Skip to main content
Top
Published in: Current Neurology and Neuroscience Reports 1/2016

01-01-2016 | Movement Disorders (S Fox, Section Editor)

Neurodegeneration with Brain Iron Accumulation

Author: Susanne A. Schneider

Published in: Current Neurology and Neuroscience Reports | Issue 1/2016

Login to get access

Abstract

Syndromes with neurodegeneration with brain iron accumulation (NBIA) are a group of neurodegenerative disorders characterized by abnormalities in brain iron metabolism with excess iron accumulation in the globus pallidus and to a lesser degree in the substantia nigra and sometimes adjacent areas. They clinically present as neurodegenerative diseases with progressive hypo- and/or hyperkinetic movement disorders and a variable degree of pyramidal, cerebellar, peripheral nerve, autonomic, cognitive and psychiatric involvement, and visual dysfunction. Several causative genes underlying NBIA have been identified which explain about 65 % of cases. Pathophysiologically, many of the NBIA syndromes map into related biochemical pathways and gene networks including mitochondrial pathways, lipid metabolism, and autophagy. Treatment for NBIA disorders remains symptomatic but a placebo-controlled double-blind study is underway. Rapid developments prompted the review of this interesting field.
Literature
2.
go back to reference Hayflick SJ. Neurodegeneration with brain iron accumulation: from genes to pathogenesis. Semin Pediatr Neurol. 2006;13:182–5.PubMedCrossRef Hayflick SJ. Neurodegeneration with brain iron accumulation: from genes to pathogenesis. Semin Pediatr Neurol. 2006;13:182–5.PubMedCrossRef
3.
go back to reference Hayflick SJ, Westaway SK, Levinson B, et al. Genetic, clinical, and radiographic delineation of Hallervorden-Spatz syndrome. N Engl J Med. 2003;348:33–40.PubMedCrossRef Hayflick SJ, Westaway SK, Levinson B, et al. Genetic, clinical, and radiographic delineation of Hallervorden-Spatz syndrome. N Engl J Med. 2003;348:33–40.PubMedCrossRef
4.
go back to reference Schneider SA, Aggarwal A, Bhatt M, et al. Severe tongue protrusion dystonia: clinical syndromes and possible treatment. Neurology. 2006;67:940–3.PubMedCrossRef Schneider SA, Aggarwal A, Bhatt M, et al. Severe tongue protrusion dystonia: clinical syndromes and possible treatment. Neurology. 2006;67:940–3.PubMedCrossRef
5.
go back to reference Marelli C, Piacentini S, Garavaglia B, Girotti F, Albanese A. Clinical and neuropsychological correlates in two brothers with pantothenate kinase-associated neurodegeneration. Movement Disorders: Off J Movement Disorder Soc. 2005;20:208–12.CrossRef Marelli C, Piacentini S, Garavaglia B, Girotti F, Albanese A. Clinical and neuropsychological correlates in two brothers with pantothenate kinase-associated neurodegeneration. Movement Disorders: Off J Movement Disorder Soc. 2005;20:208–12.CrossRef
6.
go back to reference Thomas M, Hayflick SJ, Jankovic J. Clinical heterogeneity of neurodegeneration with brain iron accumulation (Hallervorden-Spatz syndrome) and pantothenate kinase-associated neurodegeneration. Movement Disorders: Off J Movement Disorder Soc. 2004;19:36–42.CrossRef Thomas M, Hayflick SJ, Jankovic J. Clinical heterogeneity of neurodegeneration with brain iron accumulation (Hallervorden-Spatz syndrome) and pantothenate kinase-associated neurodegeneration. Movement Disorders: Off J Movement Disorder Soc. 2004;19:36–42.CrossRef
7.
go back to reference Egan RA, Weleber RG, Hogarth P, et al. Neuro-ophthalmologic and electroretinographic findings in pantothenate kinase-associated neurodegeneration (formerly Hallervorden-Spatz syndrome). Am J Ophthalmol. 2005;140:267–74.PubMedPubMedCentralCrossRef Egan RA, Weleber RG, Hogarth P, et al. Neuro-ophthalmologic and electroretinographic findings in pantothenate kinase-associated neurodegeneration (formerly Hallervorden-Spatz syndrome). Am J Ophthalmol. 2005;140:267–74.PubMedPubMedCentralCrossRef
8.
9.
go back to reference Yoon WT, Lee WY, Shin HY, Lee ST, Ki CS. Novel PANK2 gene mutations in Korean patient with pantothenate kinase-associated neurodegeneration presenting unilateral dystonic tremor. Movement Disorders: Off J Movement Disorder Soc. 2010;25:245–7.CrossRef Yoon WT, Lee WY, Shin HY, Lee ST, Ki CS. Novel PANK2 gene mutations in Korean patient with pantothenate kinase-associated neurodegeneration presenting unilateral dystonic tremor. Movement Disorders: Off J Movement Disorder Soc. 2010;25:245–7.CrossRef
10.
go back to reference Aggarwal A, Schneider SA, Houlden H, et al. Indian-subcontinent NBIA: unusual phenotypes, novel PANK2 mutations, and undetermined genetic forms. Movement Disorders: Off J Movement Disorder Soc. 2010;25:1424–31.CrossRef Aggarwal A, Schneider SA, Houlden H, et al. Indian-subcontinent NBIA: unusual phenotypes, novel PANK2 mutations, and undetermined genetic forms. Movement Disorders: Off J Movement Disorder Soc. 2010;25:1424–31.CrossRef
11.
go back to reference Chung SJ, Lee JH, Lee MC, Yoo HW, Kim GH. Focal hand dystonia in a patient with PANK2 mutation. Movement Disorders: Off J Movement Disorder Soc. 2008;23:466–8.CrossRef Chung SJ, Lee JH, Lee MC, Yoo HW, Kim GH. Focal hand dystonia in a patient with PANK2 mutation. Movement Disorders: Off J Movement Disorder Soc. 2008;23:466–8.CrossRef
12.
go back to reference Vasconcelos OM, Harter DH, Duffy C, et al. Adult Hallervorden-Spatz syndrome simulating amyotrophic lateral sclerosis. Muscle Nerve. 2003;28:118–22.PubMedCrossRef Vasconcelos OM, Harter DH, Duffy C, et al. Adult Hallervorden-Spatz syndrome simulating amyotrophic lateral sclerosis. Muscle Nerve. 2003;28:118–22.PubMedCrossRef
13.
go back to reference Antonini A, Goldwurm S, Benti R, et al. Genetic, clinical, and imaging characterization of one patient with late-onset, slowly progressive, pantothenate kinase-associated neurodegeneration. Movement Disorders: Off J Movement Disorder Soc. 2006;21:417–8.CrossRef Antonini A, Goldwurm S, Benti R, et al. Genetic, clinical, and imaging characterization of one patient with late-onset, slowly progressive, pantothenate kinase-associated neurodegeneration. Movement Disorders: Off J Movement Disorder Soc. 2006;21:417–8.CrossRef
14.
go back to reference Seo JH, Song SK, Lee PH. A novel PANK2 mutation in a patient with atypical pantothenate-kinase-associated neurodegeneration presenting with adult-onset Parkinsonism. J Clin Neurology. 2009;5:192–4.CrossRef Seo JH, Song SK, Lee PH. A novel PANK2 mutation in a patient with atypical pantothenate-kinase-associated neurodegeneration presenting with adult-onset Parkinsonism. J Clin Neurology. 2009;5:192–4.CrossRef
15.
go back to reference del Valle-Lopez P, Perez-Garcia R, Sanguino-Andres R, Gonzalez-Pablos E. Adult onset Hallervorden-Spatz disease with psychotic symptoms. Actas Esp Psiquiatr. 2011;39:260–2.PubMed del Valle-Lopez P, Perez-Garcia R, Sanguino-Andres R, Gonzalez-Pablos E. Adult onset Hallervorden-Spatz disease with psychotic symptoms. Actas Esp Psiquiatr. 2011;39:260–2.PubMed
16.
17.
go back to reference Hartig MB, Hortnagel K, Garavaglia B, et al. Genotypic and phenotypic spectrum of PANK2 mutations in patients with neurodegeneration with brain iron accumulation. Ann Neurol. 2006;59:248–56.PubMedCrossRef Hartig MB, Hortnagel K, Garavaglia B, et al. Genotypic and phenotypic spectrum of PANK2 mutations in patients with neurodegeneration with brain iron accumulation. Ann Neurol. 2006;59:248–56.PubMedCrossRef
18.
go back to reference Dusi S, Valletta L, Haack TB, et al. Exome sequence reveals mutations in CoA synthase as a cause of neurodegeneration with brain iron accumulation. Am J Hum Genet. 2014;94:11–22.PubMedPubMedCentralCrossRef Dusi S, Valletta L, Haack TB, et al. Exome sequence reveals mutations in CoA synthase as a cause of neurodegeneration with brain iron accumulation. Am J Hum Genet. 2014;94:11–22.PubMedPubMedCentralCrossRef
19.
go back to reference Santambrogio P, Dusi S, Guaraldo M, et al. Mitochondrial iron and energetic dysfunction distinguish fibroblasts and induced neurons from pantothenate kinase-associated neurodegeneration patients. Neurobiology of Disease 2015. Santambrogio P, Dusi S, Guaraldo M, et al. Mitochondrial iron and energetic dysfunction distinguish fibroblasts and induced neurons from pantothenate kinase-associated neurodegeneration patients. Neurobiology of Disease 2015.
20.
21.
go back to reference Paisan-Ruiz C, Bhatia KP, Li A, et al. Characterization of PLA2G6 as a locus for dystonia-parkinsonism. Ann Neurol. 2009;65:19–23.PubMedCrossRef Paisan-Ruiz C, Bhatia KP, Li A, et al. Characterization of PLA2G6 as a locus for dystonia-parkinsonism. Ann Neurol. 2009;65:19–23.PubMedCrossRef
22.
go back to reference Dezfouli MA, Alavi A, Rohani M, et al. PANK2 and C19orf12 mutations are common causes of neurodegeneration with brain iron accumulation. Movement Disorders: Off J Movement Disorder Soc. 2013;28:228–32.CrossRef Dezfouli MA, Alavi A, Rohani M, et al. PANK2 and C19orf12 mutations are common causes of neurodegeneration with brain iron accumulation. Movement Disorders: Off J Movement Disorder Soc. 2013;28:228–32.CrossRef
23.
go back to reference Goldman JG, Eichenseer SR, Berry-Kravis E, et al. Clinical features of neurodegeneration with brain iron accumulation due to a C19orf12 gene mutation. Movement Disorders: Off J Movement Disorder Soc. 2013;28:1462–3.CrossRef Goldman JG, Eichenseer SR, Berry-Kravis E, et al. Clinical features of neurodegeneration with brain iron accumulation due to a C19orf12 gene mutation. Movement Disorders: Off J Movement Disorder Soc. 2013;28:1462–3.CrossRef
24.•
go back to reference Hogarth P, Gregory A, Kruer MC, et al. New NBIA subtype: genetic, clinical, pathologic, and radiographic features of MPAN. Neurology. 2013;80:268–75. The authors perform a genetic screening study and demonstrate that mutations in C19orf12 are a major cause of NBIA. Clinical and radiological features of 23 MPAN patients are summarized. Histochemical and immunohistochemical work-up of brain tissue from one patient shows widespread Lewy body pathology, thereby bridging the gap to Parkinson’s disease.PubMedPubMedCentralCrossRef Hogarth P, Gregory A, Kruer MC, et al. New NBIA subtype: genetic, clinical, pathologic, and radiographic features of MPAN. Neurology. 2013;80:268–75. The authors perform a genetic screening study and demonstrate that mutations in C19orf12 are a major cause of NBIA. Clinical and radiological features of 23 MPAN patients are summarized. Histochemical and immunohistochemical work-up of brain tissue from one patient shows widespread Lewy body pathology, thereby bridging the gap to Parkinson’s disease.PubMedPubMedCentralCrossRef
25.
go back to reference Horvath R, Holinski-Feder E, Neeve VC, et al. A new phenotype of brain iron accumulation with dystonia, optic atrophy, and peripheral neuropathy. Movement Disorders: Off J Movement Disorder Soc. 2012;27:789–93.CrossRef Horvath R, Holinski-Feder E, Neeve VC, et al. A new phenotype of brain iron accumulation with dystonia, optic atrophy, and peripheral neuropathy. Movement Disorders: Off J Movement Disorder Soc. 2012;27:789–93.CrossRef
27.
go back to reference Panteghini C, Zorzi G, Venco P, et al. C19orf12 and FA2H mutations are rare in Italian patients with neurodegeneration with brain iron accumulation. Semin Pediatr Neurol. 2012;19:75–81.PubMedCrossRef Panteghini C, Zorzi G, Venco P, et al. C19orf12 and FA2H mutations are rare in Italian patients with neurodegeneration with brain iron accumulation. Semin Pediatr Neurol. 2012;19:75–81.PubMedCrossRef
28.
go back to reference Schottmann G, Stenzel W, Lutzkendorf S, Schuelke M, Knierim E. A novel frameshift mutation of C19ORF12 causes NBIA4 with cerebellar atrophy and manifests with severe peripheral motor axonal neuropathy. Clinical genetics 2013. Schottmann G, Stenzel W, Lutzkendorf S, Schuelke M, Knierim E. A novel frameshift mutation of C19ORF12 causes NBIA4 with cerebellar atrophy and manifests with severe peripheral motor axonal neuropathy. Clinical genetics 2013.
29.
go back to reference Schulte EC, Claussen MC, Jochim A, et al. Mitochondrial membrane protein associated neurodegeneration: a novel variant of neurodegeneration with brain iron accumulation. Movement Disorders: Off J Movement Disorder Soc. 2013;28:224–7.CrossRef Schulte EC, Claussen MC, Jochim A, et al. Mitochondrial membrane protein associated neurodegeneration: a novel variant of neurodegeneration with brain iron accumulation. Movement Disorders: Off J Movement Disorder Soc. 2013;28:224–7.CrossRef
30.
go back to reference Landoure G, Zhu PP, Lourenco CM, et al. Hereditary spastic paraplegia type 43 (SPG43) is caused by mutation in C19orf12. Hum Mutat. 2013;34:1357–60.PubMedCrossRef Landoure G, Zhu PP, Lourenco CM, et al. Hereditary spastic paraplegia type 43 (SPG43) is caused by mutation in C19orf12. Hum Mutat. 2013;34:1357–60.PubMedCrossRef
31.
go back to reference Deschauer M, Gaul C, Behrmann C, Prokisch H, Zierz S, Haack TB. C19orf12 mutations in neurodegeneration with brain iron accumulation mimicking juvenile amyotrophic lateral sclerosis. J Neurol. 2012;259:2434–9.PubMedCrossRef Deschauer M, Gaul C, Behrmann C, Prokisch H, Zierz S, Haack TB. C19orf12 mutations in neurodegeneration with brain iron accumulation mimicking juvenile amyotrophic lateral sclerosis. J Neurol. 2012;259:2434–9.PubMedCrossRef
32.
go back to reference Hartig MB, Iuso A, Haack T, et al. Absence of an orphan mitochondrial protein, c19orf12, causes a distinct clinical subtype of neurodegeneration with brain iron accumulation. Am J Hum Genet. 2011;89:543–50.PubMedPubMedCentralCrossRef Hartig MB, Iuso A, Haack T, et al. Absence of an orphan mitochondrial protein, c19orf12, causes a distinct clinical subtype of neurodegeneration with brain iron accumulation. Am J Hum Genet. 2011;89:543–50.PubMedPubMedCentralCrossRef
33.
go back to reference Hartig M, Prokisch H, Meitinger T, Klopstock T. Mitochondrial membrane protein-associated neurodegeneration (MPAN). Int Rev Neurobiol. 2013;110:73–84.PubMedCrossRef Hartig M, Prokisch H, Meitinger T, Klopstock T. Mitochondrial membrane protein-associated neurodegeneration (MPAN). Int Rev Neurobiol. 2013;110:73–84.PubMedCrossRef
34.
go back to reference Gagliardi M, Annesi G, Lesca G, et al. C19orf12 gene mutations in patients with neurodegeneration with brain iron accumulation. Parkinsonism Related Disorders 2015. Gagliardi M, Annesi G, Lesca G, et al. C19orf12 gene mutations in patients with neurodegeneration with brain iron accumulation. Parkinsonism Related Disorders 2015.
35.
go back to reference Kruer MC, Paisan-Ruiz C, Boddaert N, et al. Defective FA2H leads to a novel form of neurodegeneration with brain iron accumulation (NBIA). Ann Neurol. 2010;68:611–8.PubMedCrossRef Kruer MC, Paisan-Ruiz C, Boddaert N, et al. Defective FA2H leads to a novel form of neurodegeneration with brain iron accumulation (NBIA). Ann Neurol. 2010;68:611–8.PubMedCrossRef
36.
go back to reference Pensato V, Castellotti B, Gellera C, et al. Overlapping phenotypes in complex spastic paraplegias SPG11, SPG15, SPG35 and SPG48. Brain: J Neurology. 2014;137:1907–20.CrossRef Pensato V, Castellotti B, Gellera C, et al. Overlapping phenotypes in complex spastic paraplegias SPG11, SPG15, SPG35 and SPG48. Brain: J Neurology. 2014;137:1907–20.CrossRef
37.
go back to reference Cao L, Huang XJ, Chen CJ, Chen SD. A rare family with hereditary spastic paraplegia type 35 due to novel FA2H mutations: a case report with literature review. J Neurol Sci. 2013;329:1–5.PubMedCrossRef Cao L, Huang XJ, Chen CJ, Chen SD. A rare family with hereditary spastic paraplegia type 35 due to novel FA2H mutations: a case report with literature review. J Neurol Sci. 2013;329:1–5.PubMedCrossRef
38.
go back to reference Rupps R, Hukin J, Balicki M, Mercimek-Mahmutoglu S, Rolfs A, Dias C. Novel mutations in FA2H-Associated neurodegeneration: an underrecognized condition? J Child Neurol. 2013;28:1500–4.PubMedCrossRef Rupps R, Hukin J, Balicki M, Mercimek-Mahmutoglu S, Rolfs A, Dias C. Novel mutations in FA2H-Associated neurodegeneration: an underrecognized condition? J Child Neurol. 2013;28:1500–4.PubMedCrossRef
39.
go back to reference Dick KJ, Eckhardt M, Paisan-Ruiz C, et al. Mutation of FA2H underlies a complicated form of hereditary spastic paraplegia (SPG35). Hum Mutat. 2010;31:E1251–1260.PubMedCrossRef Dick KJ, Eckhardt M, Paisan-Ruiz C, et al. Mutation of FA2H underlies a complicated form of hereditary spastic paraplegia (SPG35). Hum Mutat. 2010;31:E1251–1260.PubMedCrossRef
40.
go back to reference Edvardson S, Hama H, Shaag A, et al. Mutations in the fatty acid 2-hydroxylase gene are associated with leukodystrophy with spastic paraparesis and dystonia. Am J Hum Genet. 2008;83:643–8.PubMedPubMedCentralCrossRef Edvardson S, Hama H, Shaag A, et al. Mutations in the fatty acid 2-hydroxylase gene are associated with leukodystrophy with spastic paraparesis and dystonia. Am J Hum Genet. 2008;83:643–8.PubMedPubMedCentralCrossRef
41.
go back to reference Haack TB, Hogarth P, Gregory A, Prokisch H, Hayflick SJ. BPAN: the only X-linked dominant NBIA disorder. Int Rev Neurobiol. 2013;110:85–90.PubMedCrossRef Haack TB, Hogarth P, Gregory A, Prokisch H, Hayflick SJ. BPAN: the only X-linked dominant NBIA disorder. Int Rev Neurobiol. 2013;110:85–90.PubMedCrossRef
42.
go back to reference Verhoeven WM, Egger JI, Koolen DA, et al. Beta-propeller protein-associated neurodegeneration (BPAN), a rare form of NBIA: Novel mutations and neuropsychiatric phenotype in three adult patients. Parkinsonism Related Disorders 2013. Verhoeven WM, Egger JI, Koolen DA, et al. Beta-propeller protein-associated neurodegeneration (BPAN), a rare form of NBIA: Novel mutations and neuropsychiatric phenotype in three adult patients. Parkinsonism Related Disorders 2013.
43.
go back to reference Hayflick SJ, Kruer MC, Gregory A, et al. Beta-propeller protein-associated neurodegeneration: a new X-linked dominant disorder with brain iron accumulation. Brain: J Neurology. 2013;136:1708–17.CrossRef Hayflick SJ, Kruer MC, Gregory A, et al. Beta-propeller protein-associated neurodegeneration: a new X-linked dominant disorder with brain iron accumulation. Brain: J Neurology. 2013;136:1708–17.CrossRef
44.
go back to reference Saitsu H, Nishimura T, Muramatsu K, et al. De novo mutations in the autophagy gene WDR45 cause static encephalopathy of childhood with neurodegeneration in adulthood. Nat Genet. 2013;45:445–9. 449e441.PubMedCrossRef Saitsu H, Nishimura T, Muramatsu K, et al. De novo mutations in the autophagy gene WDR45 cause static encephalopathy of childhood with neurodegeneration in adulthood. Nat Genet. 2013;45:445–9. 449e441.PubMedCrossRef
45.•
go back to reference Haack TB, Hogarth P, Kruer MC, et al. Exome sequencing reveals de novo WDR45 mutations causing a phenotypically distinct, X-linked dominant form of NBIA. Am J Hum Genet. 2012;91:1144–9. The authors (as well as Saitsu and colleagues, Ref 44 identified the underlying cause of SENDA syndrome, i.e. static encephalopathy of childhood with neurodegeneration in adulthood. In contrast to the other NBIA syndromes, de novo mutations are the mechanism. Surprisingly, three male patients had identical clinical and radiologic features as females, suggesting somatic mosaicism. Haack TB, Hogarth P, Kruer MC, et al. Exome sequencing reveals de novo WDR45 mutations causing a phenotypically distinct, X-linked dominant form of NBIA. Am J Hum Genet. 2012;91:1144–9. The authors (as well as Saitsu and colleagues, Ref 44 identified the underlying cause of SENDA syndrome, i.e. static encephalopathy of childhood with neurodegeneration in adulthood. In contrast to the other NBIA syndromes, de novo mutations are the mechanism. Surprisingly, three male patients had identical clinical and radiologic features as females, suggesting somatic mosaicism.
46.
go back to reference Zhao YG, Sun L, Miao G, et al. The autophagy gene Wdr45/Wipi4 regulates learning and memory function and axonal homeostasis. Autophagy 2015:0. Zhao YG, Sun L, Miao G, et al. The autophagy gene Wdr45/Wipi4 regulates learning and memory function and axonal homeostasis. Autophagy 2015:0.
47.•
go back to reference Nishioka K, Oyama G, Yoshino H, et al. High frequency of beta-propeller protein-associated neurodegeneration (BPAN) among patients with intellectual disability and young-onset parkinsonism. Neurobiology of aging 2015. This genetic screening study of patients with childhood intellectual disability and young-onset parkinsonism (≤40 years at onset) identified WDR45 mutations (associated with BPAN) in one quarter of patients. The data suggest a high frequency of beta-propeller protein-associated neurodegeneration mutations among this group of patients, at least in the Japanese population. Nishioka K, Oyama G, Yoshino H, et al. High frequency of beta-propeller protein-associated neurodegeneration (BPAN) among patients with intellectual disability and young-onset parkinsonism. Neurobiology of aging 2015. This genetic screening study of patients with childhood intellectual disability and young-onset parkinsonism (≤40 years at onset) identified WDR45 mutations (associated with BPAN) in one quarter of patients. The data suggest a high frequency of beta-propeller protein-associated neurodegeneration mutations among this group of patients, at least in the Japanese population.
48.
go back to reference Schneider SA, Paisan-Ruiz C, Quinn NP, et al. ATP13A2 mutations (PARK9) cause neurodegeneration with brain iron accumulation. Movement Disorders: Off J Movement Disorder Soc. 2010;25:979–84.CrossRef Schneider SA, Paisan-Ruiz C, Quinn NP, et al. ATP13A2 mutations (PARK9) cause neurodegeneration with brain iron accumulation. Movement Disorders: Off J Movement Disorder Soc. 2010;25:979–84.CrossRef
49.
go back to reference Bruggemann N, Hagenah J, Reetz K, et al. Recessively inherited parkinsonism: effect of ATP13A2 mutations on the clinical and neuroimaging phenotype. Arch Neurol. 2010;67:1357–63.PubMedCrossRef Bruggemann N, Hagenah J, Reetz K, et al. Recessively inherited parkinsonism: effect of ATP13A2 mutations on the clinical and neuroimaging phenotype. Arch Neurol. 2010;67:1357–63.PubMedCrossRef
50.
go back to reference Behrens MI, Bruggemann N, Chana P, et al. Clinical spectrum of Kufor-Rakeb syndrome in the Chilean kindred with ATP13A2 mutations. Movement Disorders: Off J Movement Disorder Soc. 2010;25:1929–37.CrossRef Behrens MI, Bruggemann N, Chana P, et al. Clinical spectrum of Kufor-Rakeb syndrome in the Chilean kindred with ATP13A2 mutations. Movement Disorders: Off J Movement Disorder Soc. 2010;25:1929–37.CrossRef
51.
go back to reference Williams DR, Hadeed A, Al-Din AS, Wreikat AL, Lees AJ. Kufor Rakeb disease: autosomal recessive, levodopa-responsive parkinsonism with pyramidal degeneration, supranuclear gaze palsy, and dementia. Movement Disorders: Off J Movement Disorder Soc. 2005;20:1264–71.CrossRef Williams DR, Hadeed A, Al-Din AS, Wreikat AL, Lees AJ. Kufor Rakeb disease: autosomal recessive, levodopa-responsive parkinsonism with pyramidal degeneration, supranuclear gaze palsy, and dementia. Movement Disorders: Off J Movement Disorder Soc. 2005;20:1264–71.CrossRef
52.
go back to reference Bras J, Verloes A, Schneider SA, Mole SE, Guerreiro RJ. Mutation of the parkinsonism gene ATP13A2 causes neuronal ceroid-lipofuscinosis. Hum Mol Genet. 2012;21:2646–50.PubMedPubMedCentralCrossRef Bras J, Verloes A, Schneider SA, Mole SE, Guerreiro RJ. Mutation of the parkinsonism gene ATP13A2 causes neuronal ceroid-lipofuscinosis. Hum Mol Genet. 2012;21:2646–50.PubMedPubMedCentralCrossRef
53.
go back to reference Tsunemi T, Krainc D. Zn(2)(+) dyshomeostasis caused by loss of ATP13A2/PARK9 leads to lysosomal dysfunction and alpha-synuclein accumulation. Hum Mol Genet. 2014;23:2791–801.PubMedPubMedCentralCrossRef Tsunemi T, Krainc D. Zn(2)(+) dyshomeostasis caused by loss of ATP13A2/PARK9 leads to lysosomal dysfunction and alpha-synuclein accumulation. Hum Mol Genet. 2014;23:2791–801.PubMedPubMedCentralCrossRef
54.
55.
go back to reference Podhajska A, Musso A, Trancikova A, et al. Common pathogenic effects of missense mutations in the P-type ATPase ATP13A2 (PARK9) associated with early-onset Parkinsonism. PLoS One. 2012;7, e39942.PubMedPubMedCentralCrossRef Podhajska A, Musso A, Trancikova A, et al. Common pathogenic effects of missense mutations in the P-type ATPase ATP13A2 (PARK9) associated with early-onset Parkinsonism. PLoS One. 2012;7, e39942.PubMedPubMedCentralCrossRef
56.
go back to reference Rinaldi DE, Corradi GR, Cuesta LM, Adamo HP, de Tezanos Pinto F. The Parkinson-associated human P5B-ATPase ATP13A2 protects against the iron-induced cytotoxicity. Biochim Biophys Acta. 1850;2015:1646–55. Rinaldi DE, Corradi GR, Cuesta LM, Adamo HP, de Tezanos Pinto F. The Parkinson-associated human P5B-ATPase ATP13A2 protects against the iron-induced cytotoxicity. Biochim Biophys Acta. 1850;2015:1646–55.
57.
go back to reference Chinnery PF, Crompton DE, Birchall D, et al. Clinical features and natural history of neuroferritinopathy caused by the FTL1 460InsA mutation. Brain: J Neurology. 2007;130:110–9.CrossRef Chinnery PF, Crompton DE, Birchall D, et al. Clinical features and natural history of neuroferritinopathy caused by the FTL1 460InsA mutation. Brain: J Neurology. 2007;130:110–9.CrossRef
59.
go back to reference McNeill A, Pandolfo M, Kuhn J, Shang H, Miyajima H. The neurological presentation of ceruloplasmin gene mutations. Eur Neurol. 2008;60:200–5.PubMedCrossRef McNeill A, Pandolfo M, Kuhn J, Shang H, Miyajima H. The neurological presentation of ceruloplasmin gene mutations. Eur Neurol. 2008;60:200–5.PubMedCrossRef
60.
go back to reference Vroegindeweij LH, van der Beek EH, Boon AJ, et al. Aceruloplasminemia presents as type 1 diabetes in non-obese adults: a detailed case series. Diabet Med 2015. Vroegindeweij LH, van der Beek EH, Boon AJ, et al. Aceruloplasminemia presents as type 1 diabetes in non-obese adults: a detailed case series. Diabet Med 2015.
61.
go back to reference Nishida K, Garringer HJ, Futamura N, et al. A novel ferritin light chain mutation in neuroferritinopathy with an atypical presentation. J Neurol Sci. 2014;342:173–7.PubMedPubMedCentralCrossRef Nishida K, Garringer HJ, Futamura N, et al. A novel ferritin light chain mutation in neuroferritinopathy with an atypical presentation. J Neurol Sci. 2014;342:173–7.PubMedPubMedCentralCrossRef
62.
go back to reference Amaral LL, Gaddikeri S, Chapman PR, et al. Neurodegeneration with brain Iron accumulation: clinicoradiological approach to diagnosis. J Neuroimaging: Off J Am Soc Neuroimaging. 2015;25:539–51.CrossRef Amaral LL, Gaddikeri S, Chapman PR, et al. Neurodegeneration with brain Iron accumulation: clinicoradiological approach to diagnosis. J Neuroimaging: Off J Am Soc Neuroimaging. 2015;25:539–51.CrossRef
63.
go back to reference Dezortova M, Herynek V, Krssak M, Kronerwetter C, Trattnig S, Hajek M. Two forms of iron as an intrinsic contrast agent in the basal ganglia of PKAN patients. Contrast Media Mol Imaging. 2012;7:509–15.PubMedCrossRef Dezortova M, Herynek V, Krssak M, Kronerwetter C, Trattnig S, Hajek M. Two forms of iron as an intrinsic contrast agent in the basal ganglia of PKAN patients. Contrast Media Mol Imaging. 2012;7:509–15.PubMedCrossRef
64.•
go back to reference Dusek PTM EM, Madai VI, Jech R, Sobesky J, Paul F, Niendorf T, et al. 7 Tesla MRI for brain iron quantification in homozygous and heterozygous PANK2 mutation carriers. Mov Disord Clin Pract. 2014;1:329–35. This study for the first time employed ultrahigh-field MRI in PKAN, both patients and relatives. Quantification of iron concentrations using quantitative susceptibility mapping revealed more than three times higher concentrations of iron in the globus pallidus, substantia nigra and internal capsule in PKAN patients, whereas 11 heterozygous PANK2 gene mutation carriers had normal brain iron concentrations. This suggests that single gene mutations are not sufficient to cause excessive iron accumulation.CrossRef Dusek PTM EM, Madai VI, Jech R, Sobesky J, Paul F, Niendorf T, et al. 7 Tesla MRI for brain iron quantification in homozygous and heterozygous PANK2 mutation carriers. Mov Disord Clin Pract. 2014;1:329–35. This study for the first time employed ultrahigh-field MRI in PKAN, both patients and relatives. Quantification of iron concentrations using quantitative susceptibility mapping revealed more than three times higher concentrations of iron in the globus pallidus, substantia nigra and internal capsule in PKAN patients, whereas 11 heterozygous PANK2 gene mutation carriers had normal brain iron concentrations. This suggests that single gene mutations are not sufficient to cause excessive iron accumulation.CrossRef
65.
go back to reference Hayflick SJ, Hartman M, Coryell J, Gitschier J, Rowley H. Brain MRI in neurodegeneration with brain iron accumulation with and without PANK2 mutations. AJNR Am J Neuroradiol. 2006;27:1230–3.PubMedPubMedCentral Hayflick SJ, Hartman M, Coryell J, Gitschier J, Rowley H. Brain MRI in neurodegeneration with brain iron accumulation with and without PANK2 mutations. AJNR Am J Neuroradiol. 2006;27:1230–3.PubMedPubMedCentral
66.•
go back to reference Delgado RF, Sanchez PR, Speckter H, et al. Missense PANK2 mutation without “eye of the tiger” sign: MR findings in a large group of patients with pantothenate kinase-associated neurodegeneration (PKAN). J Magnetic Resonance Imaging: JMRI. 2012;35:788–94. Delgado et al. examined twenty PKAN patients, one pre-symptomatic case and 13 heterozygous gene mutation carriers using 3 Tesla MRI. While all patients had the typical signal reduction within the globus pallidus and the substantia nigra, a surprising finding was the absence of the bright spot (“eye of the tiger”) in the medial part of the pallidum in six symptomatic patients. This strengthens the notion that radiological features are dynamic.PubMedCrossRef Delgado RF, Sanchez PR, Speckter H, et al. Missense PANK2 mutation without “eye of the tiger” sign: MR findings in a large group of patients with pantothenate kinase-associated neurodegeneration (PKAN). J Magnetic Resonance Imaging: JMRI. 2012;35:788–94. Delgado et al. examined twenty PKAN patients, one pre-symptomatic case and 13 heterozygous gene mutation carriers using 3 Tesla MRI. While all patients had the typical signal reduction within the globus pallidus and the substantia nigra, a surprising finding was the absence of the bright spot (“eye of the tiger”) in the medial part of the pallidum in six symptomatic patients. This strengthens the notion that radiological features are dynamic.PubMedCrossRef
67.
go back to reference Hayflick SJ, Penzien JM, Michl W, Sharif UM, Rosman NP, Wheeler PG. Cranial MRI changes may precede symptoms in Hallervorden-Spatz syndrome. Pediatr Neurol. 2001;25:166–9.PubMedCrossRef Hayflick SJ, Penzien JM, Michl W, Sharif UM, Rosman NP, Wheeler PG. Cranial MRI changes may precede symptoms in Hallervorden-Spatz syndrome. Pediatr Neurol. 2001;25:166–9.PubMedCrossRef
68.
go back to reference Chiapparini L, Savoiardo M, D’Arrigo S, et al. The “eye-of-the-tiger” sign may be absent in the early stages of classic pantothenate kinase associated neurodegeneration. Neuropediatrics. 2011;42:159–62.PubMedCrossRef Chiapparini L, Savoiardo M, D’Arrigo S, et al. The “eye-of-the-tiger” sign may be absent in the early stages of classic pantothenate kinase associated neurodegeneration. Neuropediatrics. 2011;42:159–62.PubMedCrossRef
69.
go back to reference Grandas F, Fernandez-Carballal C, Guzman-de-Villoria J, Ampuero I. Treatment of a dystonic storm with pallidal stimulation in a patient with PANK2 mutation. Movement Disorders: Off J Movement Disorder Soc. 2011;26:921–2.CrossRef Grandas F, Fernandez-Carballal C, Guzman-de-Villoria J, Ampuero I. Treatment of a dystonic storm with pallidal stimulation in a patient with PANK2 mutation. Movement Disorders: Off J Movement Disorder Soc. 2011;26:921–2.CrossRef
70.
go back to reference Fermin-Delgado R, Roa-Sanchez P, Speckter H, et al. Involvement of globus pallidus and midbrain nuclei in pantothenate kinase-associated neurodegeneration: measurement of T2 and T2* time. Clin Neuroradiol. 2013;23:11–5.PubMedCrossRef Fermin-Delgado R, Roa-Sanchez P, Speckter H, et al. Involvement of globus pallidus and midbrain nuclei in pantothenate kinase-associated neurodegeneration: measurement of T2 and T2* time. Clin Neuroradiol. 2013;23:11–5.PubMedCrossRef
71.
go back to reference Rodriguez-Raecke R, Roa-Sanchez P, Speckter H, et al. Grey matter alterations in patients with pantothenate kinase-associated neurodegeneration (PKAN). Parkinsonism Relat Disord. 2014;20:975–9.PubMedCrossRef Rodriguez-Raecke R, Roa-Sanchez P, Speckter H, et al. Grey matter alterations in patients with pantothenate kinase-associated neurodegeneration (PKAN). Parkinsonism Relat Disord. 2014;20:975–9.PubMedCrossRef
72.
go back to reference Stoeter P, Roa-Sanchez P, Speckter H, et al. Changes of cerebral white matter in patients suffering from pantothenate kinase-associated neurodegeneration (PKAN): a diffusion tensor imaging (DTI) study. Parkinsonism Related Disorders 2015. Stoeter P, Roa-Sanchez P, Speckter H, et al. Changes of cerebral white matter in patients suffering from pantothenate kinase-associated neurodegeneration (PKAN): a diffusion tensor imaging (DTI) study. Parkinsonism Related Disorders 2015.
73.
go back to reference Wu YW, Hess CP, Singhal NS, Groden C, Toro C. Idiopathic basal ganglia calcifications: an atypical presentation of PKAN. Pediatr Neurol. 2013;49:351–4.PubMedCrossRef Wu YW, Hess CP, Singhal NS, Groden C, Toro C. Idiopathic basal ganglia calcifications: an atypical presentation of PKAN. Pediatr Neurol. 2013;49:351–4.PubMedCrossRef
74.
go back to reference Kurian MA, Morgan NV, MacPherson L, et al. Phenotypic spectrum of neurodegeneration associated with mutations in the PLA2G6 gene (PLAN). Neurology. 2008;70:1623–9.PubMedCrossRef Kurian MA, Morgan NV, MacPherson L, et al. Phenotypic spectrum of neurodegeneration associated with mutations in the PLA2G6 gene (PLAN). Neurology. 2008;70:1623–9.PubMedCrossRef
75.
go back to reference Paisan-Ruiz C, Li A, Schneider SA, et al. Widespread Lewy body and tau accumulation in childhood and adult onset dystonia-parkinsonism cases with PLA2G6 mutations. Neurobiol Aging. 2012;33:814–23.PubMedPubMedCentralCrossRef Paisan-Ruiz C, Li A, Schneider SA, et al. Widespread Lewy body and tau accumulation in childhood and adult onset dystonia-parkinsonism cases with PLA2G6 mutations. Neurobiol Aging. 2012;33:814–23.PubMedPubMedCentralCrossRef
76.
go back to reference Ohta E, Takiyama Y. MRI findings in neuroferritinopathy. Neurology Res Int. 2012;2012:197438.CrossRef Ohta E, Takiyama Y. MRI findings in neuroferritinopathy. Neurology Res Int. 2012;2012:197438.CrossRef
77.
go back to reference Batla A, Adams ME, Erro R, et al. Cortical pencil lining in neuroferritinopathy: a diagnostic clue. Neurology 2015. Batla A, Adams ME, Erro R, et al. Cortical pencil lining in neuroferritinopathy: a diagnostic clue. Neurology 2015.
78.
go back to reference McNeill A, Birchall D, Hayflick SJ, et al. T2* and FSE MRI distinguishes four subtypes of neurodegeneration with brain iron accumulation. Neurology. 2008;70:1614–9.PubMedPubMedCentralCrossRef McNeill A, Birchall D, Hayflick SJ, et al. T2* and FSE MRI distinguishes four subtypes of neurodegeneration with brain iron accumulation. Neurology. 2008;70:1614–9.PubMedPubMedCentralCrossRef
79.
go back to reference Schneider SA, Bhatia KP, Hardy J. Complicated recessive dystonia Parkinsonism syndromes. Movement Disorders: Off J Movement Disorder Soc. 2009;24:490–9.CrossRef Schneider SA, Bhatia KP, Hardy J. Complicated recessive dystonia Parkinsonism syndromes. Movement Disorders: Off J Movement Disorder Soc. 2009;24:490–9.CrossRef
80.
go back to reference Chang MH, Hung WL, Liao YC, Lee YC, Hsieh PF. Eye of the tiger-like MRI in Parkinsonian variant of multiple system atrophy. J Neural Transm. 2009;116:861–6.PubMedCrossRef Chang MH, Hung WL, Liao YC, Lee YC, Hsieh PF. Eye of the tiger-like MRI in Parkinsonian variant of multiple system atrophy. J Neural Transm. 2009;116:861–6.PubMedCrossRef
81.
go back to reference Davie CA, Barker GJ, Machado C, Miller DH, Lees AJ. Proton magnetic resonance spectroscopy in Steele-Richardson-Olszewski syndrome. Movement Disorders: Off J Movement Disorder Soc. 1997;12:767–71.CrossRef Davie CA, Barker GJ, Machado C, Miller DH, Lees AJ. Proton magnetic resonance spectroscopy in Steele-Richardson-Olszewski syndrome. Movement Disorders: Off J Movement Disorder Soc. 1997;12:767–71.CrossRef
82.
go back to reference Molinuevo JL, Munoz E, Valldeoriola F, Tolosa E. The eye of the tiger sign in cortical-basal ganglionic degeneration. Movement Disorders: Off J Movement Disorder Soc. 1999;14:169–71.CrossRef Molinuevo JL, Munoz E, Valldeoriola F, Tolosa E. The eye of the tiger sign in cortical-basal ganglionic degeneration. Movement Disorders: Off J Movement Disorder Soc. 1999;14:169–71.CrossRef
83.
go back to reference Santillo AF, Skoglund L, Lindau M, et al. Frontotemporal dementia-amyotrophic lateral sclerosis complex is simulated by neurodegeneration with brain iron accumulation. Alzheimer Dis Assoc Disord. 2009;23:298–300.PubMedCrossRef Santillo AF, Skoglund L, Lindau M, et al. Frontotemporal dementia-amyotrophic lateral sclerosis complex is simulated by neurodegeneration with brain iron accumulation. Alzheimer Dis Assoc Disord. 2009;23:298–300.PubMedCrossRef
84.
go back to reference Zhang J, Zhang Y, Wang J, et al. Characterizing iron deposition in Parkinson’s disease using susceptibility-weighted imaging: an in vivo MR study. Brain Res. 2010;1330:124–30.PubMedCrossRef Zhang J, Zhang Y, Wang J, et al. Characterizing iron deposition in Parkinson’s disease using susceptibility-weighted imaging: an in vivo MR study. Brain Res. 2010;1330:124–30.PubMedCrossRef
85.
go back to reference Rossi M, Ruottinen H, Elovaara I, et al. Brain iron deposition and sequence characteristics in Parkinsonism: comparison of SWI, T(2)* maps, T(2)-weighted-, and FLAIR-SPACE. Invest Radiol. 2010;45:795–802.PubMedCrossRef Rossi M, Ruottinen H, Elovaara I, et al. Brain iron deposition and sequence characteristics in Parkinsonism: comparison of SWI, T(2)* maps, T(2)-weighted-, and FLAIR-SPACE. Invest Radiol. 2010;45:795–802.PubMedCrossRef
86.
go back to reference 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
87.
go back to reference Tsukamoto H, Inui K, Taniike M, et al. A case of Hallervorden-Spatz disease: progressive and intractable dystonia controlled by bilateral thalamotomy. Brain Dev. 1992;14:269–72.PubMedCrossRef Tsukamoto H, Inui K, Taniike M, et al. A case of Hallervorden-Spatz disease: progressive and intractable dystonia controlled by bilateral thalamotomy. Brain Dev. 1992;14:269–72.PubMedCrossRef
88.
go back to reference Balas I, Kovacs N, Hollody K. Staged bilateral stereotactic pallidothalamotomy for life-threatening dystonia in a child with Hallervorden-Spatz disease. Movement Disorders: Off J Movement Disorder Soc. 2006;21:82–5.CrossRef Balas I, Kovacs N, Hollody K. Staged bilateral stereotactic pallidothalamotomy for life-threatening dystonia in a child with Hallervorden-Spatz disease. Movement Disorders: Off J Movement Disorder Soc. 2006;21:82–5.CrossRef
89.
go back to reference Kyriagis M, Grattan-Smith P, Scheinberg A, Teo C, Nakaji N, Waugh M. Status dystonicus and Hallervorden-Spatz disease: treatment with intrathecal baclofen and pallidotomy. J Paediatr Child Health. 2004;40:322–5.PubMedCrossRef Kyriagis M, Grattan-Smith P, Scheinberg A, Teo C, Nakaji N, Waugh M. Status dystonicus and Hallervorden-Spatz disease: treatment with intrathecal baclofen and pallidotomy. J Paediatr Child Health. 2004;40:322–5.PubMedCrossRef
90.
go back to reference Justesen CR, Penn RD, Kroin JS, Egel RT. Stereotactic pallidotomy in a child with Hallervorden-Spatz disease. Case report. J Neurosurg. 1999;90:551–4.PubMedCrossRef Justesen CR, Penn RD, Kroin JS, Egel RT. Stereotactic pallidotomy in a child with Hallervorden-Spatz disease. Case report. J Neurosurg. 1999;90:551–4.PubMedCrossRef
91.
go back to reference Castelnau P, Cif L, Valente EM, et al. Pallidal stimulation improves pantothenate kinase-associated neurodegeneration. Ann Neurol. 2005;57:738–41.PubMedCrossRef Castelnau P, Cif L, Valente EM, et al. Pallidal stimulation improves pantothenate kinase-associated neurodegeneration. Ann Neurol. 2005;57:738–41.PubMedCrossRef
92.
go back to reference Mikati MA, Yehya A, Darwish H, Karam P, Comair Y. Deep brain stimulation as a mode of treatment of early onset pantothenate kinase-associated neurodegeneration. Eur J Paediatr Neurol. 2009;13:61–4.PubMedCrossRef Mikati MA, Yehya A, Darwish H, Karam P, Comair Y. Deep brain stimulation as a mode of treatment of early onset pantothenate kinase-associated neurodegeneration. Eur J Paediatr Neurol. 2009;13:61–4.PubMedCrossRef
93.
go back to reference Krause M, Fogel W, Tronnier V, et al. Long-term benefit to pallidal deep brain stimulation in a case of dystonia secondary to pantothenate kinase-associated neurodegeneration. Movement Disorders: Off J Movement Disorder Soc. 2006;21:2255–7.CrossRef Krause M, Fogel W, Tronnier V, et al. Long-term benefit to pallidal deep brain stimulation in a case of dystonia secondary to pantothenate kinase-associated neurodegeneration. Movement Disorders: Off J Movement Disorder Soc. 2006;21:2255–7.CrossRef
94.
go back to reference Schneider SA, Dusek P, Hardy J, Westenberger A, Jankovic J, Bhatia KP. Genetics and pathophysiology of neurodegeneration with brain iron accumulation (NBIA). Curr Neuropharmacol. 2013;11:59–79.PubMedPubMedCentral Schneider SA, Dusek P, Hardy J, Westenberger A, Jankovic J, Bhatia KP. Genetics and pathophysiology of neurodegeneration with brain iron accumulation (NBIA). Curr Neuropharmacol. 2013;11:59–79.PubMedPubMedCentral
95.
go back to reference Li X, Jankovic J, Le W. Iron chelation and neuroprotection in neurodegenerative diseases. J Neural Transm. 2011;118:473–7.PubMedCrossRef Li X, Jankovic J, Le W. Iron chelation and neuroprotection in neurodegenerative diseases. J Neural Transm. 2011;118:473–7.PubMedCrossRef
96.
go back to reference Zhu W, Li X, Xie W, et al. Genetic iron chelation protects against proteasome inhibition-induced dopamine neuron degeneration. Neurobiol Dis. 2010;37:307–13.PubMedCrossRef Zhu W, Li X, Xie W, et al. Genetic iron chelation protects against proteasome inhibition-induced dopamine neuron degeneration. Neurobiol Dis. 2010;37:307–13.PubMedCrossRef
97.•
go back to reference Zorzi G, Zibordi F, Chiapparini L, et al. Iron-related MRI images in patients with pantothenate kinase-associated neurodegeneration (PKAN) treated with deferiprone: results of a phase II pilot trial. Movement Disorders: Off J Movement Disorder Soc. 2011;26:1756–9. Zorzi et al. assessed the safety and efficacy of the oral iron-chelating agent deferiprone on clinical features and magnetic resonance pallidal iron concentration in PKAN. While there was significant median reduction in globus pallidus iron content, there was no clinical benefit. This may suggest that iron wash-out may be incapable of rescuing damaged tissue. Larger studies are underway to further explore this issue.CrossRef Zorzi G, Zibordi F, Chiapparini L, et al. Iron-related MRI images in patients with pantothenate kinase-associated neurodegeneration (PKAN) treated with deferiprone: results of a phase II pilot trial. Movement Disorders: Off J Movement Disorder Soc. 2011;26:1756–9. Zorzi et al. assessed the safety and efficacy of the oral iron-chelating agent deferiprone on clinical features and magnetic resonance pallidal iron concentration in PKAN. While there was significant median reduction in globus pallidus iron content, there was no clinical benefit. This may suggest that iron wash-out may be incapable of rescuing damaged tissue. Larger studies are underway to further explore this issue.CrossRef
98.
go back to reference Kalman B, Lautenschlaeger R, Kohlmayer F, et al. An international registry for neurodegeneration with brain iron accumulation. Orphanet J Rare Dis. 2012;7:66.PubMedPubMedCentralCrossRef Kalman B, Lautenschlaeger R, Kohlmayer F, et al. An international registry for neurodegeneration with brain iron accumulation. Orphanet J Rare Dis. 2012;7:66.PubMedPubMedCentralCrossRef
99.
go back to reference Srinivasan B, Baratashvili M, van der Zwaag M, Kanon B, Colombelli C, Lambrechts RA, Schaap O, Nollen EA, Podgoršek A, Kosec G, Petković H, Hayflick S, Tiranti V, Reijngoud DJ, Grzeschik NA, Sibon OC. Extracellular 4'-phosphopantetheine is a source for intracellular coenzyme A synthesis. Nat Chem Biol. 2015 Oct;11(10):784–92. Srinivasan B, Baratashvili M, van der Zwaag M, Kanon B, Colombelli C, Lambrechts RA, Schaap O, Nollen EA, Podgoršek A, Kosec G, Petković H, Hayflick S, Tiranti V, Reijngoud DJ, Grzeschik NA, Sibon OC. Extracellular 4'-phosphopantetheine is a source for intracellular coenzyme A synthesis. Nat Chem Biol. 2015 Oct;11(10):784–92.
100.
go back to reference Schneider SA, Zorzi G, Nardocci N. Pathophysiology and treatment of neurodegeneration with brain iron accumulation in the pediatric population. Curr Treat Options Neurol. 2013;15:652–67.PubMedCrossRef Schneider SA, Zorzi G, Nardocci N. Pathophysiology and treatment of neurodegeneration with brain iron accumulation in the pediatric population. Curr Treat Options Neurol. 2013;15:652–67.PubMedCrossRef
Metadata
Title
Neurodegeneration with Brain Iron Accumulation
Author
Susanne A. Schneider
Publication date
01-01-2016
Publisher
Springer US
Published in
Current Neurology and Neuroscience Reports / Issue 1/2016
Print ISSN: 1528-4042
Electronic ISSN: 1534-6293
DOI
https://doi.org/10.1007/s11910-015-0608-3

Other articles of this Issue 1/2016

Current Neurology and Neuroscience Reports 1/2016 Go to the issue

Movement Disorders (S Fox, Section Editor)

Advances in Biomarker Research in Parkinson’s Disease

Infection (J Halperin, Section Editor)

Infection and Stroke: an Update on Recent Progress