The search for environmental causes of PD has however been hampered by lack of understanding the prodromal phase of PD development and the difficulties in exposure assessment during this prolonged period. On the other hand, the existence of this prodromal period, along with an increasingly better understanding of PD prodromal symptoms, provides an exciting opportunity to identify environmental factors that initiate PD pathogenesis and/or modify its progression. For prevention efforts, this prodromal stage is of a major interest. Targeting factors that enter the body via the nose or gut has become even more important since the discovery of α-synuclein aggregates in the enteric and olfactory nervous systems. In this paper, we speculate about novel research hypotheses and approaches that may help us better define the role of environment in PD etiology, especially during its extended and complex prodromal phase.
Collapse
Affiliation(s)
- Honglei Chen
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Beate Ritz
- Department of Epidemiology and Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
12
|
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and has a growing socioeconomic impact due to demographic changes in the industrial nations. There are several forms of PD, a fraction of which (<5%) are monogenic, i. e. caused by mutations in single genes. At present, six genes have been established for the clinically classical form of parkinsonism including three autosomal dominantly (SNCA, LRRK2, VPS35) and three autosomal recessively inherited ones (Parkin, PINK1, DJ-1). In addition, there are a plethora of genes causing atypical forms of parkinsonism. In contrast, idiopathic PD is of a multifactorial nature. Genome-wide association studies have established a total of 26 genetic loci for this form of the disease; however, for most of these loci the underlying functional genetic variants have not yet been identified and the respective disease mechanisms remain unresolved. Furthermore, there are a number of environmental and life style factors that are associated with idiopathic PD. Exposure to pesticides and possibly a history of head trauma represent genuine risk factors. Other PD-associated factors, such as smoking and intake of coffee and alcohol may not represent risk factors per se and the cause-effect relationship has not yet been elucidated for most of these factors. A patient with a positive family history and/or an early age of disease onset should undergo counseling with respect to a possible monogenic form of the disease. Disease prediction based on genetic, environmental and life style factors is not yet possible for idiopathic PD and potential gene-specific therapies are currently in the development or early testing phase.
Collapse
Affiliation(s)
- C M Lill
- Institut für Neurogenetik, Universitätsklinikum Schleswig Holstein, Campus Lübeck, Universität zu Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Deutschland
| | - C Klein
- Institut für Neurogenetik, Universitätsklinikum Schleswig Holstein, Campus Lübeck, Universität zu Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Deutschland.
| |
Collapse
|
13
|
Chuang YH, Lee PC, Vlaar T, Mulot C, Loriot MA, Hansen J, Lill CM, Ritz B, Elbaz A. Pooled analysis of the HLA-DRB1 by smoking interaction in Parkinson disease. Ann Neurol 2017; 82:655-664. [PMID: 28981958 DOI: 10.1002/ana.25065] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 10/02/2017] [Accepted: 10/04/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Inflammatory response plays an important role in Parkinson disease (PD). Previous studies have reported an association between human leukocyte antigen (HLA)-DRB1 and the risk of PD. There has also been growing interest in investigating whether inflammation-related genes interact with environmental factors such as smoking to influence PD risk. We performed a pooled analysis of the interaction between HLA-DRB1 and smoking in PD in 3 population-based case-control studies from Denmark and France. METHODS We included 2,056 cases and 2,723 controls from 3 PD studies (Denmark, France) that obtained information on smoking through interviews. Genotyping of the rs660895 polymorphism in the HLA-DRB1 region was based on saliva or blood DNA samples. To assess interactions, we used logistic regression with product terms between rs660895 and smoking. We performed random-effects meta-analysis of marginal associations and interactions. RESULTS Both carrying rs660895-G (AG vs AA: odds ratio [OR] = 0.81; GG vs AA: OR = 0.56; p-trend = 0.003) and ever smoking (OR = 0.56, p < 0.001) were inversely associated with PD. A multiplicative interaction was observed between rs660895 and smoking using codominant, additive (interaction parameter = 1.37, p = 0.005), and dominant (interaction parameter = 1.54, p = 0.001) genetic models without any heterogeneity (I² = 0.0%); the inverse association of rs660895-(AG+GG) with PD seen in never smokers (OR = 0.64, p < 0.001) disappeared among ever smokers (OR = 1.00, p = 0.99). Similar interactions were observed when we investigated light and heavy smokers separately. INTERPRETATION Our study provides the first evidence that smoking modifies the previously reported inverse association of rs660895-G with PD, and suggests that smoking and HLA-DRB1 are involved in common pathways, possibly related to neuroinflammation. Ann Neurol 2017;82:655-664.
Collapse
Affiliation(s)
- Yu-Hsuan Chuang
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA
| | - Pei-Chen Lee
- Department of Health Care Management, College of Health Technology, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Tim Vlaar
- Universite Paris-Saclay, Univ. Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
| | - Claire Mulot
- INSERM UMR-S 1147, CRB EPIGENETEC, Univ. Paris Descartes, Sorbonne Paris Cite, 75006 Paris, France
| | - Marie-Anne Loriot
- Assistance Publique-Hopitaux de Paris, Hopital Europeen Georges Pompidou, Biochimie, Pharmacogenetique et Oncologie Moleculaire, 75015 Paris, France
| | - Johnni Hansen
- Danish Cancer Society Research Center, Danish Cancer Society, Copenhagen, Denmark
| | - Christina M Lill
- Genetic and Molecular Epidemiology Group, Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Beate Ritz
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA.,Department of Neurology, School of Medicine, University of California, Los Angeles, Los Angeles, CA.,Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, CA
| | - Alexis Elbaz
- Universite Paris-Saclay, Univ. Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
| |
Collapse
|
14
|
Simon DK, Wu C, Tilley BC, Lohmann K, Klein C, Payami H, Wills AM, Aminoff MJ, Bainbridge J, Dewey R, Hauser RA, Schaake S, Schneider JS, Sharma S, Singer C, Tanner CM, Truong D, Wei P, Wong PS, Yang T. Caffeine, creatine, GRIN2A and Parkinson's disease progression. J Neurol Sci 2017; 375:355-359. [PMID: 28320167 DOI: 10.1016/j.jns.2017.02.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 10/20/2022]
Abstract
Caffeine is neuroprotective in animal models of Parkinson's disease (PD) and caffeine intake is inversely associated with the risk of PD. This association may be influenced by the genotype of GRIN2A, which encodes an NMDA-glutamate-receptor subunit. In two placebo-controlled studies, we detected no association of caffeine intake with the rate of clinical progression of PD, except among subjects taking creatine, for whom higher caffeine intake was associated with more rapid progression. We now have analyzed data from 420 subjects for whom DNA samples and caffeine intake data were available from a placebo-controlled study of creatine in PD. The GRIN2A genotype was not associated with the rate of clinical progression of PD in the placebo group. However, there was a 4-way interaction between GRIN2A genotype, caffeine, creatine and the time since baseline. Among subjects in the creatine group with high levels of caffeine intake, but not among those with low caffeine intake, the GRIN2A T allele was associated with more rapid progression (p=0.03). These data indicate that the deleterious interaction between caffeine and creatine with respect to rate of progression of PD is influenced by GRIN2A genotype. This example of a genetic factor interacting with environmental factors illustrates the complexity of gene-environment interactions in the progression of PD.
Collapse
Affiliation(s)
- David K Simon
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA.
| | - Cai Wu
- Department of Biostatistics, University of Texas Health Science Center School of Public Health at Houston, Houston, TX 77030, USA.
| | - Barbara C Tilley
- Department of Biostatistics, University of Texas Health Science Center School of Public Health at Houston, Houston, TX 77030, USA.
| | - Katja Lohmann
- Institute of Neurogenetics, University of Luebeck, 23538 Luebeck, Germany.
| | - Christine Klein
- Institute of Neurogenetics, University of Luebeck, 23538 Luebeck, Germany.
| | - Haydeh Payami
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35233, USA; Center for Genomic Medicine, HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA.
| | - Anne-Marie Wills
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Michael J Aminoff
- Department of Neurology, University of California, San Francisco, USA.
| | - Jacquelyn Bainbridge
- University of Colorado, Anschutz Medical Campus, Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Clinical Pharmacy and Neurology, Aurora, CO, USA.
| | - Richard Dewey
- University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Robert A Hauser
- Department of Neurology, University of South Florida, Tampa, FL, USA.
| | - Susen Schaake
- Institute of Neurogenetics, University of Luebeck, 23538 Luebeck, Germany.
| | - Jay S Schneider
- Department of Pathology, Anatomy and Cell Biology, Parkinson's Disease Research Unit, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Saloni Sharma
- Clinical Trials Coordination Center, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Carlos Singer
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA.
| | - Caroline M Tanner
- Parkinson's Disease Research Education and Clinical Center, San Francisco Veteran's Affairs Medical Center, Department of Neurology, University of California, San Francisco, CA, USA.
| | - Daniel Truong
- The Parkinson's and Movement Disorder Institute, Fountain Valley, CA, USA.
| | - Peng Wei
- Department of Biostatistics, University of Texas Health Science Center School of Public Health at Houston, Houston, TX 77030, USA.
| | - Pei Shieen Wong
- University of Colorado, Anschutz Medical Campus, Skaggs School of Pharmacy and Pharmaceutical Sciences, Department of Clinical Pharmacy and Neurology, Aurora, CO, USA; Singapore General Hospital, Singapore 169608, Singapore.
| | - Tianzhong Yang
- Department of Biostatistics, University of Texas Health Science Center School of Public Health at Houston, Houston, TX 77030, USA.
| |
Collapse
|
15
|
Inzelberg R, Flash S, Friedman E, Azizi E. Cutaneous malignant melanoma and Parkinson disease: Common pathways? Ann Neurol 2016; 80:811-820. [PMID: 27761938 DOI: 10.1002/ana.24802] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 09/03/2016] [Accepted: 10/10/2016] [Indexed: 12/25/2022]
Abstract
The mechanisms underlying the high prevalence of cutaneous malignant melanoma (CMM) in Parkinson disease (PD) are unclear, but plausibly involve common pathways. 129Ser-phosphorylated α-synuclein, a pathological PD hallmark, is abundantly expressed in CMM, but not in normal skin. In inherited PD, PARK genes harbor germline mutations; the same genes are somatically mutated in CMM, or their encoded proteins are involved in melanomagenesis. Conversely, genes associated with CMM affect PD risk. PD/CMM-targeted cells share neural crest origin and melanogenesis capability. Pigmentation gene variants may underlie their susceptibility. We review putative genetic intersections that may be suggestive of shared pathways in neurodegeneration/melanomagenesis. Ann Neurol 2016;80:811-820.
Collapse
Affiliation(s)
- Rivka Inzelberg
- Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv
- Center of Advanced Technologies in Rehabilitation, Sheba Medical Center, Tel Hashomer
| | - Shira Flash
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv
| | - Eitan Friedman
- Susanne Levy Gertner Oncogenetics Unit, Institute of Human Genetics, Sheba Medical Center, Tel Hashomer
- Departments of Internal Medicine and Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv
| | - Esther Azizi
- Department of Dermatology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
16
|
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: 24.4] [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
|
17
|
Chuang YH, Lill CM, Lee PC, Hansen J, Lassen CF, Bertram L, Greene N, Sinsheimer JS, Ritz B. Gene-Environment Interaction in Parkinson's Disease: Coffee, ADORA2A, and CYP1A2. Neuroepidemiology 2016; 47:192-200. [PMID: 28135712 DOI: 10.1159/000450855] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/16/2016] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Drinking caffeinated coffee has been reported to provide protection against Parkinson's disease (PD). Caffeine is an adenosine A2A receptor (encoded by the gene ADORA2A) antagonist that increases dopaminergic neurotransmission and Cytochrome P450 1A2 (gene: CYP1A2) metabolizes caffeine; thus, gene polymorphisms in ADORA2A and CYP1A2 may influence the effect coffee consumption has on PD risk. METHODS In a population-based case-control study (PASIDA) in Denmark (1,556 PD patients and 1,606 birth year- and gender-matched controls), we assessed interactions between lifetime coffee consumption and 3 polymorphisms in ADORA2A and CYP1A2 for all subjects, and incident and prevalent PD cases separately using logistic regression models. We also conducted a meta-analysis combining our results with those from previous studies. RESULTS We estimated statistically significant interactions for ADORA2A rs5760423 and heavy vs. light coffee consumption in incident (OR interaction = 0.66 [95% CI 0.46-0.94], p = 0.02) but not prevalent PD. We did not observe interactions for CYP1A2 rs762551 and rs2472304 in incident or prevalent PD. In meta-analyses, PD associations with daily coffee consumption were strongest among carriers of variant alleles in both ADORA2A and CYP1A2. CONCLUSION We corroborated results from a previous report that described interactions between ADORA2A and CYP1A2 polymorphisms and coffee consumption. Our results also suggest that survivor bias may affect results of studies that enroll prevalent PD cases.
Collapse
Affiliation(s)
- Yu-Hsuan Chuang
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles (UCLA), USA
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Genetic Profile, Environmental Exposure, and Their Interaction in Parkinson's Disease. PARKINSONS DISEASE 2016; 2016:6465793. [PMID: 26942037 PMCID: PMC4752982 DOI: 10.1155/2016/6465793] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/05/2016] [Accepted: 01/10/2016] [Indexed: 12/15/2022]
Abstract
The discovery of causative mutations for Parkinson's disease (PD) as well as their functional characterization in cellular and animal models has provided crucial insight into the pathogenesis of this disorder. Today, we know that PD pathogenesis involves multiple related processes including mitochondrial dysfunction, oxidative and nitrative stress, microglial activation and inflammation, and aggregation of α-synuclein and impaired autophagy. However, with the exception of a few families with Mendelian inheritance, the cause of PD in most individuals is yet unknown and the identified genetic susceptibility factors have only small effect size. Epidemiologic studies have found increased risk of PD associated with exposure to environmental toxicants such as pesticides, organic solvents, metals, and air pollutants, while reduced risk of PD associated with smoking cigarettes and coffee consumption. The role of environmental exposure, as well as the contribution of single genetic risk factors, is still controversial. In most of PD cases, disease onset is probably triggered by a complex interplay of many genetic and nongenetic factors, each of which conveys a minor increase in the risk of disease. This review summarizes the current knowledge on causal mutation for PD, susceptibility factors increasing disease risk, and the genetic factors that modify the impact of environmental exposure.
Collapse
|
19
|
Elbaz A, Carcaillon L, Kab S, Moisan F. Epidemiology of Parkinson's disease. Rev Neurol (Paris) 2016; 172:14-26. [DOI: 10.1016/j.neurol.2015.09.012] [Citation(s) in RCA: 221] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 08/25/2015] [Accepted: 09/01/2015] [Indexed: 12/25/2022]
|
20
|
Elbaz A, Moisan F. The scientific bases to consider Parkinson's disease an occupational disease in agriculture professionals exposed to pesticides in France. J Epidemiol Community Health 2015; 70:319-21. [PMID: 26507749 DOI: 10.1136/jech-2015-205455] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 10/08/2015] [Indexed: 11/03/2022]
Affiliation(s)
- Alexis Elbaz
- INSERM, Centre for Research in Epidemiology and Population Health, U1018, Epidemiology of ageing and age related diseases, Villejuif, France University Paris-Sud, UMRS 1018, Villejuif, France Département santé travail, Institut de veille sanitaire (InVS), Saint-Maurice, France
| | - Frédéric Moisan
- Département santé travail, Institut de veille sanitaire (InVS), Saint-Maurice, France
| |
Collapse
|
21
|
Lill CM, Hansen J, Olsen JH, Binder H, Ritz B, Bertram L. Impact of Parkinson's disease risk loci on age at onset. Mov Disord 2015; 30:847-50. [PMID: 25914293 DOI: 10.1002/mds.26237] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/16/2015] [Accepted: 03/20/2015] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The aim of this study was to assess whether recently identified Parkinson's disease (PD) risk genes also influence age at onset in PD. METHODS We genotyped 23 single-nucleotide polymorphisms in 1,526 Danish PD patients and performed linear regression analyses with age at onset. The combined impact of PD risk loci on age at onset was assessed by linear regression analyses using a weighted genetic risk score. RESULTS The strongest effects were observed with rs12726330 in GBA (beta = -3.63, P = 2.0 × 10(-5) ) and rs34311866 in TMEM175/GAK (beta = -1.19, P = 4.0 × 10(-3) ), corresponding to a 3.6-y and 1.2-y decrease of age at onset per risk allele, respectively. The weighted genetic risk score yielded significant association with reduced onset age (P = 3.98 × 10(-3) ), although the variance explained was small (0.6%), and the effect was mostly driven by polymorphisms in GBA and TMEM175/GAK. CONCLUSIONS Overall, our study indicates that GBA and TMEM175/GAK significantly alter age at onset in PD.
Collapse
Affiliation(s)
- Christina M Lill
- Platform for Genome Analytics, Institutes of Neurogenetics & Integrative and Experimental Genomics, University of Lübeck, Lübeck, Germany
| | - Johnni Hansen
- Institute of Cancer Epidemiology, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Epidemiology and Environmental Sciences, School of Public Health, University of California, Los Angeles, California, USA
| | - Jørgen H Olsen
- Institute of Cancer Epidemiology, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Harald Binder
- Institute for Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center Mainz, Mainz, Germany
| | - Beate Ritz
- Department of Epidemiology and Environmental Sciences, School of Public Health, University of California, Los Angeles, California, USA
| | - 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
| |
Collapse
|
22
|
Hamza TH, Hill-Burns EM, Scott WK, Vance JM, Factor SA, Zabetian CP, Payami H. Glutamate receptor gene GRIN2A, coffee, and Parkinson disease. PLoS Genet 2014; 10:e1004774. [PMID: 25411979 PMCID: PMC4238944 DOI: 10.1371/journal.pgen.1004774] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Taye H Hamza
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America; New England Research Institutes Inc., Watertown, Massachusetts, United States of America
| | - Erin M Hill-Burns
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America
| | - William K Scott
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Jeffrey M Vance
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Stewart A Factor
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Cyrus P Zabetian
- VA Puget Sound Health Care System and Department of Neurology, University of Washington, Seattle, Washington, United States of America
| | - Haydeh Payami
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, New York, United States of America; Department of Biomedical Science, School of Public Health, State University of New York, Albany, New York, United States of America
| |
Collapse
|