Elsevier

Neurobiology of Disease

Volume 62, February 2014, Pages 172-178
Neurobiology of Disease

Pooled analysis of iron-related genes in Parkinson's disease: Association with transferrin

https://doi.org/10.1016/j.nbd.2013.09.019Get rights and content

Highlights

  • 16 iron homeostasis genes were considered for association with Parkinson's disease.

  • Data from 3 case–control studies (United States, Australia, and France) were pooled.

  • 3′ region of transferrin (TF) was associated with PD (OR = 0.83 [0.71–0.96]).

  • Transferrin receptor 2 (TFR2) genetic variation may also be associated with PD.

  • TF-TFR2 findings suggest roles for iron misregulation or mitochondrial dysfunction.

Abstract

Pathologic features of Parkinson's disease (PD) include death of dopaminergic neurons in the substantia nigra, presence of α-synuclein containing Lewy bodies, and iron accumulation in PD-related brain regions. The observed iron accumulation may be contributing to PD etiology but it also may be a byproduct of cell death or cellular dysfunction. To elucidate the possible role of iron accumulation in PD, we investigated genetic variation in 16 genes related to iron homeostasis in three case–control studies from the United States, Australia, and France. After screening 90 haplotype tagging single nucleotide polymorphisms (SNPs) within the genes of interest in the US study population, we investigated the five most promising gene regions in two additional independent case–control studies. For the pooled data set (1289 cases, 1391 controls) we observed a protective association (OR = 0.83, 95% CI: 0.71–0.96) between PD and a haplotype composed of the A allele at rs1880669 and the T allele at rs1049296 in transferrin (TF; GeneID: 7018). Additionally, we observed a suggestive protective association (OR = 0.87, 95% CI: 0.74-1.02) between PD and a haplotype composed of the G allele at rs10247962 and the A allele at rs4434553 in transferrin receptor 2 (TFR2; GeneID: 7036). We observed no associations in our pooled sample for haplotypes in SLC40A1, CYB561, or HFE. Taken together with previous findings in model systems, our results suggest that TF or a TF-TFR2 complex may have a role in the etiology of PD, possibly through iron misregulation or mitochondrial dysfunction within dopaminergic neurons.

Introduction

Brain iron accumulation, beyond that seen in age-matched controls, is frequently observed in Parkinson's disease (PD) affected brains (reviewed in (Gerlach et al., 2006)). Histochemical comparisons of postmortem brain tissue have found increased levels of iron deposits in the substantia nigra (SN) of Parkinsonian brains (Dexter et al., 1987, Sofic et al., 1991) and specifically in neurons of the SN (Oakley et al., 2007). These pathologic findings are further supported by magnetic resonance imaging (Bartzokis et al., 1999, Gorell et al., 1995, Martin et al., 2008, Michaeli et al., 2007) and transcranial ultrasound (Becker et al., 1995, Mehnert et al., 2010, Walter et al., 2002) studies that observed iron deposits in the SN in living idiopathic PD patients.

It is debatable whether this accumulation of iron is a cause, co-factor, or consequence of the dopaminergic (DA) cell death in PD. Iron is a very potent oxidation–reduction agent that can create oxidative stress in cells and prior work suggests that neurons may be more sensitive to alterations in iron (LaVaute et al., 2001, Moos et al., 1998) than other cell types in the brain. Iron is also hypothesized to aggravate some key pathogenic processes related to PD including alpha-synuclein fibril formation (Olivares et al., 2009, Uversky et al., 2001) and mitochondrial dysfunction (Horowitz and Greenamyre, 2010, Lin et al., 2001). Finally, iron may simply be a remnant of neuronal cell death (He et al., 2003).

Animal models provide some potential insight. Genetic knockouts of some iron-related genes, when not lethal, produce altered brain iron levels (LaVaute et al., 2001, Moos and Morgan, 2004, Patel et al., 2002). Iron administration to unaltered animals has resulted in brain iron accumulation (Sengstock et al., 1993) or iron accumulation with a decrease in dopamine levels (Sziraki et al., 1998, Wesemann et al., 1994). Iron feeding studies in animals have also observed decreased dopamine levels with a large excess of dietary iron (Kaur et al., 2007) and with iron in combination with toxins (Levenson et al., 2004, Peng et al., 2007). These observations suggest that iron is either a cause or a co-factor, not a consequence, of dopaminergic cells damage, possibly due to the role iron plays in the synthesis of tyrosine hydroxylase (Snyder and Connor, 2009). Therefore, we hypothesize that small imbalances in ferrous or ferric brain iron contribute to one or more of the pathogenic processes contributing to neurodegeneration in PD and propose to investigate this hypothesis by evaluating the associations between PD and iron-related genes.

To date, epidemiologic studies investigating associations between occupational or dietary iron exposure and PD have been unpersuasive, and the few reports of iron-related genes and PD have been predominantly in small studies, investigating rare exonic SNPs, and inconclusive (reviewed in (Rhodes and Ritz, 2008)). Nevertheless, studies in experimental models of PD support a role for iron in the etiology or progression of PD (e.g. (Ben-Shachar and Youdim, 1991, Fredriksson et al., 2001, Kaur et al., 2003)). To facilitate the investigation of the iron-PD hypothesis, we pooled data from three independent case–control studies: one each from the United States (US), Australia (AU), and France (FR). All studies were designed for the investigation of genetic variation; the US and FR studies were also designed to investigate the influence of pesticide exposure on PD etiology and, therefore, include subjects with an increased likelihood of exposure to pesticides. In this report, we present the results from our two-phase study of promoter region, intronic, and exonic SNPs specifically selected to span each of 16 iron-related candidate genes and their associations with PD in a pooled sample of 1286 idiopathic PD cases and 1391 controls.

Section snippets

Methods

For our two-phase design we screened a larger number of genetic variants in an initial study, the US Study described below, followed by genotyping of selected variants in two additional studies based on findings from the initial study. All three studies (Supplemental Table 1) were pooled for the final analysis.

Phase 1, screening in US Study

From the 84 SNPs screened in the US Study (phase 1; Supplementary Table 2), 12 out of 31 regions (haplotype blocks) contained one or more SNPs with a sufficient screening p-value for consideration in the phase 1 haplotype analysis. These 12 regions were in 10 of the 15 genes evaluated in the US Study: FRRS1, SLC40A1, TF (2 regions), CP (2 regions), HFE, TFR2, IREB2, CYB561, FECH, and HAMP (no SNPs in LTF, TFRC, FTMT, SLC11A2, and HEPH met the screening criterion). In the haplotype analysis of

Discussion

Based on the well-documented accumulation of iron in the substantia nigra (SN) of Parkinson's disease (PD) brains we pursued a hypothesis generating effort investigating genetic variants of iron-related genes for association with PD. Our results are intriguing and suggest of a role for genetic variants in transferrin and transferrin receptor 2 in the etiology or pathology of PD. While these findings must be considered exploratory, we observed a 15–20% decrease in risk of PD for subjects with

Acknowledgments

We are exceptionally grateful to all study participants, whose generosity made this research possible. We thank Drs. Jerome Rotter and Kent Taylor (Cedars-Sinai Medical Center) for their generous support in genotyping the US Study. The US study was funded in part by NIEHS Grants R01-ES010544, U54-ES012078, P01-ES016732; the Michael J. Fox Foundation; The Parkinson Alliance; the American Parkinson Disease Association; and the Veterans Administration Healthcare System (SW PADRECC). The Australian

References (76)

  • C.W. Levenson et al.

    Role of dietary iron restriction in a mouse model of Parkinson's disease

    Exp. Neurol.

    (2004)
  • E. Lin et al.

    Regulation of the 75-Kda subunit of mitochondrial complex I by iron

    J. Biol. Chem.

    (2001)
  • P.G. Mastroberardino et al.

    A novel transferrin/Tfr2-mediated mitochondrial iron transport system is disrupted in Parkinson's disease

    Neurobiol. Dis.

    (2009)
  • S. Purcell et al.

    Plink: a tool set for whole-genome association and population-based linkage analyses

    Am. J. Hum. Genet.

    (2007)
  • S.L. Rhodes et al.

    Genetics of iron regulation and the possible role of iron in Parkinson's disease

    Neurobiol. Dis.

    (2008)
  • D.J. Schaid et al.

    Score tests for association between traits and haplotypes when linkage phase is ambiguous

    Am. J. Hum. Genet.

    (2002)
  • A.M. Snyder et al.

    Iron, the substantia nigra and related neurological disorders

    Biochim. Biophys. Acta

    (2009)
  • I. Sziraki et al.

    Manganese: a transition metal protects nigrostriatal neurons from oxidative stress in the iron-induced animal model of Parkinsonism

    Neuroscience

    (1998)
  • T. Tanaka et al.

    A genome-wide association analysis of serum iron concentrations

    Blood

    (2010)
  • V.N. Uversky et al.

    Metal-triggered structural transformations, aggregation, and fibrillation of human alpha-synuclein. a possible molecular Nk between Parkinson's disease and heavy metal exposure

    J. Biol. Chem.

    (2001)
  • J.M. Vance et al.

    Gene-environment interactions in Parkinson's disease and other forms of Parkinsonism

    Neurotoxicology

    (2010)
  • J.D. Wall et al.

    Assessing the performance of the haplotype block model of linkage disequilibrium

    Am. J. Hum. Genet.

    (2003)
  • J.B. Whitfield et al.

    Effects of Hfe C282y and H63d polymorphisms and polygenic background on iron stores in a large community sample of twins

    Am. J. Hum. Genet.

    (2000)
  • X. Zhang et al.

    Neuroprotection by iron chelator against proteasome inhibitor-induced nigral degeneration

    Biochem. Biophys. Res. Commun.

    (2005)
  • P. Aisen

    The G277s mutation in transferrin does not disturb function

    Br. J. Haematol.

    (2003)
  • J.K. Andersen

    Paraquat and iron exposure as possible synergistic environmental risk factors in Parkinson's disease

    Neurotox. Res.

    (2003)
  • J.C. Barrett et al.

    Haploview: analysis and visualization of Ld and haplotype maps

    Bioinformatics

    (2005)
  • G. Becker et al.

    Degeneration of substantia nigra in chronic Parkinson's disease visualized by transcranial color-coded real-time sonography

    Neurology

    (1995)
  • D. Ben-Shachar et al.

    Intranigral iron injection induces behavioral and biochemical “Parkinsonism” in rats

    J. Neurochem.

    (1991)
  • B. Benyamin et al.

    Common variants in Tmprss6 are associated with iron status and erythrocyte volume

    Nat. Genet.

    (2009)
  • R. Blanco-Rojo et al.

    Four variants in transferrin and Hfe genes as potential markers of iron deficiency anaemia risk: an association study in menstruating women

    Nutr. Metab. (Lond.)

    (2011)
  • J.H. Bower et al.

    Incidence and distribution of Parkinsonism in Olmsted County, Minnesota, 1976–1990

    Neurology

    (1999)
  • S. Costello et al.

    Parkinson's disease and residential exposure to maneb and paraquat from agricultural applications in the Central Valley of California

    Am. J. Epidemiol.

    (2009)
  • D.C. Crawford et al.

    Definition and clinical importance of haplotypes

    Annu. Rev. Med.

    (2005)
  • M.J. Daly et al.

    High-resolution haplotype structure in the human genome

    Nat. Genet.

    (2001)
  • J. Delanghe et al.

    Human transferrin G277s mutation and iron deficiency in pregnancy

    Br. J. Haematol.

    (2006)
  • H. Drakesmith et al.

    Hepcidin and the iron-infection axis

    Science

    (2012)
  • F. Dutheil et al.

    Interaction between Abcb1 and professional exposure to organochlorine insecticides in Parkinson disease

    Arch. Neurol.

    (2010)
  • Cited by (0)

    View full text