Chapter 41 - Dentatorubral–pallidoluysian atrophy

https://doi.org/10.1016/B978-0-444-51892-7.00041-3Get rights and content

Abstract

Dentatorubral–pallidoluysian atrophy (DRPLA) is a rare autosomal dominant neurodegenerative disorder clinically characterized by various combinations of cerebellar ataxia, choreoathetosis, myoclonus, epilepsy, dementia, and psychiatric symptoms. The most striking clinical features of DRPLA are the considerable heterogeneity in clinical presentation, depending on the age of onset, and the prominent genetic anticipation. DRPLA is caused by unstable expansion of CAG repeats coding for polyglutamine stretches located in exon 5 of the DRPLA gene. DRPLA is characterized by prominent anticipation, with paternal transmission resulting in more prominent anticipation than does maternal transmission, which is now understood based on the intergenerational stability of the CAG repeats. DRPLA protein (also called atrophin-1) is localized in the nucleus and functions as a transcription co-regulator. Recent immunohistochemical studies on autopsied tissues of patients with DRPLA have demonstrated that diffuse accumulation of mutant DRPLA protein (atrophin-1) in the neuronal nuclei, rather than the formation of neuronal intranuclear inclusions (NIIs), is the predominant pathologic condition and involves a wide range of central nervous system regions far beyond the systems previously reported to be affected. Thus, age-dependent and CAG repeat-dependent intranuclear accumulation of mutant DRPLA leading to nuclear dysfunctions are suggested to be the essential pathophysiologic mechanisms in DRPLA.

Introduction

Dentatorubral–pallidoluysian atrophy (DRPLA) is a rare autosomal dominant neurodegenerative disorder clinically characterized by various combinations of cerebellar ataxia, choreoathetosis, myoclonus, epilepsy, dementia, and psychiatric symptoms (MIM #125370) (Naito and Oyanagi, 1982). The term DRPLA was originally used by Smith et al. to describe a neuropathological condition associated with severe neuronal loss, particularly in the dentatorubral and pallidoluysian systems of the central nervous system, in a sporadic case without a family history ( Smith et al., 1958, Smith, 1975). The hereditary form of DRPLA was first described in 1972 by Naito and his colleagues (Naito et al., 1972). Since then, a number of reports on Japanese pedigrees with similar clinical presentations have been published (Oyanagi and Naito, 1977, Tanaka et al., 1977, Hirayama et al., 1981, Iizuka et al., 1984, Suzuki et al., 1985, Iizuka and Hirayama, 1986, Akashi et al., 1987, Iwabuchi, 1987, Iwabuchi et al., 1987, Naito et al., 1987), and DRPLA has been established as a distinct disease entity.

The gene for DRPLA was discovered in 1994 and an unstable CAG trinucleotide repeat expansion in the protein-coding region of this gene was found to be the causative mutation for DRPLA (Koide et al., 1994, Nagafuchi et al., 1994a). To date, at least nine diseases have been found as being caused by expansion of CAG repeats coding for polyglutamine stretches; these include spinal and bulbar muscular atrophy (SBMA) (La Spada et al., 1991), Huntington's disease (HD) (Huntington's Disease Collaborative Research Group, 1993), spinocerebellar ataxia type 1 (SCA1) (Orr et al., 1993), DRPLA (Koide et al., 1994, Nagafuchi et al., 1994b), Machado–Joseph disease (also called SCA3) (Kawaguchi et al., 1994), SCA2 (Imbert et al., 1996, Pulst et al., 1996, Sanpei et al., 1996), SCA6 (Zhuchenko et al., 1997), SCA7 (David et al., 1997), and SCA17 (Nakamura et al., 2001).

In this chapter, the clinical and molecular genetic aspects of DRPLA are described. Recent progress in the study of the molecular mechanisms of neurodegeneration caused by expanded polyglutamine stretches is also discussed.

Section snippets

Epidemiology

The prevalence rate of SCAs in the Japanese population has been estimated to be 18.5/100 000 (Tsuji et al., 2008). Sporadic ataxias accounted for 67.2% of SCAs, and hereditary ataxias for 28.8%. Among the hereditary ataxias, autosomal dominant ataxias by far predominate in the Japanese population. Among the autosomal dominant ataxias, Machado–Joseph disease (SCA3) and SCA6 are the common SCAs, and DRPLA is the third most common (Fig. 41.1) (Tsuji et al., 2008). The relative frequencies of SCA1

Clinical presentations of DRPLA

The most striking clinical features of DRPLA are the considerable heterogeneity in clinical presentation depending on the age of onset and the prominent genetic anticipation. Naito and Oyanagi (1982) reported that juvenile-onset patients (onset before the age of 20) frequently exhibit a phenotype of progressive myoclonus epilepsy (PME), characterized by progressive ataxia, seizures, myoclonus, and intellectual deterioration. Epileptic seizures are a common feature in all patients with onset

Clinical genetics

The mode of inheritance of DRPLA is autosomal dominant with a high penetrance. Although DRPLA has been reported to occur predominantly in Japanese individuals, cases with similar clinical features have been described in other ethnic groups (Titica and van Bogaert, 1946, De Barsy et al., 1968, Farmer et al., 1989). Since the discovery of the gene for DRPLA (Koide et al., 1994, Nagafuchi et al., 1994b), CAG repeat expansion of the DRPLA gene has been demonstrated in families with various ethnic

Neuroimaging

MRI findings for DRPLA are characterized by progressive atrophy of the cerebellum and brainstem structures. Multiple regression analysis revealed that both the patient's age at MRI scan and the size of expanded CAG repeats correlate with the atrophic changes of the midsagittal structures (cerebellar vermis and brainstem), suggesting that both the age and the size of expanded CAG repeats independently affect the atrophic changes of the cerebellum and brainstem. It has been known that DRPLA

Neuropathology

The major neuropathological changes detected by conventional neuropathological observations consist of combined degeneration of the dentatorubral and pallidoluysian systems of the central nervous system (Naito and Oyanagi, 1982, Takahashi et al., 1988, Yamada et al., 2000). In addition, cerebral white matter damage has been described. The autopsy study of the white matter lesions showed diffuse myelin pallor, axonal preservation, and reactive astrogliosis in the cerebral white matter, with only

Molecular genetics of DRPLA

DRPLA is characterized by prominent anticipation (Koide et al., 1994, Nagafuchi et al., 1994a; Ikeuchi et al., 1995a, b, c; Naito, 1995, Ueno et al., 1995). Paternal transmission results in more prominent anticipation (26–29 years/generation) than does maternal transmission (14–15 years/generation). Strong parental bias on the degree of anticipation observed in HD (Huntington's Disease Collaborative Research Group, 1993) and SCA1 (Orr et al., 1993) suggested that unstable CAG repeat expansion

Treatment

There are no treatments available to prevent disease progression in DRPLA. The principle of therapy for DRPLA is, therefore, basically a symptomatic approach. Antiepileptic drugs should be given to patients with epileptic seizures. Haloperidol can be given when patients show choreoathetosis affecting the quality of daily life.

References (73)

  • J.R. Burke et al.

    The Haw River syndrome: dentatorubropallidoluysian atrophy (DRPLA) in an African-American family

    Nat Genet

    (1994)
  • M.Y. Chung et al.

    Evidence for a mechanism predisposing to intergenerational CAG repeat instability in spinocerebellar ataxia type I

    Nat Genet

    (1993)
  • M. Connarty et al.

    Molecular re-investigation of patients with Huntington's disease in Wessex reveals a family with dentatorubral and pallidoluysian atrophy

    Hum Genet

    (1996)
  • G. David et al.

    Cloning of the SCA7 gene reveals a highly unstable CAG repeat expansion

    Nat Genet

    (1997)
  • T.H. De Barsy et al.

    La Dyssynergie cerebelleuse myoclonique (R. Hunt): Affection autonome ou rariante du type degeneratif de l'epilepsie-myoclonie progressive (Unvericht-Lundborg) Appoche anatomo-alinique

    J Neurol Sci

    (1968)
  • A. Destee et al.

    The first identified French family with dentatorubral-pallidoluysian atrophy

    Mov Disord

    (2000)
  • M. Duyao et al.

    Trinucleotide repeat length instability and age of onset in Huntington's disease

    Nat Genet

    (1993)
  • T.W. Farmer et al.

    Ataxia, chorea, seizures, and dementia. Pathologic features of a newly defined familial disorder

    Arch Neurol

    (1989)
  • A. Filla et al.

    Relative frequencies of CAG expansions in spinocerebellar ataxia and dentatorubropallidoluysian atrophy in 116 Italian families

    Eur Neurol

    (2000)
  • Y. Hayashi et al.

    Hereditary dentatorubral-pallidoluysian atrophy – ubiquitinated filamentous inclusions in the cerebellar dentate nucleus neurons

    Acta Neuropathol (Berl)

    (1998)
  • K. Hirayama et al.

    Clinicopathological study of dentatorubropallidoluysian atrophy. Part I - Its clinical form and analysis of symptomatology [in Japanese]

    Adv Neurol

    (1981)
  • Huntington's Disease Collaborative Research Group

    A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington's disease chromosomes

    Cell

    (1993)
  • S. Igarashi et al.

    Suppression of aggregate formation and apoptosis by transglutaminase inhibitors in cells expressing truncated DRPLA protein with an expanded polyglutamine stretch

    Nat Genet

    (1998)
  • R. Iizuka et al.

    Dentato-rubro-pallido-luysian atrophy

  • R. Iizuka et al.

    Dentato-rubro-pallido-luysian atrophy: a clinico-pathological study

    J Neurol Neurosurg Psychiatry

    (1984)
  • T. Ikeuchi et al.

    Dentatorubral-pallidoluysian atrophy (DRPLA). Molecular basis for wide clinical features of DRPLA

    Clin Neurosci

    (1995)
  • T. Ikeuchi et al.

    Dentatorubral-pallidoluysian atrophy: clinical features are closely related to unstable expansions of trinucleotide (CAG) repeat

    Ann Neurol

    (1995)
  • G. Imbert et al.

    Cloning of the gene for spinocerebellar ataxia 2 reveals a locus with high sensitivity to expanded CAG/glutamine repeats [see comments]

    Nat Genet

    (1996)
  • K. Iwabuchi

    Clinico-pathological studies on dentato-rubro-pallido-luysian atrophy (DRPLA)

    Yokohama Igaku

    (1987)
  • K. Iwabuchi et al.

    A clinicopathological study on familial cases of dentatorubro-pallidoluysian atrophy (DRPLA)

    Clin Neurol

    (1987)
  • Y. Kawaguchi et al.

    CAG expansions in a novel gene for Machado-Joseph disease at chromosome 14q32.1

    Nat Genet

    (1994)
  • R. Koide et al.

    Unstable expansion of CAG repeat in hereditary dentatorubral-pallidoluysian atrophy (DRPLA)

    Nat Genet

    (1994)
  • R. Koide et al.

    Atrophy of the cerebellum and brainstem in dentatorubral pallidoluysian atrophy. Influence of CAG repeat size on MRI findings

    Neurology

    (1997)
  • A.R. La Spada et al.

    Androgen receptor gene mutations in X-linked spinal and bulbar muscular atrophy

    Nature

    (1991)
  • I. Le Ber et al.

    Prevalence of dentatorubral-pallidoluysian atrophy in a large series of white patients with cerebellar ataxia

    Arch Neurol

    (2003)
  • I.H. Lee et al.

    Dentatorubropallidoluysian atrophy in Chinese

    Arch Neurol

    (2001)
  • Cited by (63)

    • Parkinsonism and ataxia

      2022, Journal of the Neurological Sciences
      Citation Excerpt :

      However, a hypokinetic-rigid phenotype accompanying cerebellar signs can be present [11], typically in patients with lower CAG repeats, with or without CAA interruptions [12]. Like SCA17, Dentatorubral–pallidoluysian atrophy (DRPLA) is a rare neurodegenerative disease caused by CAG repeat expansion and is included in both SCAs and HDL syndromes due to its diverse motor phenotypes, where parkinsonian features are rarely encountered [13]. Although classically defined as a “pure” cerebellar ataxia, reports of SCA6-associated parkinsonian syndromes, mainly akinetic-rigid, are abundant [14,15], likely accompanied by dysautonomia, myoclonus, dystonia, and cognitive impairment.

    • DRPLA: An unusual disease or an underestimated cause of ataxia in Brazil?

      2021, Parkinsonism and Related Disorders
      Citation Excerpt :

      DRPLA is caused by abnormal expansion of CAG trinucleotide repeats over 48 tandem copies in full-penetrance alleles in exon 5 of ATN1 gene (12p13.31) [1,3]. There is marked intrafamilial variability and both clinical presentation and genetic anticipation have a direct correlation with the size of CAG expansion repeats [1,3]. Clinical presentation and disease severity depend mainly on the age of onset and length of CAG expansion repeats [1,2].

    • Principles and Practice of Movement Disorders

      2021, Principles and Practice of Movement Disorders
    • Epilepsy genetics-considerations for clinical practice today and for the future

      2020, Rosenberg’s Molecular and Genetic Basis of Neurological and Psychiatric Disease: Volume 2
    View all citing articles on Scopus
    View full text