#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Serum Iron Levels and the Risk of Parkinson Disease: A Mendelian Randomization Study


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.

Please see later in the article for the Editors' Summary


Vyšlo v časopise: Serum Iron Levels and the Risk of Parkinson Disease: A Mendelian Randomization Study. PLoS Med 10(6): e32767. doi:10.1371/journal.pmed.1001462
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pmed.1001462

Souhrn

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.

Please see later in the article for the Editors' Summary


Zdroje

1. CrichtonRR, DexterDT, WardRJ (2011) Brain iron metabolism and its perturbation in neurological diseases. J Neural Transm 118: 301–314.

2. ZeccaL, YoudimMB, RiedererP, ConnorJR, CrichtonRR (2004) Iron, brain ageing and neurodegenerative disorders. Nat Rev Neurosci 5: 863–873.

3. DusekP, JankovicJ, LeW (2012) Iron dysregulation in movement disorders. Neurobiol Dis 46: 1–18.

4. FornoLS (1996) Neuropathology of Parkinson's disease. J Neuropathol Exp Neurol 55: 259–272.

5. SpillantiniMG, SchmidtML, LeeVM, TrojanowskiJQ, JakesR, et al. (1997) Alpha-synuclein in Lewy bodies. Nature 388: 839–840.

6. MarianiS, VentrigliaM, SimonelliI, DonnoS, BucossiS, et al. (2013) Fe and Cu do not differ in Parkinson's disease: a replication study plus meta-analysis. Neurobiol Aging 34: 632–633.

7. Davey SmithG, EbrahimS (2005) What can mendelian randomisation tell us about modifiable behavioural and environmental exposures? BMJ 330: 1076–1079.

8. Davey SmithG, EbrahimS (2003) ‘Mendelian randomization’: Can genetic epidemiology contribute to understanding environmental determinants of disease? Int J Epidemiol 32: 1–22.

9. PierceBL, AhsanH, VanderweeleTJ (2011) Power and instrument strength requirements for mendelian randomization studies using multiple genetic variants. Int J Epidemiol 40: 740–752.

10. PalmerTM, LawlorDA, HarbordRM, SheehanNA, TobiasJH, et al. (2012) Using multiple genetic variants as instrumental variables for modifiable risk factors. Stats Methods Med Res 21: 223–242.

11. BenyaminB, FerreiraMA, WillemsenG, GordonS, MiddelbergRP, et al. (2009) Common variants in TMPRSS6 are associated with iron status and erythrocyte volume. Nat Genet 41: 1173–1175.

12. LillCM, RoehrJT, McQueenMB, KavvouraFK, BagadeS, et al. (2012) Comprehensive research synopsis and systematic meta-analyses in parkinson's disease genetics: The PDGene database. PLoS Genet 8: e1002548 doi:10.1371/journal.pgen.1002548

13. GrecoV, De MarcoEV, RoccaFE, AnnesiF, CivitelliD, et al. (2011) Association study between four polymorphisms in the HFE, TF and TFR genes and Parkinson's disease in southern Italy. Neurol Sci 32: 525–527.

14. HallingJ, PetersenMS, GrandjeanP, WeiheP, BrosenK (2008) Genetic predisposition to parkinson's disease: CYP2D6 and HFE in the Faroe Islands. Pharmacogenet Genomics 18: 209–212.

15. GuerreiroRJ, BrasJM, SantanaI, JanuarioC, SantiagoB, et al. (2006) Association of HFE common mutations with Parkinson's disease, Alzheimer's disease and mild cognitive impairment in a portuguese cohort. BMC Neurol 6: 24.

16. DekkerMC, GiesbergenPC, NjajouOT, van SwietenJC, HofmanA, et al. (2003) Mutations in the hemochromatosis gene (HFE), Parkinson's disease and parkinsonism. Neurosci Lett 348: 117–119.

17. BorieC, GaspariniF, VerpillatP, BonnetAM, AgidY, et al. (2002) Association study between iron-related genes polymorphisms and Parkinson's disease. J Neurol 249: 801–804.

18. BuchananDD, SilburnPA, ChalkJB, Le CouteurDG, MellickGD (2002) The Cys282Tyr polymorphism in the HFE gene in Australian Parkinson's disease patients. Neurosci Lett 327: 91–94.

19. AamodtAH, StovnerLJ, ThorstensenK, LydersenS, WhiteLR, et al. (2007) Prevalence of haemochromatosis gene mutations in Parkinson's disease. J Neurol, Neurosurg Psychiatry 78: 315–317.

20. BiasiottoG, GoldwurmS, FinazziD, TunesiS, ZecchinelliA, et al. (2008) HFE gene mutations in a population of Italian Parkinson's disease patients. Parkinsonism Relat Disord 14: 426–430.

21. AkbasN, HochstrasserH, DeplazesJ, TomiukJ, BauerP, et al. (2006) Screening for mutations of the HFE gene in Parkinson's disease patients with hyperechogenicity of the substantia nigra. Neurosci Lett 407: 16–19.

22. PankratzN, BeechamGW, DeStefanoAL, DawsonTM, DohenyKF, et al. (2012) Meta-analysis of Parkinson's disease: Identification of a novel locus, RIT2. Ann Neurol 71: 370–384.

23. DoCB, TungJY, DorfmanE, KieferAK, DrabantEM, et al. (2011) Web-based genome-wide association study identifies two novel loci and a substantial genetic component for Parkinson's disease. PLoS Genet 7: e1002141 doi:10.1371/journal.pgen.1002141

24. International Parkinson Disease Genomics Consortium (2011) NallsMA, PlagnolV, HernandezDG, SharmaM, et al. (2011) Imputation of sequence variants for identification of genetic risks for Parkinson's disease: A meta-analysis of genome-wide association studies. Lancet 377: 641–649.

25. International Parkinson's Disease Genomics Consortium (IPDGC), Wellcome Trust Case Control Consortium 2 (WTCCC2) (2011) A two-stage meta-analysis identifies several new loci for Parkinson's disease. PLoS Genet 7: e1002142 doi:10.1371/journal.pgen.1002142

26. PankratzN, WilkJB, LatourelleJC, DeStefanoAL, HalterC, et al. (2009) Genomewide association study for susceptibility genes contributing to familial Parkinson disease. Hum Genet 124: 593–605.

27. FungHC, ScholzS, MatarinM, Simon-SanchezJ, HernandezD, et al. (2006) Genome-wide genotyping in Parkinson's disease and neurologically normal controls: first stage analysis and public release of data. Lancet Neurol 5: 911–916.

28. Simon-SanchezJ, SchulteC, BrasJM, SharmaM, GibbsJR, et al. (2009) Genome-wide association study reveals genetic risk underlying Parkinson's disease. Nat Genet 41: 1308–1312.

29. EdwardsTL, ScottWK, AlmonteC, BurtA, PowellEH, et al. (2010) Genome-wide association study confirms SNPs in SNCA and the MAPT region as common risk factors for Parkinson disease. Ann Hum Genet 74: 97–109.

30. HamzaTH, ZabetianCP, TenesaA, LaederachA, MontimurroJ, et al. (2010) Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson's disease. Nat Genet 42: 781–785.

31. HigginsJP, ThompsonSG, DeeksJJ, AltmanDG (2003) Measuring inconsistency in meta-analyses. BMJ 327: 557–560.

32. D DidelezV, MengS (2010) Assumptions of IV methods for observational epidemiology. Stat Sci 25: 22–40.

33. BautistaLE, SmeethL, HingoraniAD, CasasJP (2006) Estimation of bias in nongenetic observational studies using “mendelian triangulation”. Ann Epidemiol 16: 675–680.

34. ThomasDC, LawlorDA, ThompsonJR (2007) Re: Estimation of bias in nongenetic observational studies using “mendelian triangulation” by Bautista et al. Ann Epidemiol 17: 511–513.

35. PierceBL, AhsanH, VanderweeleTJ (2011) Power and instrument strength requirements for Mendelian randomization studies using multiple genetic variants. Int J Epidemiol 40: 740–752.

36. TannerCM, GoldmanSM (1996) Epidemiology of Parkinson's disease. Neurol Clin 14: 317–335.

37. RitzB, AscherioA, CheckowayH, MarderKS, NelsonLM, et al. (2007) Pooled analysis of tobacco use and risk of Parkinson disease. Arch Neurol 64: 990–997.

38. HernanMA, TakkoucheB, Caamano-IsornaF, Gestal-OteroJJ (2002) A meta-analysis of coffee drinking, cigarette smoking, and the risk of Parkinson's disease. Ann Neurol 52: 276–284.

39. LinertW, BridgeMH, HuberM, BjugstadKB, GrossmanS, et al. (1999) In vitro and in vivo studies investigating possible antioxidant actions of nicotine: Relevance to Parkinson's and Alzheimer's diseases. Biochim Biophy Acta 1454: 143–152.

40. MorckTA, LynchSR, CookJD (1983) Inhibition of food iron absorption by coffee. Am J Clin Nutr 37: 416–420.

41. ZijpIM, KorverO, TijburgLB (2000) Effect of tea and other dietary factors on iron absorption. Crit Rev Food Sci Nutr 40: 371–398.

42. KupershmidtL, AmitT, Bar-AmO, YoudimMB, WeinrebO (2012) Neuroprotection by the multitarget iron chelator M30 on age-related alterations in mice. Mech Ageing Dev 133: 267–274.

43. BergD, RoggendorfW, SchroderU, KleinR, TatschnerT, et al. (2002) Echogenicity of the substantia nigra: Association with increased iron content and marker for susceptibility to nigrostriatal injury. Arch Neurol 59: 999–1005.

44. WalterU, WittR, WoltersA, WittstockM, BeneckeR (2012) Substantia nigra echogenicity in Parkinson's disease: Relation to serum iron and C-reactive protein. J Neural Transm 119: 53–57.

45. LogroscinoG, ChenH, WingA, AscherioA (2006) Blood donations, iron stores, and risk of Parkinson's disease. Mov Disord 21: 835–838.

46. SavicaR, GrossardtBR, CarlinJM, IcenM, BowerJH, et al. (2009) Anemia or low hemoglobin levels preceding Parkinson disease: a case-control study. Neurology 73: 1381–1387.

47. LevensonCW, CutlerRG, LadenheimB, CadetJL, HareJ, et al. (2004) Role of dietary iron restriction in a mouse model of Parkinson's disease. Exp Neurol 190: 506–514.

48. MiyakeY, TanakaK, FukushimaW, SasakiS, KiyoharaC, et al. (2011) Dietary intake of metals and risk of Parkinson's disease: a case-control study in Japan. J Neurol Sci 306: 98–102.

49. RamseyAJ, HillasPJ, FitzpatrickPF (1996) Characterization of the active site iron in tyrosine hydroxylase. Redox states of the iron. J Biol Chem 271: 24395–24400.

50. BeardJ, EriksonKM, JonesBC (2003) Neonatal iron deficiency results in irreversible changes in dopamine function in rats. J Nutr 133: 1174–1179.

51. LevensonCW, TassabehjiNM (2004) Iron and ageing: an introduction to iron regulatory mechanisms. Ageing Res Rev 3: 251–263.

52. RhodesSL, RitzB (2008) Genetics of iron regulation and the possible role of iron in Parkinson's disease. Neurobiol Dis 32: 183–195.

53. CastiglioniE, FinazziD, GoldwurmS, PezzoliG, ForniG, et al. (2010) Analysis of nucleotide variations in genes of iron management in patients of Parkinson's disease and other movement disorders. Parkinsons Dis 2011: 827693.

54. CastiglioniE, FinazziD, GoldwurmS, LeviS, PezzoliG, et al. (2010) Sequence variations in mitochondrial ferritin: Distribution in healthy controls and different types of patients. Genet Test Mol Biomarkers 14: 793–796.

55. EzquerraM, CampdelacreuJ, MunozE, TolosaE (2005) Association study of the G258S transferrin gene polymorphism and Parkinson's disease in the Spanish population. J Neurol 252: 1269–1270.

56. HeQ, DuT, YuX, XieA, SongN, et al. (2011) DMT1 polymorphism and risk of Parkinson's disease. Neurosci Lett 501: 128–131.

57. FederJN, GnirkeA, ThomasW, TsuchihashiZ, RuddyDA, et al. (2003) The discovery of the new haemochromatosis gene. 1996. J Hepatol 38: 704–709.

58. DuX, SheE, GelbartT, TruksaJ, LeeP, et al. (2008) The serine protease TMPRSS6 is required to sense iron deficiency. Science 320: 1088–1092.

59. FederJN, GnirkeA, ThomasW, TsuchihashiZ, RuddyDA, et al. (1996) A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis. Nat Genet 13: 399–408.

60. BradleyLA, JohnsonDD, PalomakiGE, HaddowJE, RobertsonNH, eta (1998) Hereditary haemochromatosis mutation frequencies in the general population. J Med Screen 5: 34–36.

61. WaheedA, ParkkilaS, ZhouXY, TomatsuS, TsuchihashiZ, et al. (1997) Hereditary hemochromatosis: effects of C282Y and H63D mutations on association with beta2-microglobulin, intracellular processing, and cell surface expression of the HFE protein in COS-7 cells. Proc Natl Acad Sci U S A 94: 12384–12389.

62. FederJN, TsuchihashiZ, IrrinkiA, LeeVK, MapaFA, et al. (1997) The hemochromatosis founder mutation in HLA-H disrupts beta2-microglobulin interaction and cell surface expression. J Biol Chem 272: 14025–14028.

63. LebrónJA, BennettMJ, VaughnDE, ChirinoAJ, SnowPM, et al. (1998) Crystal structure of the hemochromatosis protein HFE and characterization of its interaction with transferrin receptor. Cell 93: 111–123.

64. AnP, WuQ, WangH, GuanY, MuM, et al. (2012) TMPRSS6, but not TF, TFR2 or BMP2 variants are associated with increased risk of iron-deficiency anemia. Hum Mol Genet 21: 2124–2131.

65. NaiA, PaganiA, SilvestriL, CampostriniN, CorbellaM, et al. (2011) TMPRSS6 rs855791 modulates hepcidin transcription in vitro and serum hepcidin levels in normal individuals. Blood 118: 4459–4462.

66. FleissJL (1993) The statistical basis of meta-analysis. Stat Methods Med Res 2: 121–145.

67. TamuraT, GoldenbergRL, HouJ, JohnstonKE, CliverSP, RameySL, et al. (2002) Cord serum ferritin concentrations and mental and psychomotor development of children at five years of age. J Ped 186: 458–463.

68. HarbordRM, DidelezV, PalmerTM, MengS, SterneJA, et al. (2013) Severity of bias of a simple estimator of the causal odds ratio in mendelian randomization studies. Stat Med 32: 1246-1258.

Štítky
Interné lekárstvo

Článok vyšiel v časopise

PLOS Medicine


2013 Číslo 6
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#