1
|
Yi M, Li J, Jian S, Li B, Huang Z, Shu L, Zhang Y. Quantitative and causal analysis for inflammatory genes and the risk of Parkinson's disease. Front Immunol 2023; 14:1119315. [PMID: 36926335 PMCID: PMC10011457 DOI: 10.3389/fimmu.2023.1119315] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/08/2023] [Indexed: 03/08/2023] Open
Abstract
Background The dysfunction of immune system and inflammation contribute to the Parkinson's disease (PD) pathogenesis. Cytokines, oxidative stress, neurotoxin and metabolism associated enzymes participate in neuroinflammation in PD and the genes involved in them have been reported to be associated with the risk of PD. In our study, we performed a quantitative and causal analysis of the relationship between inflammatory genes and PD risk. Methods Standard process was performed for quantitative analysis. Allele model (AM) was used as primary outcome analysis and dominant model (DM) and recessive model (RM) were applied to do the secondary analysis. Then, for those genes significantly associated with the risk of PD, we used the published GWAS summary statistics for Mendelian Randomization (MR) to test the causal analysis between them. Results We included 36 variants in 18 genes for final pooled analysis. As a result, IL-6 rs1800795, TNF-α rs1799964, PON1 rs854560, CYP2D6 rs3892097, HLA-DRB rs660895, BST1 rs11931532, CCDC62 rs12817488 polymorphisms were associated with the risk of PD statistically with the ORs ranged from 0.66 to 3.19 while variants in IL-1α, IL-1β, IL-10, MnSOD, NFE2L2, CYP2E1, NOS1, NAT2, ABCB1, HFE and MTHFR were not related to the risk of PD. Besides, we observed that increasing ADP-ribosyl cyclase (coded by BST1) had causal effect on higher PD risk (OR[95%CI] =1.16[1.10-1.22]) while PON1(coded by PON1) shown probably protective effect on PD risk (OR[95%CI] =0.81[0.66-0.99]). Conclusion Several polymorphisms from inflammatory genes of IL-6, TNF-α, PON1, CYP2D6, HLA-DRB, BST1, CCDC62 were statistically associated with the susceptibility of PD, and with evidence of causal relationships for ADP-ribosyl cyclase and PON1 on PD risk, which may help understand the mechanisms and pathways underlying PD pathogenesis.
Collapse
Affiliation(s)
- Minhan Yi
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,School of Life Sciences, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiaxin Li
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shijie Jian
- School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Binbin Li
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zini Huang
- Bangor College, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Li Shu
- National Health Commission Key Laboratory for Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| | - Yuan Zhang
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
2
|
Loughnan R, Ahern J, Tompkins C, Palmer CE, Iversen J, Thompson WK, Andreassen O, Jernigan T, Sugrue L, Dale A, Boyle MET, Fan CC. Association of Genetic Variant Linked to Hemochromatosis With Brain Magnetic Resonance Imaging Measures of Iron and Movement Disorders. JAMA Neurol 2022; 79:919-928. [PMID: 35913729 PMCID: PMC9344392 DOI: 10.1001/jamaneurol.2022.2030] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/26/2022] [Indexed: 12/31/2022]
Abstract
Importance Hereditary hemochromatosis (HH) is an autosomal recessive genetic disorder that leads to iron overload. Conflicting results from previous research has led some to believe the brain is spared the toxic effects of iron in HH. Objective To test the association of the strongest genetic risk variant for HH on brainwide measures sensitive to iron deposition and the rates of movement disorders in a substantially larger sample than previous studies of its kind. Design, Setting, and Participants This cross-sectional retrospective study included participants from the UK Biobank, a population-based sample. Genotype, health record, and neuroimaging data were collected from January 2006 to May 2021. Data analysis was conducted from January 2021 to April 2022. Disorders tested included movement disorders (International Statistical Classification of Diseases and Related Health Problems, Tenth Revision [ICD-10], codes G20-G26), abnormalities of gait and mobility (ICD-10 codes R26), and other disorders of the nervous system (ICD-10 codes G90-G99). Exposures Homozygosity for p.C282Y, the largest known genetic risk factor for HH. Main Outcomes and Measures T2-weighted and T2* signal intensity from brain magnetic resonance imaging scans, measures sensitive to iron deposition, and clinical diagnosis of neurological disorders. Results The total cohort consisted of 488 288 individuals (264 719 female; ages 49-87 years, largely northern European ancestry), 2889 of whom were p.C282Y homozygotes. The neuroimaging analysis consisted of 836 individuals: 165 p.C282Y homozygotes (99 female) and 671 matched controls (399 female). A total of 206 individuals were excluded from analysis due to withdrawal of consent. Neuroimaging analysis showed that p.C282Y homozygosity was associated with decreased T2-weighted and T2* signal intensity in subcortical motor structures (basal ganglia, thalamus, red nucleus, and cerebellum; Cohen d >1) consistent with substantial iron deposition. Across the whole UK Biobank (2889 p.C282Y homozygotes, 485 399 controls), we found a significantly increased prevalence for movement disorders in male homozygotes (OR, 1.80; 95% CI, 1.28-2.55; P = .001) but not female individuals (OR, 1.09; 95% CI, 0.70-1.73; P = .69). Among the 31 p.C282Y male homozygotes with a movement disorder, only 10 had a concurrent HH diagnosis. Conclusions and Relevance These findings indicate increased iron deposition in subcortical motor circuits in p.C282Y homozygotes and confirm an increased association with movement disorders in male homozygotes. Early treatment in HH effectively prevents the negative consequences of iron overload in the liver and heart. Our work suggests that screening for p.C282Y homozygosity in high-risk individuals also has the potential to reduce brain iron accumulation and to reduce the risk of movement disorders among male individuals who are homozygous for this mutation.
Collapse
Affiliation(s)
- Robert Loughnan
- Department of Cognitive Science, University of California, San Diego, La Jolla
- Population Neuroscience and Genetics, University of California, San Diego, La Jolla
| | - Jonathan Ahern
- Department of Cognitive Science, University of California, San Diego, La Jolla
| | - Cherisse Tompkins
- Department of Cognitive Science, University of California, San Diego, La Jolla
| | - Clare E. Palmer
- Center for Human Development, University of California, San Diego, La Jolla
| | - John Iversen
- Center for Human Development, University of California, San Diego, La Jolla
| | - Wesley K. Thompson
- Population Neuroscience and Genetics, University of California, San Diego, La Jolla
- Division of Biostatistics, Department of Radiology, University of California, San Diego, La Jolla
- Center for Population Neuroscience and Genetics, Laureate Institute for Brain Research, Tulsa, Oklahoma
| | - Ole Andreassen
- NORMENT Centre, Division of Mental Health and Addiction, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Terry Jernigan
- Department of Cognitive Science, University of California, San Diego, La Jolla
- Center for Human Development, University of California, San Diego, La Jolla
- Department of Radiology, University of California, San Diego School of Medicine, La Jolla
- Department of Psychiatry, University of California, San Diego School of Medicine, La Jolla
| | - Leo Sugrue
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
- Department of Psychiatry, University of California, San Francisco
| | - Anders Dale
- Department of Cognitive Science, University of California, San Diego, La Jolla
- Department of Radiology, University of California, San Diego School of Medicine, La Jolla
- Department of Neuroscience, University of California, San Diego School of Medicine, La Jolla
- Center for Multimodal Imaging and Genetics, University of California, San Diego School of Medicine, La Jolla
| | - Mary E. T. Boyle
- Department of Cognitive Science, University of California, San Diego, La Jolla
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Chun Chieh Fan
- Population Neuroscience and Genetics, University of California, San Diego, La Jolla
- Center for Multimodal Imaging and Genetics, University of California, San Diego School of Medicine, La Jolla
- Center for Population Neuroscience and Genetics, Laureate Institute for Brain Research, Tulsa, Oklahoma
| |
Collapse
|
3
|
Kim Y, Stahl MC, Huang X, Connor JR. H63D variant of the homeostatic iron regulator (HFE) gene alters α-synuclein expression, aggregation, and toxicity. J Neurochem 2020; 155:177-190. [PMID: 32574378 DOI: 10.1111/jnc.15107] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022]
Abstract
Pathological features of Parkinson's disease include the formation of Lewy bodies containing α-synuclein and the accumulation of iron in the substantia nigra. Previous studies have suggested that iron accumulation contributes to the Parkinson's disease pathology through reactive oxygen species production and accelerated α-synuclein aggregation. This study examines the effects of commonly occurring H63D variant of the homeostatic iron regulatory (HFE) gene on α-synuclein pathology in cell culture and animal models. H63D HFE expression in SH-SY5Y cells lowered endogenous α-synuclein levels and significantly decreased pre-formed fibril-induced α-synuclein aggregation. H63D HFE cells were also protected from pre-formed fibril-induced apoptosis. Autophagic flux, a major pathway for α-synuclein clearance, was increased in H63D HFE cells. Expression of REDD1 was elevated and rapamycin treatment was unable to further induce autophagy, indicating mTORC1 inhibition as the main mechanism of autophagy induction. Moreover, siRNA knockdown of REDD1 in H63D HFE cells decreased autophagic flux and increased the sensitivity to PFF-mediated toxicity. While iron chelator (deferiprone) treatment rescued WT HFE cells from pre-formed fibril toxicity, it exacerbated or was unable to rescue H63D HFE cells. In the in vivo pre-formed fibril intracranial injection model, H67D Hfe (mouse homolog of the human H63D HFE variant) C57BL/6J × 129 mice showed less α-synuclein aggregation and less decline in motor function compared to WT Hfe. Collectively, this study suggests that H63D HFE variant modifies α-synuclein pathology through the induction of autophagy and has the potential to impact the pathogenesis and treatment response in Parkinson's disease.
Collapse
Affiliation(s)
- Yunsung Kim
- Department of Neurology, Penn State College of Medicine, Hershey, PA, USA
| | - Mark C Stahl
- Department of Neurology, Penn State College of Medicine, Hershey, PA, USA
- Neurocrine Biosciences, San Diego, CA, USA
| | - Xuemei Huang
- Department of Neurology, Penn State College of Medicine, Hershey, PA, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Translational Brain Research Center, Penn State College of Medicine, Hershey, PA, USA
| | - James R Connor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Translational Brain Research Center, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
4
|
Kim Y, Connor JR. The roles of iron and HFE genotype in neurological diseases. Mol Aspects Med 2020; 75:100867. [PMID: 32654761 DOI: 10.1016/j.mam.2020.100867] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/21/2020] [Accepted: 05/24/2020] [Indexed: 12/13/2022]
Abstract
Iron accumulation is a recurring pathological phenomenon in many neurological diseases including Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis, and others. Iron is essential for normal development and functions of the brain; however, excess redox-active iron can also lead to oxidative damage and cell death. Especially for terminally differentiated cells like neurons, regulation of reactive oxygen species is critical for cell viability. As a result, cellular iron level is tightly regulated. Although iron accumulation related to neurological diseases has been well documented, the pathoetiological contributions of the homeostatic iron regulator (HFE), which controls cellular iron uptake, is less understood. Furthermore, a common HFE variant, H63D HFE, has been identified as a modifier of multiple neurological diseases. This review will discuss the roles of iron and HFE in the brain as well as their impact on various disease processes.
Collapse
Affiliation(s)
- Yunsung Kim
- Penn State College of Medicine, Department of Neurosurgery, Hershey, PA, USA
| | - James R Connor
- Penn State College of Medicine, Department of Neurosurgery, Hershey, PA, USA.
| |
Collapse
|
5
|
Song IY, Snyder AM, Kim Y, Neely EB, Wade QW, Connor JR. The Nrf2-mediated defense mechanism associated with HFE genotype limits vulnerability to oxidative stress-induced toxicity. Toxicology 2020; 441:152525. [PMID: 32540480 DOI: 10.1016/j.tox.2020.152525] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/25/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023]
Abstract
There is considerable interest in gene and environment interactions in neurodegenerative diseases. The HFE (homeostatic iron regulator) gene variant (H63D) is highly prevalent in the population and has been investigated as a disease modifier in multiple neurodegenerative diseases. We have developed a mouse model to interrogate the impact of this gene variant in a model of paraquat toxicity. Using primary astrocytes, we found that the H67D-Hfe(equivalent of the human H63D variant) astrocytes are less vulnerable than the WT-Hfe astrocytes to paraquat-induced cell death, mitochondrial damage, and cellular senescence. We hypothesized that the Hfe variant-associated protection is a result of the activation of the Nrf2 antioxidant defense system and found a significant increase in Nrf2 levels after paraquat exposure in the H67D-Hfe astrocytes than the WT-Hfe astrocytes. Moreover, decreasing Nrf2 by molecular or pharmaceutical manipulation resulted in increased vulnerability to paraquat in the H67D-Hfe astrocytes. To further elucidate the role of Hfe variant genotype in neuroprotection mediated by astrocytes, we added media from the paraquat-treated astrocytes to differentiated SH-SY5Y neuroblastoma cells and found a significantly larger reduction in the viability when treated with WT-Hfe astrocyte media than the H67D-Hfe astrocyte media possibly due to higher secretion of IL-6 observed in the WT-Hfe astrocytes. To further explore the mechanism of Nrf2 protection, we measured NQO1, the Nrf2-mediated antioxidant, in primary astrocytes and found a significantly higher NQO1 level in the H67D-Hfe astrocytes. To consider the translational potential of our findings, we utilized the PPMI (Parkinson's Progression Markers Initiative) clinical database and found that, consistent with the mouse study, H63D-HFE carriers had a significantly higher NQO1 level in the CSF than the WT-HFE carriers. Consistent with our previous reports on H63D-HFE in disease, these data further suggest that HFE genotype in the human population impacts the antioxidant defense system and can therefore alter pathogenesis.
Collapse
Affiliation(s)
- Insung Y Song
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania 17033, United States.
| | - Amanda M Snyder
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania 17033, United States
| | - Yunsung Kim
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania 17033, United States
| | - Elizabeth B Neely
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania 17033, United States
| | - Quinn W Wade
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania 17033, United States
| | - James R Connor
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania 17033, United States
| |
Collapse
|
6
|
Selective vulnerability in α-synucleinopathies. Acta Neuropathol 2019; 138:681-704. [PMID: 31006067 PMCID: PMC6800835 DOI: 10.1007/s00401-019-02010-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/13/2019] [Accepted: 04/05/2019] [Indexed: 12/11/2022]
Abstract
Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy are neurodegenerative disorders resulting in progressive motor/cognitive deficits among other symptoms. They are characterised by stereotypical brain cell loss accompanied by the formation of proteinaceous aggregations of the protein α-synuclein (α-syn), being, therefore, termed α-synucleinopathies. Although the presence of α-syn inclusions is a common hallmark of these disorders, the exact nature of the deposited protein is specific to each disease. Different neuroanatomical regions and cellular populations manifest a differential vulnerability to the appearance of protein deposits, cell dysfunction, and cell death, leading to phenotypic diversity. The present review describes the multiple factors that contribute to the selective vulnerability in α-synucleinopathies. We explore the intrinsic cellular properties in the affected regions, including the physiological and pathophysiological roles of endogenous α-syn, the metabolic and genetic build-up of the cells and their connectivity. These factors converge with the variability of the α-syn conformational strains and their spreading capacity to dictate the phenotypic diversity and regional vulnerability of each disease. Finally, we describe the exogenous and environmental factors that potentially contribute by igniting and modulating the differential pathology in α-synucleinopathies. In conclusion, we think that it is the confluence of this disruption of the cellular metabolic state and α-syn structural equilibrium through the anatomical connectivity which appears to initiate cascades of pathological processes triggered by genetic, environmental, or stochastic events that result in the "death by a thousand cuts" profile of α-synucleinopathies.
Collapse
|
7
|
Nixon AM, Meadowcroft MD, Neely EB, Snyder AM, Purnell CJ, Wright J, Lamendella R, Nandar W, Huang X, Connor JR. HFE Genotype Restricts the Response to Paraquat in a Mouse Model of Neurotoxicity. J Neurochem 2018; 145:299-311. [PMID: 29315562 DOI: 10.1111/jnc.14299] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/05/2017] [Accepted: 01/03/2018] [Indexed: 12/30/2022]
Abstract
Parkinson's disease is marked clinically by motor dysfunction and pathologically by dopaminergic cell loss in the substantia nigra and iron accumulation in the substantia nigra. The driver underlying iron accumulation remains unknown and could be genetic or environmental. The HFE protein is critical for the regulation of cellular iron uptake. Mutations within this protein are associated with increased iron accumulation including in the brain. We have focused on the commonly occurring H63D variant of the HFE gene as a disease modifier in a number of neurodegenerative diseases. To investigate the role of H63D HFE genotype, we generated a mouse model in which the wild-type (WT) HFE gene is replaced by the H67D gene variant (mouse homolog of the human H63D gene variant). Using paraquat toxicity as the model for Parkinson's disease, we found that WT mice responded as expected with significantly greater motor function, loss of tyrosine hydroxylase staining and increase microglial staining in the substantia nigra, and an increase in R2 relaxation rate within the substantia nigra of the paraquat-treated mice compared to their saline-treated counterparts. In contrast, the H67D mice showed a remarkable resistance to paraquat treatment; specifically differing from the WT mice with no changes in motor function or changes in R2 relaxation rates following paraquat exposure. At baseline, there were differences between the H67D HFE mice and WT mice in gut microbiome profile and increased L-ferritin staining in the substantia nigra that could account for the resistance to paraquat. Of particular note, the H67D HFE mice regardless of whether or not they were treated with paraquat had significantly less tyrosine hydroxylase immunostaining than WT. Our results clearly demonstrate that the HFE genotype impacts the expression of tyrosine hydroxylase in the substantia nigra, the gut microbiome and the response to paraquat providing additional support that the HFE genotype is a disease modifier for Parkinson's disease. Moreover, the finding that the HFE mutant mice are resistant to paraquat may provide a model in which to study resistant mechanisms to neurotoxicants.
Collapse
Affiliation(s)
- Anne M Nixon
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Mark D Meadowcroft
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
- Department of Radiology, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Elizabeth B Neely
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Amanda M Snyder
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - Carson J Purnell
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | | | - Regina Lamendella
- Wright Labs, Huntingdon, Pennsylvania, USA
- Department of Microbiology, Juniata College, Huntingdon, Pennsylvania, USA
| | - Wint Nandar
- Department of Sports Medicine and Nutrition, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xuemei Huang
- Department of Neurology, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| | - James R Connor
- Department of Neurosurgery, M.S. Hershey Penn State University College of Medicine, M.S. Hershey Medical Center, Hershey, Pennsylvania, USA
| |
Collapse
|
8
|
ASSESSMENT OF DRUSEN AND OTHER RETINAL DEGENERATIVE CHANGES IN PATIENTS WITH HEREDITARY HEMOCHROMATOSIS. Retina 2018; 38:594-599. [DOI: 10.1097/iae.0000000000001577] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
9
|
Arshad AR, Sulaiman SA, Saperi AA, Jamal R, Mohamed Ibrahim N, Abdul Murad NA. MicroRNAs and Target Genes As Biomarkers for the Diagnosis of Early Onset of Parkinson Disease. Front Mol Neurosci 2017; 10:352. [PMID: 29163029 PMCID: PMC5671573 DOI: 10.3389/fnmol.2017.00352] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/13/2017] [Indexed: 12/21/2022] Open
Abstract
Among the neurodegenerative disorders, Parkinson's disease (PD) ranks as the second most common disorder with a higher prevalence in individuals aged over 60 years old. Younger individuals may also be affected with PD which is known as early onset PD (EOPD). Despite similarities between the characteristics of EOPD and late onset PD (LODP), EOPD patients experience much longer disease manifestations and poorer quality of life. Although some individuals are more prone to have EOPD due to certain genetic alterations, the molecular mechanisms that differentiate between EOPD and LOPD remains unclear. Recent findings in PD patients revealed that there were differences in the genetic profiles of PD patients compared to healthy controls, as well as between EOPD and LOPD patients. There were variants identified that correlated with the decline of cognitive and motor symptoms as well as non-motor symptoms in PD. There were also specific microRNAs that correlated with PD progression, and since microRNAs have been shown to be involved in the maintenance of neuronal development, mitochondrial dysfunction and oxidative stress, there is a strong possibility that these microRNAs can be potentially used to differentiate between subsets of PD patients. PD is mainly diagnosed at the late stage, when almost majority of the dopaminergic neurons are lost. Therefore, identification of molecular biomarkers for early detection of PD is important. Given that miRNAs are crucial in controlling the gene expression, these regulatory microRNAs and their target genes could be used as biomarkers for early diagnosis of PD. In this article, we discussed the genes involved and their regulatory miRNAs, regarding their roles in PD progression, based on the findings of significantly altered microRNAs in EOPD studies. We also discussed the potential of these miRNAs as molecular biomarkers for early diagnosis.
Collapse
Affiliation(s)
- Ahmad R. Arshad
- UKM Medical Centre, UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Malaysia
| | - Siti A. Sulaiman
- UKM Medical Centre, UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Malaysia
| | - Amalia A. Saperi
- UKM Medical Centre, UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Malaysia
| | - Rahman Jamal
- UKM Medical Centre, UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Malaysia
| | - Norlinah Mohamed Ibrahim
- Department of Medicine, Faculty of Medicine, UKM Medical Centre, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Malaysia
| | - Nor Azian Abdul Murad
- UKM Medical Centre, UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Bandar Tun Razak, Malaysia
| |
Collapse
|
10
|
Levodopa Responsive Parkinsonism in Patients with Hemochromatosis: Case Presentation and Literature Review. Case Rep Neurol Med 2017; 2017:5146723. [PMID: 28424751 PMCID: PMC5382304 DOI: 10.1155/2017/5146723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 03/19/2017] [Indexed: 11/29/2022] Open
Abstract
Hemochromatosis is an autosomal recessive disorder which leads to abnormal iron deposition in the parenchyma of multiple organs causing tissue damage. Accumulation of iron in the brain has been postulated to be associated with several neurodegenerative diseases including Parkinson's disease. The excess iron promotes Parkin and α-synuclein aggregation in the neurons. Excess iron has also been noted in substantia nigra on MRI especially using susceptibility weighted imaging in patients with Parkinson's disease. We present a case of a young male with alleles for both C282Y and H63D who presented with signs of Parkinsonism and demonstrated significant improvement with levodopa treatment.
Collapse
|
11
|
Hare DJ, Cardoso BR, Raven EP, Double KL, Finkelstein DI, Szymlek-Gay EA, Biggs BA. Excessive early-life dietary exposure: a potential source of elevated brain iron and a risk factor for Parkinson's disease. NPJ Parkinsons Dis 2017; 3:1. [PMID: 28649601 PMCID: PMC5460187 DOI: 10.1038/s41531-016-0004-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 11/08/2016] [Accepted: 11/11/2016] [Indexed: 11/28/2022] Open
Abstract
Iron accumulates gradually in the ageing brain. In Parkinson's disease, iron deposition within the substantia nigra is further increased, contributing to a heightened pro-oxidant environment in dopaminergic neurons. We hypothesise that individuals in high-income countries, where cereals and infant formulae have historically been fortified with iron, experience increased early-life iron exposure that predisposes them to age-related iron accumulation in the brain. Combined with genetic factors that limit iron regulatory capacity and/or dopamine metabolism, this may increase the risk of Parkinson's diseases. We propose to (a) validate a retrospective biomarker of iron exposure in children; (b) translate this biomarker to adults; (c) integrate it with in vivo brain iron in Parkinson's disease; and (d) longitudinally examine the relationships between early-life iron exposure and metabolism, brain iron deposition and Parkinson's disease risk. This approach will provide empirical evidence to support therapeutically addressing brain iron deposition in Parkinson's diseases and produce a potential biomarker of Parkinson's disease risk in preclinical individuals.
Collapse
Affiliation(s)
- Dominic J Hare
- Department of Medicine (Royal Melbourne Hospital) at the Doherty Institute, The University of Melbourne, Parkville, Melbourne, VIC Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Melbourne, VIC Australia
- Elemental Bio-imaging Facility, University of Technology Sydney, Broadway, NSW Australia
| | - Bárbara Rita Cardoso
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Melbourne, VIC Australia
- Department of Pharmaceutical Sciences, Department of Food and Experimental Nutrition, University of São Paulo, São Paulo, Brazil
| | - Erika P Raven
- Center for Functional and Molecular Imaging, Georgetown University Medical Centre, Washington DC, USA
- Advanced Magnetic Resonance Imaging Section, Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD USA
| | - Kay L Double
- Sydney Medical School, University of Sydney, Darlington, NSW Australia
- Brain and Mind Centre, University of Sydney, Camperdown, NSW Australia
| | - David I Finkelstein
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Melbourne, VIC Australia
| | - Ewa A Szymlek-Gay
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC Australia
| | - Beverley-Ann Biggs
- Department of Medicine (Royal Melbourne Hospital) at the Doherty Institute, The University of Melbourne, Parkville, Melbourne, VIC Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Parkville, Melbourne, VIC Australia
| |
Collapse
|
12
|
Parkinson's Disease: The Mitochondria-Iron Link. PARKINSONS DISEASE 2016; 2016:7049108. [PMID: 27293957 PMCID: PMC4886095 DOI: 10.1155/2016/7049108] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 12/14/2022]
Abstract
Mitochondrial dysfunction, iron accumulation, and oxidative damage are conditions often found in damaged brain areas of Parkinson's disease. We propose that a causal link exists between these three events. Mitochondrial dysfunction results not only in increased reactive oxygen species production but also in decreased iron-sulfur cluster synthesis and unorthodox activation of Iron Regulatory Protein 1 (IRP1), a key regulator of cell iron homeostasis. In turn, IRP1 activation results in iron accumulation and hydroxyl radical-mediated damage. These three occurrences-mitochondrial dysfunction, iron accumulation, and oxidative damage-generate a positive feedback loop of increased iron accumulation and oxidative stress. Here, we review the evidence that points to a link between mitochondrial dysfunction and iron accumulation as early events in the development of sporadic and genetic cases of Parkinson's disease. Finally, an attempt is done to contextualize the possible relationship between mitochondria dysfunction and iron dyshomeostasis. Based on published evidence, we propose that iron chelation-by decreasing iron-associated oxidative damage and by inducing cell survival and cell-rescue pathways-is a viable therapy for retarding this cycle.
Collapse
|
13
|
Duan C, Wang M, Zhang Y, Wei X, Huang Y, Zhang H, Cheng L, Gai Z. C282Y and H63D Polymorphisms in Hemochromatosis Gene and Risk of Parkinson's Disease: A Meta-Analysis. Am J Alzheimers Dis Other Demen 2016; 31:201-7. [PMID: 26340960 PMCID: PMC10852941 DOI: 10.1177/1533317515602220] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE A meta-analysis was performed to better clarify the association between hemochromatosis (HFE) gene and the risk of Parkinson's disease (PD). METHODS Pooled odds ratio (OR) with 95% confidence interval (CI) was calculated from fixed- and random-effect models. Heterogeneity among studies was evaluated using the I(2) and Q test. Egger's test was used to estimate the publication bias. RESULTS We identified 8 articles with 9 independent studies for this meta-analysis. The present meta-analysis showed no significant association of Y allele with the risk of PD in dominant (OR = 0.87, 95% CI = 0.70-1.09), recessive (OR = 1.58, 95% CI = 0.61-4.10), and codominant (OR = 0.88, 95% CI = 0.72-1.09) models for C282Y. There were also no significant associations of D allele with the risk of PD in dominant (OR = 1.04, 95% CI = 0.87-1.24), recessive (OR = 1.23, 95% CI = 0.70-2.18), and codominant (OR = 1.04, 95% CI = 0.89-1.22) genetic models for H63D. No publication bias was detected. CONCLUSION The meta-analysis indicated that C282Y and H63D polymorphisms in the HFE gene might not be associated with PD.
Collapse
Affiliation(s)
| | - Meiyun Wang
- Ji'nan Children's Hospital, Ji'nan, Shandong, China
| | - Yan Zhang
- Ji'nan Center For Disease Control And Prevention, Ji'nan, Shandong, China
| | - Xuxia Wei
- Ji'nan Children's Hospital, Ji'nan, Shandong, China
| | - Yan Huang
- Ji'nan Children's Hospital, Ji'nan, Shandong, China
| | | | - Lu Cheng
- Ji'nan Children's Hospital, Ji'nan, Shandong, China
| | - Zhongtao Gai
- Ji'nan Children's Hospital, Ji'nan, Shandong, China
| |
Collapse
|
14
|
Xia J, Xu H, Jiang H, Xie J. The association between the C282Y and H63D polymorphisms of HFE gene and the risk of Parkinson's disease: A meta-analysis. Neurosci Lett 2015; 595:99-103. [PMID: 25863172 DOI: 10.1016/j.neulet.2015.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/27/2015] [Accepted: 04/04/2015] [Indexed: 12/18/2022]
Abstract
Impaired brain iron homeostasis has been considered as an important mechanism in Parkinson's diseases (PD). There are indications that C282Y and H63D polymorphisms of HFE genes involved in iron metabolism might contribute to the pathogenesis of PD in some cases. However, the investigation of the relationship between PD and the two polymorphisms had produced contradictory results. We performed a meta-analysis to assess the C282Y and H63D polymorphisms of HFE in PD susceptibility. PubMed, EMBASE and Web of Science were systematically searched to identify relevant researches. The strict selection criteria and exclusion standard were applied. Odds ratios (ORs) with 95% confidence intervals (CIs) were used to assess the strength of associations. A fixed-effect or random-effect model was selected, depending on the results of the heterogeneity test. Fifteen studies were included in the meta-analysis (eight studies with 1631 cases and 4548 controls for C282Y; seven studies with 1192 cases and 4065 controls for H63D). For the C282Y polymorphism, significant associations were observed in the Recessive model (YY vs CY+CC: OR=0.22, 95% CI=0.09-0.57, P=0.002). This indicated that the C282Y polymorphism in HFE might be a potential protective factor for PD. However, no significant associations were found for any genetic model for the H63D polymorphism, suggesting that the H63D polymorphism might not be associated with PD.
Collapse
Affiliation(s)
- Jianjian Xia
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao 266071, China
| | - Huamin Xu
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao 266071, China
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao 266071, China
| | - Junxia Xie
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Medical College of Qingdao University, Qingdao 266071, China.
| |
Collapse
|
15
|
Graham J, Hobson D, Ponnampalam A. High affinity hemoglobin and Parkinson’s disease. Med Hypotheses 2014; 83:819-21. [DOI: 10.1016/j.mehy.2014.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 09/03/2014] [Accepted: 10/20/2014] [Indexed: 10/24/2022]
|
16
|
Pretorius E, Swanepoel AC, Buys AV, Vermeulen N, Duim W, Kell DB. Eryptosis as a marker of Parkinson's disease. Aging (Albany NY) 2014; 6:788-819. [PMID: 25411230 PMCID: PMC4247384 DOI: 10.18632/aging.100695] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/24/2014] [Indexed: 12/20/2022]
Abstract
A major trend in recent Parkinson's disease (PD) research is the investigation of biological markers that could help in identifying at-risk individuals or to track disease progression and response to therapies. Central to this is the knowledge that inflammation is a known hallmark of PD and of many other degenerative diseases. In the current work, we focus on inflammatory signalling in PD, using a systems approach that allows us to look at the disease in a more holistic way. We discuss cyclooxygenases, prostaglandins, thromboxanes and also iron in PD. These particular signalling molecules are involved in PD pathophysiology, but are also very important in an aberrant coagulation/hematology system. We present and discuss a hypothesis regarding the possible interaction of these aberrant signalling molecules implicated in PD, and suggest that these molecules may affect the erythrocytes of PD patients. This would be observable as changes in the morphology of the RBCs and of PD patients relative to healthy controls. We then show that the RBCs of PD patients are indeed rather dramatically deranged in their morphology, exhibiting eryptosis (a kind of programmed cell death). This morphological indicator may have useful diagnostic and prognostic significance.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa
| | - Albe C Swanepoel
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa
| | - Antoinette V Buys
- Microscopy and Microanalysis Unit, University of Pretoria, Arcadia 0007, South Africa
| | - Natasha Vermeulen
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa
| | - Wiebren Duim
- Department of Neurology Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa
| | - Douglas B Kell
- School of Chemistry and The Manchester Institute of Biotechnology, The University of Manchester, Manchester M1 7DN, Lancs, UK
| |
Collapse
|
17
|
Delatycki MB, Tai G, Corben L, Yiu EM, Evans-Galea MV, Stephenson SEM, Gurrin L, Allen KJ, Lynch D, Lockhart PJ. HFE p.C282Y heterozygosity is associated with earlier disease onset in Friedreich ataxia. Mov Disord 2014; 29:940-3. [PMID: 24390816 DOI: 10.1002/mds.25795] [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: 07/27/2013] [Revised: 11/26/2013] [Accepted: 12/03/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Friedreich ataxia (FRDA) generally results from reduced frataxin, a mitochondrial protein involved in iron metabolism. We assessed whether HFE p.C282Y and/or p.H63D heterozygosity modifies age at disease onset or disease severity in individuals with FRDA. METHODS One hundred seventy individuals with FRDA were assessed for the association of HFE p.C282Y and p.H63D with (1) age at disease onset and (2) Friedreich Ataxia Rating Scale (FARS) score. RESULTS After adjusting for the smaller FXN GAA repeat size and sex, individuals with FRDA and heterozygous for p.C282Y had disease onset on average 3.72 years earlier than those homozygous for the wild-type amino acid (P = 0.02). Neither mutation affected disease severity as measured by FARS. CONCLUSIONS It is hypothesized that the association between p.C282Y heterozygosity and an earlier age at FRDA onset relates to exacerbation of the already dysregulated iron metabolism that plays a major role in the pathogenesis of FRDA.
Collapse
Affiliation(s)
- Martin B Delatycki
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Childrens Research Institute, Parkville, Victoria, Australia; Clinical Genetics, Austin Health, Heidelberg, Victoria, Australia; Department of Paediatrics, University of Melbourne, Royal Children's Hospital, Parkville, Victoria, Australia; School of Psychology and Psychiatry, Monash University, Clayton, Victoria, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Nigral iron elevation is an invariable feature of Parkinson's disease and is a sufficient cause of neurodegeneration. BIOMED RESEARCH INTERNATIONAL 2014; 2014:581256. [PMID: 24527451 PMCID: PMC3914334 DOI: 10.1155/2014/581256] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 10/28/2013] [Indexed: 12/22/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by motor deficits accompanying degeneration of substantia nigra pars compactor (SNc) neurons. Although familial forms of the disease exist, the cause of sporadic PD is unknown. Symptomatic treatments are available for PD, but there are no disease modifying therapies. While the neurodegenerative processes in PD may be multifactorial, this paper will review the evidence that prooxidant iron elevation in the SNc is an invariable feature of sporadic and familial PD forms, participates in the disease mechanism, and presents as a tractable target for a disease modifying therapy.
Collapse
|
19
|
Pichler I, Del Greco M. F, Gögele M, Lill CM, Bertram L, Do CB, Eriksson N, Foroud T, Myers RH, Nalls M, Keller MF, Benyamin B, Whitfield JB, Pramstaller PP, Hicks AA, Thompson JR, Minelli C. Serum iron levels and the risk of Parkinson disease: a Mendelian randomization study. PLoS Med 2013; 10:e1001462. [PMID: 23750121 PMCID: PMC3672214 DOI: 10.1371/journal.pmed.1001462] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 04/24/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Although levels of iron are known to be increased in the brains of patients with Parkinson disease (PD), epidemiological evidence on a possible effect of iron blood levels on PD risk is inconclusive, with effects reported in opposite directions. Epidemiological studies suffer from problems of confounding and reverse causation, and mendelian randomization (MR) represents an alternative approach to provide unconfounded estimates of the effects of biomarkers on disease. We performed a MR study where genes known to modify iron levels were used as instruments to estimate the effect of iron on PD risk, based on estimates of the genetic effects on both iron and PD obtained from the largest sample meta-analyzed to date. METHODS AND FINDINGS We used as instrumental variables three genetic variants influencing iron levels, HFE rs1800562, HFE rs1799945, and TMPRSS6 rs855791. Estimates of their effect on serum iron were based on a recent genome-wide meta-analysis of 21,567 individuals, while estimates of their effect on PD risk were obtained through meta-analysis of genome-wide and candidate gene studies with 20,809 PD cases and 88,892 controls. Separate MR estimates of the effect of iron on PD were obtained for each variant and pooled by meta-analysis. We investigated heterogeneity across the three estimates as an indication of possible pleiotropy and found no evidence of it. The combined MR estimate showed a statistically significant protective effect of iron, with a relative risk reduction for PD of 3% (95% CI 1%-6%; p = 0.001) per 10 µg/dl increase in serum iron. CONCLUSIONS Our study suggests that increased iron levels are causally associated with a decreased risk of developing PD. Further studies are needed to understand the pathophysiological mechanism of action of serum iron on PD risk before recommendations can be made.
Collapse
Affiliation(s)
- Irene Pichler
- Respiratory Epidemiology and Public Health, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Fabiola Del Greco M.
- Respiratory Epidemiology and Public Health, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Martin Gögele
- Respiratory Epidemiology and Public Health, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Christina M. Lill
- Neuropsychiatric Genetics Group, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
- Department of Neurology, Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Lars Bertram
- Neuropsychiatric Genetics Group, Department of Vertebrate Genomics, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Chuong B. Do
- 23andMe, Inc., Mountain View, California, United States of America
| | | | - Tatiana Foroud
- Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Richard H. Myers
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | | | - Michael Nalls
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Margaux F. Keller
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Biological Anthropology, Temple University, Philadelphia, Pennsylvania, United States of America
| | | | | | - Beben Benyamin
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
- Queensland Brain Institute, The University of Queensland, Queensland, Australia
| | - John B. Whitfield
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | | | - Peter P. Pramstaller
- Respiratory Epidemiology and Public Health, National Heart and Lung Institute, Imperial College, London, United Kingdom
- Department of Neurology, General Central Hospital, Bolzano, Italy
- Department of Neurology, University of Lübeck, Lübeck, Germany
| | - Andrew A. Hicks
- Respiratory Epidemiology and Public Health, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - John R. Thompson
- Department of Health Sciences, University of Leicester, Leicester, United Kingdom
| | - Cosetta Minelli
- Respiratory Epidemiology and Public Health, National Heart and Lung Institute, Imperial College, London, United Kingdom
| |
Collapse
|
20
|
Li GF, Pan YZ, Sirois P, Li K, Xu YJ. Iron homeostasis in osteoporosis and its clinical implications. Osteoporos Int 2012; 23:2403-8. [PMID: 22525981 DOI: 10.1007/s00198-012-1982-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 02/23/2012] [Indexed: 12/14/2022]
Abstract
Osteoporosis has until now been considered to be a disease associated with abnormal calcium metabolism. However, an increasing number of clinical observations strongly suggest the association of iron overload with bone diseases, particularly in osteoporosis in menopausal women. The recent identification of hepcidin sheds new light into the crucial role of iron homeostasis in bone metabolism. Decreasing iron overload in cell studies as well as in animal experiments has been shown to improve bone cell metabolism and growth in vitro and in vivo. In view of the significant iron overload found in the aging population, especially in females, the therapeutic potential of lowering iron overload for the treatment of osteoporosis is suggested.
Collapse
Affiliation(s)
- G F Li
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | | | | | | | | |
Collapse
|
21
|
Transition metal abnormalities in progressive dementias. Biometals 2011; 25:337-50. [DOI: 10.1007/s10534-011-9504-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 10/21/2011] [Indexed: 02/06/2023]
|
22
|
Abstract
Iron accumulation in the brain and increased oxidative stress are consistent observations in many neurodegenerative diseases. Thus, we have begun examination into gene mutations or allelic variants that could be associated with loss of iron homeostasis. One of the mechanisms leading to iron overload is a mutation in the HFE gene, which is involved in iron metabolism. The 2 most common HFE gene variants are C282Y (1.9%) and H63D (8.9%). The C282Y HFE variant is more commonly associated with hereditary hemochromatosis, which is an autosomal recessive disorder, characterized by iron overload in a number of systemic organs. The H63D HFE variant appears less frequently associated with hemochromatosis, but its role in the neurodegenerative diseases has received more attention. At the cellular level, the HFE mutant protein resulting from the H63D HFE gene variant is associated with iron dyshomeostasis, increased oxidative stress, glutamate release, tau phosphorylation, and alteration in inflammatory response, each of which is under investigation as a contributing factor to neurodegenerative diseases. Therefore, the HFE gene variants are proposed to be genetic modifiers or a risk factor for neurodegenerative diseases by establishing an enabling milieu for pathogenic agents. This review will discuss the current knowledge of the association of the HFE gene variants with neurodegenerative diseases: amyotrophic lateral sclerosis, Alzheimer's disease, Parkinson's disease, and ischemic stroke. Importantly, the data herein also begin to dispel the long-held view that the brain is protected from iron accumulation associated with the HFE mutations.
Collapse
Affiliation(s)
- Wint Nandar
- Department of Neurosurgery, Pennsylvania State University, M. S. Hershey Medical Center, Hershey, PA 17033, USA
| | | |
Collapse
|
23
|
Greco V, De Marco EV, Rocca FE, Annesi F, Civitelli D, Provenzano G, Tarantino P, Scornaienchi V, Pucci F, Salsone M, Novellino F, Morelli M, Paglionico S, Gambardella A, Quattrone A, Annesi G. Association study between four polymorphisms in the HFE, TF and TFR genes and Parkinson's disease in southern Italy. Neurol Sci 2011; 32:525-7. [PMID: 21384276 DOI: 10.1007/s10072-011-0504-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Accepted: 02/16/2011] [Indexed: 12/30/2022]
Abstract
Iron overload may lead to neurodegenerative disorders such as Parkinson's disease (PD) and alterations of iron-related genes might be involved in the pathogenesis of this disease. The gene of haemochromatosis (HFE) encodes the HFE protein which interacts with the transferrin receptor (TFR), lowering its affinity for iron-bound transferrin (TF). We examined four known polymorphisms, C282Y and H63D in the HFE gene, G258S in the TF gene and S82G in the TFR gene, in 181 sporadic PD patients and 180 controls from Southern Italy to investigate their possible role in susceptibility to PD. No significant differences were found in genotype and allele frequencies between PD and controls for all the polymorphisms studied, suggesting that these variants do not contribute significantly to the risk of PD.
Collapse
Affiliation(s)
- Valentina Greco
- Institute of Neurological Sciences, National Research Council, Contrada Burga, 87050 Mangone, Cosenza, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kell DB. Towards a unifying, systems biology understanding of large-scale cellular death and destruction caused by poorly liganded iron: Parkinson's, Huntington's, Alzheimer's, prions, bactericides, chemical toxicology and others as examples. Arch Toxicol 2010; 84:825-89. [PMID: 20967426 PMCID: PMC2988997 DOI: 10.1007/s00204-010-0577-x] [Citation(s) in RCA: 286] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 07/14/2010] [Indexed: 12/11/2022]
Abstract
Exposure to a variety of toxins and/or infectious agents leads to disease, degeneration and death, often characterised by circumstances in which cells or tissues do not merely die and cease to function but may be more or less entirely obliterated. It is then legitimate to ask the question as to whether, despite the many kinds of agent involved, there may be at least some unifying mechanisms of such cell death and destruction. I summarise the evidence that in a great many cases, one underlying mechanism, providing major stresses of this type, entails continuing and autocatalytic production (based on positive feedback mechanisms) of hydroxyl radicals via Fenton chemistry involving poorly liganded iron, leading to cell death via apoptosis (probably including via pathways induced by changes in the NF-κB system). While every pathway is in some sense connected to every other one, I highlight the literature evidence suggesting that the degenerative effects of many diseases and toxicological insults converge on iron dysregulation. This highlights specifically the role of iron metabolism, and the detailed speciation of iron, in chemical and other toxicology, and has significant implications for the use of iron chelating substances (probably in partnership with appropriate anti-oxidants) as nutritional or therapeutic agents in inhibiting both the progression of these mainly degenerative diseases and the sequelae of both chronic and acute toxin exposure. The complexity of biochemical networks, especially those involving autocatalytic behaviour and positive feedbacks, means that multiple interventions (e.g. of iron chelators plus antioxidants) are likely to prove most effective. A variety of systems biology approaches, that I summarise, can predict both the mechanisms involved in these cell death pathways and the optimal sites of action for nutritional or pharmacological interventions.
Collapse
Affiliation(s)
- Douglas B Kell
- School of Chemistry and the Manchester Interdisciplinary Biocentre, The University of Manchester, Manchester M1 7DN, UK.
| |
Collapse
|
25
|
Savica R, Grossardt BR, Carlin JM, Icen M, Bower JH, Ahlskog JE, Maraganore DM, Steensma DP, Rocca WA. Anemia or low hemoglobin levels preceding Parkinson disease: a case-control study. Neurology 2009; 73:1381-7. [PMID: 19858460 DOI: 10.1212/wnl.0b013e3181bd80c1] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVE It has been suggested that anemia may be a risk factor for dementia, for restless legs syndrome, and for Parkinson disease (PD). Thus, we investigated the association of anemia with the subsequent risk of PD using a case-control study design. METHODS We used the medical records-linkage system of the Rochester Epidemiology Project to identify 196 subjects who developed PD in Olmsted County, Minnesota, from 1976 through 1995. Each incident case was matched by age (+/-1 year) and sex to a general population control. We reviewed the complete medical records of cases and controls in the system to detect anemia defined using the World Health Organization criteria. RESULTS Anemia was more common in the history of cases than of controls (odds ratio 2.00, 95% confidence interval 1.31-3.06, p = 0.001). The association remained significant after adjustment for cigarette smoking, exposure to pesticides, or hysterectomy (in women). The association was not significantly different between men and women, or between PD patients with or without rest tremor. Analyses stratified by time of onset of anemia showed a greater association for anemia that started 20 to 29 years before the onset of PD. Hemoglobin levels were slightly but consistently lower in cases than in controls across all ages. CONCLUSIONS Our results support an association between anemia experienced early in life and the later development of Parkinson disease. The interpretation of this association remains uncertain.
Collapse
Affiliation(s)
- R Savica
- Department of Neurology, College of Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Snyder AM, Connor JR. Iron, the substantia nigra and related neurological disorders. Biochim Biophys Acta Gen Subj 2008; 1790:606-14. [PMID: 18778755 DOI: 10.1016/j.bbagen.2008.08.005] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Revised: 07/29/2008] [Accepted: 08/12/2008] [Indexed: 12/14/2022]
Abstract
BACKGROUND Iron status is higher in the substantia nigra than in other brain regions but can fluctuate as function of diet and genetics and disease. Of particular note is the compartmentalization of the iron-enrichment in this region; the pars reticulata contains higher levels of stainable iron as compared to the pars compacta. The latter area is where the dopaminergic neurons reside. How this compartmentalization impacts the interpretation of data that iron contributes to cell death as in Parkinson's disease or iron deficiency contributes to altered dopaminergic activity is unknown. Nonetheless, that iron can influence neuronal cell death and dopamine function is clear. METHODS The mechanisms by which iron may be managed in the substantia nigra, particularly in the neuromelanin cells where minimal levels of ferritin the iron storage protein have been detected are addressed. The current approaches to detect iron in the substantia nigra are also reviewed. In addition, the potential mechanisms by which iron enrichment may occur in the substantia nigra are explored. GENERAL SIGNIFICANCE This review attempts to provide a critical evaluation of the many avenues of exploration into the role of iron in one of the most iron-enriched and clinically investigated areas of the brain, the substantia nigra.
Collapse
Affiliation(s)
- Amanda M Snyder
- Department of Neurosurgery, Penn State University, M. S. Hershey Medical Center, Hershey, PA 17033, USA
| | | |
Collapse
|
27
|
Rhodes SL, Ritz B. Genetics of iron regulation and the possible role of iron in Parkinson's disease. Neurobiol Dis 2008; 32:183-95. [PMID: 18675357 DOI: 10.1016/j.nbd.2008.07.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Revised: 07/01/2008] [Accepted: 07/02/2008] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is acknowledged as the second most common neurodegenerative disorder after Alzheimer's Disease. Older age may be the only unequivocal risk factor for PD although the male to female ratio is consistently greater than 1 in populations of European ancestry. Characteristic features of PD include dopaminergic neuron death in the substantia nigra (SN) pars compacta, accumulation of alpha-synuclein inclusions known as Lewy bodies in the SN, and brain iron accumulation beyond that observed in non-PD brains of a similar age. In this review article, we will provide an overview of human and animal studies investigating the contributions of iron in PD, a summary of human studies of iron-related genes in PD, a review of the literature on the genetics of iron metabolism, and some hypotheses on possible roles for iron in the pathogenic processes of PD including potential interactions between iron and other factors associated with Parkinson's disease.
Collapse
Affiliation(s)
- Shannon L Rhodes
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | |
Collapse
|
28
|
Biasiotto G, Goldwurm S, Finazzi D, Tunesi S, Zecchinelli A, Sironi F, Pezzoli G, Arosio P. HFE gene mutations in a population of Italian Parkinson's disease patients. Parkinsonism Relat Disord 2008; 14:426-30. [DOI: 10.1016/j.parkreldis.2007.10.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Revised: 10/16/2007] [Accepted: 10/31/2007] [Indexed: 12/14/2022]
|
29
|
|
30
|
Genetic predisposition to Parkinson's disease: CYP2D6 and HFE in the Faroe Islands. Pharmacogenet Genomics 2008; 18:209-12. [PMID: 18300942 DOI: 10.1097/fpc.0b013e3282f5106e] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To investigate whether the genetic variants of CYP2D6 and HFE are more frequent in Parkinson's disease (PD) patients compared with controls in a population where the prevalence of these variants and PD are increased. METHODS Blood samples were collected from 79 PD patients and 154 controls in the Faroe Islands. Genotyping for the 'CYP2D6*3, *4, *6 and *9' alleles and for the C282Y and H63D mutations were performed by real-time polymerase chain reaction before Taqman assessment. RESULTS The frequency of CYP2D6 poor metabolizers among the patients was not higher compared with the frequency found in the control group (chi2 test, P=0.86). The odds ratio was 0.92 (95% confidence interval: 0.44-1.90). Neither was a difference in HFE genotype or allele frequencies found between the patients and the controls, and the C282Y and H63D mutation carrier frequencies did not reveal any difference (chi2 test, P=0.50 and 0.60, respectively). CONCLUSION This study does not support an association between PD and mutations of the CYP2D6 and HFE genes, although a weak association cannot be excluded. The high frequency of PD in the Faroes is most likely the result of interactions between multiple genetic and environmental factors, still to be identified.
Collapse
|
31
|
Jiang H, Song N, Wang J, Ren LY, Xie JX. Peripheral iron dextran induced degeneration of dopaminergic neurons in rat substantia nigra. Neurochem Int 2007; 51:32-6. [PMID: 17490790 DOI: 10.1016/j.neuint.2007.03.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2007] [Revised: 03/26/2007] [Accepted: 03/28/2007] [Indexed: 01/08/2023]
Abstract
Iron accumulation is considered to be involved in the pathogenesis of Parkinson's disease. To demonstrate the relationship between peripheral iron overload and dopaminergic neuron loss in rat substantia nigra (SN), in the present study we used fast cyclic voltammetry, tyrosine hydroxylase (TH) immunohistochemistry, Perls' iron staining, and high performance liquid chromatography-electrochemical detection to study the degeneration of dopaminergic neurons and increased iron content in the SN of iron dextran overloaded animals. The findings showed that peripheral iron dextran overload increased the iron staining positive cells and reduced the number of TH-immunoreactive neurons in the SN. As a result, dopamine release and content, as well as its metabolites contents were decreased in caudate putamen. Even more dramatic changes were found in chronic overload group. These results suggest that peripheral iron dextran can increase the iron level in the SN, where excessive iron causes the degeneration of dopaminergic neurons. The chronic iron overload may be more destructive to dopaminergic neurons than the acute iron overload.
Collapse
Affiliation(s)
- Hong Jiang
- Department of Physiology, Medical College of Qingdao University, No. 308 Ningxia Road, Qingdao 266071, China
| | | | | | | | | |
Collapse
|