U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Adam MP, Feldman J, Mirzaa GM, et al., editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2024.

Cover of GeneReviews®

GeneReviews® [Internet].

Show details

VPS13A Disease

Synonyms: Chorea-Acanthocytosis (ChAc), Choreoacanthocytosis

, MD, , DPhil, , PhD, , MD, , PhD, , PhD, , MD, PhD, , MB, ChB, PhD, , MD, PhD, and , MD.

Author Information and Affiliations

Initial Posting: ; Last Update: March 30, 2023.

Estimated reading time: 46 minutes

Summary

Clinical characteristics.

VPS13A disease, caused by VPS13A loss-of-function pathogenic variants, is characterized by a spectrum of movement disorders (chorea, dystonia, tics, sometimes parkinsonism); predominant orofacial choreic and dystonic movements and tics (with involuntary tongue protrusion on attempted swallowing, habitual tongue and lip biting resulting in self-mutilation, involuntary vocalizations); dysarthria and dysphagia; psychiatric, cognitive, and behavioral changes ("frontal lobe type"); seizures; and progressive neuromuscular involvement. Huntingtonism (triad of progressive movement disorder and cognitive and behavioral alterations) is a typical presentation. Phenotypic variability is considerable even within the same family, including for monozygotic twins. Mean age of onset is about 30 years. VPS13A disease runs a chronic progressive course and may lead to major disability within a few years. Some affected individuals are bedridden or wheelchair dependent by the third decade. Age at death ranges from 28 to 61 years; several instances of sudden unexplained death or death during epileptic seizures have been reported.

Diagnosis/testing.

The diagnosis of VPS13A disease is established in a proband with suggestive findings and biallelic pathogenic variants in VPS13A identified by molecular genetic testing.

Management.

Treatment of manifestations: There is no cure for VPS13A disease. Supportive treatment to improve quality of life, maximize function, and reduce complications is recommended. This ideally involves multidisciplinary care by specialists in relevant fields of neurology, psychiatry, physiatry, physical therapy (PT), occupational therapy (OT), speech-language therapy, feeding, neuropsychology, and medical genetics. Pharmacotherapy for movement disorders may include dopamine antagonists/depleters such as atypical neuroleptics or tetrabenazine (or its derivatives) for limb and trunk dystonia and orofaciolingual dystonia (which may also benefit from botulinum toxin). Issues with mobility, activities of daily living, and need for assistive devices can be addressed by physiatry, PT, and OT. In persons with dysphagia, feeding assistance can include speech therapy and gastrostomy tube placement as needed to reduce weight loss and/or risk of aspiration. For dysarthria or mutism, therapy can include the use of technical means for augmentative and alternative communication, such as speech-generating devices. Seizure management can include use of phenytoin, clobazam, valproate, and levetiracetam. For psychiatric/behavioral issues, antidepressant or antipsychotic medications are used per conventional approaches.

Surveillance: Regular monitoring of existing manifestations, the individual's response to pharmacotherapy and other supportive care, and the emergence of new manifestations is recommended per the multidisciplinary treating specialists.

Agents/circumstances to avoid: Seizure-provoking circumstances (e.g., sleep deprivation, alcohol intake) and anticonvulsants that may worsen involuntary movements (e.g., carbamazepine, lamotrigine).

Genetic counseling.

VPS13A disease is inherited in an autosomal recessive manner. If both parents are known to be heterozygous for a VPS13A pathogenic variant, each sib of an affected individual has at conception a 25% chance of being affected, a 50% chance of being an asymptomatic carrier, and a 25% chance of being unaffected and not a carrier. Once the VPS13A pathogenic variants have been identified in an affected family member, carrier testing for at-risk relatives, prenatal testing for a pregnancy at increased risk, and preimplantation genetic testing are possible.

Diagnosis

"VPS13A disease" refers to the disorder commonly known as chorea-acanthocytosis; the genetically precise term "VPS13A disease" is preferred because the presence of chorea and/or acanthocytosis is neither necessary nor sufficient to diagnose the disorder [Walker & Danek 2021].

Suggestive Findings

VPS13A disease should be suspected in probands with the following clinical, laboratory, and imaging findings and family history. "Red flag" findings are summarized in Table 1, and more detailed information follows the table.

Table 1.

VPS13A Disease: Red Flag Findings

VPS13A Disease: Red Flag Findings
Clinical findings Triad of progressive movement disorder (e.g., chorea, dystonia, in later stages parkinsonism), cognitive alterations, & behavioral alterations ("huntingtonism")
Prominent orofacial chorea, dystonia (feeding dystonia), & tics
Epileptic seizures
Weak to absent tendon stretch reflexes
Laboratory findings HyperCKemia
Presence of acanthocytes in peripheral blood 1
Neuroimaging Atrophy of the caudate on neuroimaging
1.

Acanthocytosis is neither necessary nor sufficient to diagnose the disorder.

Clinical Findings

Suggestive clinical findings include:

  • Huntingtonism triad of progressive deterioration of movement, cognition, and behavior
  • Progressive movement disorder
    • Commonly chorea and dystonia in early disease stages
    • Sometimes a parkinsonian syndrome, especially in later disease stages
    • Predominant orofacial choreic and dystonic movements and tics
  • Dysarthria and dysphagia with resultant weight loss
  • Progressive cognitive and behavioral changes (of "frontal lobe type," i.e., executive dysfunction, impaired social cognition) [Walterfang et al 2008]
  • Psychosis
  • Seizures, which can be the initial manifestation; sometimes suggestive of a familial temporal lobe epilepsy [Al-Asmi et al 2005, Scheid et al 2009]
  • Progressive neuromuscular involvement characterized by distal muscle wasting and weakness. This can be subclinical (only creatine kinase [CK] elevation). Electromyography commonly reveals chronic denervation and, in some instances, also myopathic changes [Vaisfeld et al 2021].
  • Peripheral neuropathy with impaired deep tendon reflexes and vibration sense contributing to muscle weakness and atrophy. Electrophysiologic tests demonstrate a sensory or sensorimotor axonopathy.

Family History

Family history is consistent with autosomal recessive inheritance (e.g., affected sibs and/or parental consanguinity). Absence of a known family history does not preclude the diagnosis.

Supportive Laboratory Findings

Muscle and liver enzymes, markers of hemolysis

  • Increased serum concentration of muscle CK is observed in the majority of individuals.
  • Less commonly, serum concentrations of aspartate transaminase and alanine transaminase are increased.
  • Levels of haptoglobin can be reduced and levels of lactate dehydrogenase can be increased due to chronic (subclinical) hemolysis [Rampoldi et al 2002].

Acanthocytosis. Acanthocytes usually comprise 5%-50% of the red blood cell population in individuals with VPS13A disease; however, in some cases, acanthocytes may be absent [Bayreuther et al 2010] or may appear only late in the disease course [Sorrentino et al 1999].

Note: (1) The proportion of acanthocytes does not correlate with disease severity. (2) Presence of acanthocytes is neither "specific" nor "sensitive" for the diagnosis of VPS13A disease. (3) The following methods, which are appropriate for testing, are not ubiquitously available.

  • A standard routine procedure is to dilute blood 1:1 with 0.9% saline containing 10 U/mL heparin, and examine it using phase-contrast microscopy after 30 minutes' incubation in a shaker and wet blood smear preparation. In control samples, fewer than 6.3% of cells are speculated [Storch et al 2005].
    Note: (1) Dry blood smears seem inadequate [Alawneh et al 2012]. (2) The suggestion to perform blood smears on three (or more) occasions [Sokolov et al 2012] in order to exclude acanthocytosis is unfounded.
  • Scanning electron microscopy of erythrocytes fixed with glutaraldehyde is probably the most reliable method of detecting acanthocytes but is not routinely available. Distinguishing acanthocytes from erythrocytes of other shapes can be difficult, as their definitions may appear insufficient in the individual case [Peikert et al 2022b]. Although use of artificial neural networks to discriminate acanthocytes from other abnormally shaped erythrocytes might be useful [Simionato et al 2021, Peikert et al 2022b], this is not yet clinically available.
  • Decreased erythrocyte sedimentation rate may emerge as a simpler indirect indicator of acanthocytosis [Darras et al 2021].

Detection of absent or marked reduction of VPS13A protein (formerly chorein). See Molecular Genetics, VSP13A-specific laboratory technical considerations.

Neuroimaging

Cranial CT and brain MRI reveal atrophy of the caudate nuclei with dilatation of the anterior horns of the lateral ventricles. The extent of basal ganglia atrophy is best appreciated on sections in the frontal plane. MRI may show T2-weighted signal increase in the caudate and putamen; occasionally iron deposition may be observed [Lee et al 2011, Kaul et al 2013].

In addition to the caudate nucleus, the putamen also shows significant and marked reduction in volume compared with controls [Walterfang et al 2011b].

Hippocampal sclerosis and atrophy are also frequently seen [Al-Asmi et al 2005, Huppertz et al 2008, Scheid et al 2009, Mente et al 2017].

There may be mild generalized cerebral cortical atrophy.

Although cerebellar atrophy has been reported in a few individuals, to date genetic diagnoses have not been established in these individuals [Tsai et al 1997, Katsube et al 2009, Jiang et al 2012, Sharma et al 2014].

Establishing the Diagnosis

The diagnosis of VPS13A disease is established in a proband with suggestive findings and biallelic pathogenic (or likely pathogenic) variants in VPS13A identified by molecular genetic testing (see Table 2).

Note: (1) Per ACMG/AMP variant interpretation guidelines, the terms "pathogenic variant" and "likely pathogenic variant" are synonymous in a clinical setting, meaning that both are considered diagnostic and can be used for clinical decision making [Richards et al 2015]. Reference to "pathogenic variants" in this GeneReview is understood to include likely pathogenic variants. (2) Identification of biallelic VPS13A variants of uncertain significance (or of one known VPS13A pathogenic variant and one VPS13A variant of uncertain significance) does not establish or rule out the diagnosis.

Molecular genetic testing approaches. Gene-targeted testing requires that the clinician determines which gene(s) are likely involved, whereas genomic testing does not. Because the phenotype of VPS13A disease is broad, individuals with the distinctive findings described in Suggestive Findings are likely to be diagnosed using gene-targeted testing (see Option 1), whereas those with a phenotype indistinguishable from many other inherited disorders with generalized chorea and/or epileptic seizures may be more likely to be diagnosed using genomic testing (see Option 2).

Note: Single-gene testing (sequence analysis of VPS13A, followed by gene-targeted deletion/duplication analysis) is rarely useful and typically NOT recommended.

Option 1

A chorea/dystonia multigene panel that includes VPS13A and other genes of interest (see Differential Diagnosis) may be considered to identify the genetic cause of the condition while limiting identification of variants of uncertain significance and pathogenic variants in genes that do not explain the underlying phenotype. Note: (1) The genes included in the panel and the diagnostic sensitivity of the testing used for each gene vary by laboratory and are likely to change over time. (2) Some multigene panels may include genes not associated with the condition discussed in this GeneReview. (3) In some laboratories, panel options may include a custom laboratory-designed panel and/or custom phenotype-focused exome analysis that includes genes specified by the clinician. (4) Methods used in a panel may include sequence analysis, deletion/duplication analysis, and/or other non-sequencing-based tests.

For an introduction to multigene panels click here. More detailed information for clinicians ordering genetic tests can be found here.

Option 2

Comprehensive genomic testing does not require the clinician to determine which genes are likely involved. Exome sequencing is most commonly used; genome sequencing is also possible.

For an introduction to comprehensive genomic testing click here. More detailed information for clinicians ordering genomic testing can be found here.

Table 2.

Molecular Genetic Testing Used in VPS13A Disease

Gene 1MethodProportion of Pathogenic Variants 2 Detectable by Method
VPS13A Sequence analysis 370%-90% 4
Gene-targeted deletion/duplication analysis 510%-30% 6
1.
2.

See Molecular Genetics for information on variants detected in this gene.

3.

Sequence analysis detects variants that are benign, likely benign, of uncertain significance, likely pathogenic, or pathogenic. Variants may include small intragenic deletions/insertions and missense, nonsense, and splice site variants; typically, exon or whole-gene deletions/duplications are not detected. For issues to consider in interpretation of sequence analysis results, click here.

4.

Rampoldi et al [2001], Dobson-Stone et al [2002], Tomiyasu et al [2011], Nishida et al [2019], and data derived from the subscription-based professional view of Human Gene Mutation Database [Stenson et al 2020]

5.

Gene-targeted deletion/duplication analysis detects intragenic deletions or duplications. Methods used may include a range of techniques such as quantitative PCR, long-range PCR, multiplex ligation-dependent probe amplification (MLPA), and a gene-targeted microarray designed to detect single-exon deletions or duplications.

6.

Deletion of exons 60-61 seems common in the Japanese population [Ueno et al 2001, Tomiyasu et al 2011], and deletion of exons 70-73 has been observed in the French Canadian population [Dobson-Stone et al 2005]; therefore, the proportion of pathogenic variants detected by sequence or gene-targeted deletion/duplication analysis varies by population.

Clinical Characteristics

Clinical Description

VPS13A disease is characterized by a progressive movement disorder, orofacial choreic and dystonic movements and tics, dysarthria and dysphagia, progressive cognitive and behavioral changes, psychosis, seizures, and progressive neuropathy and myopathy. Phenotypic variability is considerable and requires consideration of the diagnosis in a wide range of clinical conditions (including epilepsy, myopathy, and Tourette syndrome), with the huntingtonism triad as one typical presentation ("Huntington disease-like"). Acanthocytes may be found in blood smears, but the relevance of their presence or absence has been overstated (see Nomenclature). Mean age of onset in VPS13A disease is about age 30 years. VPS13A disease runs a chronic progressive course and may lead to major disability within a few years. Table 3 provides an overview of the major clinical findings.

Table 3.

Select Features of VPS13A Disease

FeatureRelative PrevalenceComment
Limb & trunk chorea +++Most prominent in early disease stages
Prominent orofacial chorea, dystonia, & tics Orofacial chorea +++
Tongue protrusion / feeding dystonia ++Suggests diagnosis when present
Tongue & lip biting ++Highly suggestive for diagnosis when present; may be caused by behavioral compulsion or tic
Involuntary vocalizations ++Typically meet criteria for tics
Parkinsonism +Typically more prevalent at later disease stages, but can also occur early or be presenting feature
Dysphagia +++
Dysarthria +++
Cognitive decline ++Variable
Behavioral/psychiatric changes ++Variable; compulsive behaviors can be prominent
Epilepsy ++Can predate other features
Axonal neuropathy ++Often mild, but diminished tendon reflexes are a "red flag"
Myopathy +Often mild, yet some persons have severe weakness/atrophy
Oculomotor abnormalities +Rarely conspicuous

+++ = very common; ++ = common; + = uncommon

Limb and trunk chorea is common and can include flinging arm and leg movements, shoulder shrugs, and pelvic thrusts. Stance and gait are typically affected by involuntary movements such as foot or leg chorea and dystonia. Violent trunk spasms may occur with sudden flexion or extension movements; head drops and head banging with a risk of head and neck injury can also occur [Schneider et al 2010].

A peculiar "rubber person gait" may appear due to a sudden lapse of muscle tone in the trunk or legs [Thomas & Jankovic 2003, Termsarasab & Frucht 2018] and may be interpreted as being functional (psychogenic). Impaired postural reflexes may result in falls, as may sudden buckling of knees [Yamamoto et al 1982] and equinovarus foot deformity, the latter related to dystonia as well as atrophy of the peroneal muscles.

The choreic syndrome gradually evolves into predominant parkinsonism with dystonia in about one third of affected individuals. Increased rigid muscle tone, rest tremor, impaired postural reflexes, bradykinesia, facial masking, and micrographia may appear. Occasionally, parkinsonism may be the presenting manifestation.

Orofacial chorea, dystonia, and tics. Predominance of orofacial chorea is very common. The involuntary movements that affect the face, mouth, tongue, pharynx, and larynx are the most characteristic.

Action-induced dystonic protrusion of the tongue while feeding is typical and causes the tongue to push the food out of the mouth. "Feeding dystonia" is the term commonly applied for this pattern of movement [Bader et al 2010].

Continuous tongue and lip biting caused by behavioral compulsion or tic/chorea can lead to self-mutilation, which can result in serious and challenging infections of the oral region. Affected individuals typically try to avoid this by keeping an object such as a handkerchief between the teeth, which may function either as a sensory trick to reduce dystonia or as a mechanical obstacle.

Involuntary vocalizations (vocal tics) are present in about two thirds of affected individuals [Saiki et al 2004]. The variety of described vocalizations (tics) include clicking, gasping, sighing, whistling, blowing, sucking, grunting noises, perseveration of word elements or phrases, and continuous humming.

There may be habitual teeth grinding (bruxism), spitting, or involuntary belching [Wihl et al 2001, Sibon et al 2004].

Dysphagia. The oral phase of swallowing is often impaired (while pharyngeal and esophageal phases seem intact), resulting in dysphagia with drooling and reduced caloric intake and potentially severe weight loss.

Dysarthria is common; slurred speech may be a presenting manifestation. In the course of VPS13A disease, communication may become limited to grunting or whispering. The hyperkinetic orofacial state may eventually progress to mutism [Aasly et al 1999].

Cognitive decline is common. Memory and executive functions, such as the ability to sustain concentration over time, planning, and modifying behavior, seem particularly affected. These findings resemble those of frontotemporal dementia [Danek et al 2004].

Behavioral/psychiatric changes. Changes in personality and behavior along with psychopathologic abnormalities occur in about two thirds of affected individuals [Danek et al 2004]. Apathy, depression, and bradyphrenia (slowness of thought) can be seen, but hyperactivity, irritability, distractibility, and emotional instability can also be observed. Individuals may behave in a disinhibited manner that can include sexual disinhibition. They may show obsessive-compulsive behavior including trichotillomania [Lossos et al 2005, Walterfang et al 2008] and self-inflicted chronic excoriations on the head [Walker et al 2006]. Loss of insight, self-neglect, anxiety, paranoia, aggression against others, and autoaggression are observed. Suicide and suicidal ideation are part of the disease spectrum [Sorrentino et al 1999].

Epilepsy is observed in almost half of affected individuals and can be the initial manifestation [Al-Asmi et al 2005]. It usually manifests as generalized tonic-clonic seizures and is probably secondarily generalized, for example, from temporal lobe foci [Scheid et al 2009, Bader et al 2011]. There may be prolonged states of memory impairment and confusion most likely corresponding to nonconvulsive seizures [Bader et al 2011, Mente et al 2017].

EEG may show temporal spikes, both interictally and with seizure onset [Scheid et al 2009].

Neuropathy and myopathy. Nerve and muscle involvement resulting in ankle areflexia is seen in almost all affected individuals and muscle atrophy and weakness in at least half of affected individuals. Symptoms suggestive of motor neuron disease have been reported [Neutel et al 2012]. Primary myopathic alterations can also be detected [Vaisfeld et al 2021]. Sensory loss is usually slight or may only be detected as reduced vibration sense. Pyramidal tract signs are usually absent, but bilateral extensor plantar responses were noted in one individual [Neutel et al 2012], and upper motor neuron involvement was found post mortem in another [Miki et al 2010].

Ocular motor abnormalities have been noted on occasion, with apraxia of lid opening, intermittent blepharospasm, frequent square wave jerks, slowing of saccades (mainly vertical), and reduced saccadic range [Gradstein et al 2005].

Other clinical findings

  • Dilated cardiomyopathy is rare [Kageyama et al 2007]; however, mild cardiac involvement may be more common than previously thought [Quick et al 2021].
  • Splenomegaly is occasionally noted and may be caused by erythrocyte dysfunction and hemolysis, as shown by reduced levels of hemoglobin and haptoglobin.
  • Hepatomegaly may be present, along with elevated liver enzymes; to date the clinical significance of this is unclear.
  • Autonomic nervous system dysfunction was described in one affected individual [Kihara et al 2002].
  • In a few individuals, sleep disturbance was demonstrated by polysomnography [Dolenc-Groselj et al 2004].
  • Chance co-occurrences with other conditions may complicate the clinical diagnostic process [Anheim et al 2010].

Other studies

  • MR spectroscopy may reveal abnormal proton spectra from the basal ganglia [Niemelä et al 2020].
  • Tracer imaging studies of the type presently available in most major medical centers may support a suspicion of VPS13A disease. Regional cerebral glucose metabolism, measured using 18F-fluorodeoxy-glucose positron emission tomography, shows striatal hypometabolism [Ehrlich & Walker 2017, Niemelä et al 2020].
  • Imaging of dopaminergic and serotoninergic transmission. Although ratios of binding to striatal dopamine transporters and serotonin transporters in the hypothalamus midbrain area as determined using (123)I-beta-CIT-SPECT fell within the normal ranges in two affected individuals, a significant difference in binding to presynaptic striatal dopamine transporters was observed [Müller-Vahl et al 2007]. Presynaptic dopaminergic deficiency was identified in some individuals [Niemelä et al 2020].
  • CT scanning of leg muscles reveals a selective pattern of symmetric fatty change [Ishikawa et al 2000].
  • Cerebrospinal fluid studies, when reported, have been normal.
  • Serum neurofilament light chain is significantly increased in affected individuals compared to healthy controls [Peikert et al 2020], as it is in many other neurodegenerative disorders.
  • Peripheral nerve biopsy has demonstrated loss of myelinated fibers, particularly those of larger diameter. Unmyelinated fibers may also be affected. Signs of regeneration have been observed [Sorrentino et al 1999].
  • Muscle biopsy revealed findings indicative of both neurogenic and myopathic atrophy [Vaisfeld et al 2021]. "Nemaline" rods in muscle have been reported, although their exact composition is unknown [Tamura et al 2005].

Neuropathology. Systematic neuropathologic studies are still lacking. Click here (pdf) for more information.

Prognosis. Life expectancy is reduced. Age at death ranges from 28 to 61 years.

Several instances of sudden unexplained death or death during epileptic seizures have been reported [Walker et al 2019].

Genotype-Phenotype Correlations

To date, available data are inconclusive with regard to genotype-phenotype correlations involving clinical manifestations and laboratory findings for VPS13A disease.

Nomenclature

To incorporate the genetic etiology of the disorder, Walker & Danek [2021] proposed that chorea-acanthocytosis be renamed VPS13A disease. The term "VPS13A disease" is preferred because the presence of chorea and/or acanthocytosis is neither necessary nor sufficient to diagnose the disorder [Walker & Danek 2021.]

"Neuroacanthocytosis" – a nonspecific umbrella term that may refer to any disorder with neurologic abnormalities and acanthocytosis (including McLeod neuroacanthocytosis syndrome) – should be used with great caution and is not a substitute for a definitive genetic diagnosis [Walker & Danek 2021].

The term "Levine-Critchley syndrome" is obsolete [Velayos-Baeza et al 2011; Danek et al, unpublished data].

Other outdated terms include "chorea-amyotrophy-acanthocytosis syndrome" and "familial amyotrophic chorea with acanthocytosis."

Prevalence

The number of individuals with VPS13A disease worldwide is estimated to be approximately 1:1,000,000 [Jung et al 2011].

The following observations might speak to founder effects; however, to date the overall prevalence of the following variants and VPS13A disease in these populations is unknown (see Table 8).

Differential Diagnosis

Because of the protean manifestations of VPS13A disease, a wide range of disorders needs to be considered in the differential diagnosis, including the general categories of parkinsonian syndromes (see Parkinson Disease Overview), hereditary dystonia, choreiform and other movement disorders, epilepsy disorders, and neuromuscular disorders.

Table 4.

Genes of Interest in the Differential Diagnosis of VPS13A Disease

Gene(s)DisorderMOIFeatures of Disorder
Overlapping w/VPS13A DiseaseDistinguishing from VPS13A Disease
ANGPTL3
APOB
Hypobetalipoproteinemia (See APOB-Related Familial Hypobetalipoproteinemia.)AR
  • Acanthocytosis
  • Dysarthria, neuropathy, & areflexia
  • Absence of basal ganglia movement disorder
  • Hallmark findings of pigmentary retinopathy, vitamin E deficiency, & steatorrhea
  • Spinocerebellar syndrome & sensorimotor neuropathy
ATN1 DRPLA AD
  • Choreoathetosis
  • Epilepsy
Ataxia
ATP7B Wilson disease AR
  • ↑ liver enzymes
  • Tremor, poor coordination, loss of fine motor control, chorea, & choreoathetosis OR rigid dystonia (mask-like facies, rigidity, gait disturbance, pseudobulbar involvement)
  • Psychiatric disturbance (depression, neurotic behaviors, disorganization of personality &, occasionally, intellectual deterioration)
  • Low serum copper & ceruloplasmin concentrations & ↑ urinary copper excretion, esp after chelator challenging
  • Prominent MRI abnormalities during disease progression
ELAC2 Combined oxidative phosphorylation deficiency-17 (COXPD17) (OMIM 615440)ARChorea, psychosis, acanthocytosis 1Apart from a single adult case w/ELAC2 pathogenic variant1 clinical findings & presentation age (early childhood) in COXPD17 differ greatly from VPS13A disease.
HPRT1 Lesch-Nyhan disease (See HPRT1 Disorders.)XL
  • Cognitive & behavioral disturbances
  • Self-injurious behavior (biting of lips, cheeks fingers, hands; head/limb banging)
  • Neurologic dysfunction (dystonia, choreoathetosis, opisthotonos)
  • Age at manifestation (early childhood) very different from VPS13A disease
  • Hyperuricemia
HTT Huntington disease (HD)AD
  • Chorea syndrome, changes of personality & behavior, & imaging findings in HD & VPS13A disease are almost identical.
  • Parkinsonism is typical for juvenile HD (Westphal variant) & transition to parkinsonism is not uncommon in late-stage HD.
  • Seizures are much more common in VPS13A disease than in HD.
  • ↑ serum concentrations of CK or liver enzymes & acanthocytosis are unusual for HD.
  • ↓ ankle reflexes are more prevalent in VPS13A disease.
  • The neuropathology of HD is more widespread & involves the cerebral cortex.
JPH3 Huntington disease-like 2 (HDL2)ADHuntingtonism typically presenting in midlife w/progression to death over 10-20 yrs
  • Acanthocytes are not present in great majority of affected persons.
  • Serum CK is normal.
  • Myopathy & seizures are absent.
  • HDL2 has been described exclusively in persons w/African ancestry.
MTTP Abetalipoproteinemia AR
  • Acanthocytosis
  • Dysarthria, neuropathy, & areflexia
  • Hallmark findings: presence of pigmentary retinopathy, vitamin E deficiency, steatorrhea, & absence of basal ganglia movement disorder
  • Spinocerebellar syndrome & sensorimotor neuropathy
PANK2 Pantothenate kinase-associated neurodegeneration (PKAN) (See also Neurodegeneration with Brain Iron Accumulation Disorders Overview.)AR
  • Early childhood onset of progressive dystonia, dysarthria, rigidity, & choreoathetosis
  • "Atypical" presentation: onset at age >10 yrs, prominent speech defects, psychiatric disturbances, & more gradual progression of disease
  • Acanthocytes often observed
  • "Eye of the tiger" MRI finding (identified on transverse images of globus pallidus as central region of hyperintensity surrounded by rim of hypointensity) in PKAN
  • Much younger age of disease onset
XK McLeod neuroacanthocytosis syndrome (also referred to as McLeod syndrome [MLS] or XK disease 2)XL
  • CNS manifestations (movement disorder, cognitive impairment, & psychiatric symptoms)
  • Neuromuscular manifestations (mostly subclinical sensorimotor axonopathy, muscle weakness, or atrophy)
  • Red blood cell acanthocytosis & compensated hemolysis
  • Usually later onset in MLS of some features shared w/VPS13A disease (e.g., huntingtonism, feeding dystonia, & head drops)
  • The McLeod blood group phenotype 3 distinguishes MLS from VPS13A disease (in which Kell blood group antigen expression is normal).
  • Malignant arrhythmias & cardiomyopathy are common.

AD = autosomal dominant; AR = autosomal recessive; CNS = central nervous system; MOI = mode of inheritance; XL = X-linked

1.
2.

Because the term "neuroacanthocytosis" refers to several genetically and phenotypically distinct disorders, the terms McLeod syndrome or XK disease are preferred by the authors [Walker & Danek 2021; Authors, personal observation].

3.

Hematologically, MLS is defined as a specific blood group phenotype (named after the first proband, Hugh McLeod; "McLeod blood group phenotype") that results from absent expression of the Kx erythrocyte antigen and weakened expression of Kell blood group antigens. Note: Transfusions of Kx-positive blood products should be avoided in persons w/the McLeod blood group phenotype. Kx-negative blood or, if possible, banked autologous or homologous blood should be used for transfusions.

Tourette syndrome is often diagnosed during initial stages of VPS13A disease [Saiki et al 2004, Müller-Vahl et al 2007, Walterfang et al 2008, Walterfang et al 2011a]. Its picture of motor and vocal tics, obsessive-compulsive behavior, and impaired impulse control can resemble the VPS13A disease spectrum.

Management

Evaluations Following Initial Diagnosis

To establish the extent of disease and needs in an individual diagnosed with VPS13A disease, the evaluations summarized in Table 5 (if not performed as part of the evaluation that led to the diagnosis) are recommended.

Table 5.

Recommended Evaluations Following Initial Diagnosis in Individuals with VPS13A Disease

System/ConcernEvaluationComment
Neurologic Assess for movement disorder(s).
  • Apply appropriate scales according to predominant movement disorder (e.g., for chorea UHDRS-TMS, for dystonia UDRS or FMDRS, for parkinsonism MDS-UPDRS Part III).
  • Perform structural brain imaging (if not performed previously or not available for review); MRI preferred.
Assess for seizures.
  • Assess seizure semiology & frequency.
  • Perform structural brain imaging (if not performed previously or not available for review); MRI preferred to assess for hippocampal sclerosis or other epileptogenic lesions.
  • Perform EEG. 1
Assess for neuromuscular involvement.
  • Assess muscle weakness or atrophy, DTRs, gross motor & fine motor skills, mobility, ADL, & need for adaptive devices.
  • Determine serum CK, ALT, AST, & LDH.
  • Perform EMG & NCV studies.
Mobility, ADL, & need for adaptive devices Eval by physiatrist, PT, OT
  • Assess need for protective devices (to counteract head banging & repeated falls).
  • Assess living situation (to ↓ risk of falls).
  • Assess need for AFOs for foot drop secondary to muscle weakness/dystonia.
Cognitive To incl motor & speech-language eval & general cognitive skills eval
  • Assess executive deficits & memory.
  • Perform formal neuropsychological eval &/or short tests such as MoCA. 2
  • Consider involving OT & neuropsychologist if needed.
Behavioral/
Psychiatric
Assess for OCD, personality change, anxiety, depression, bipolar disorder, schizo-affective disorder.
  • Perform standardized psychiatric assessment; eval of symptom-oriented psychotherapeutic & psychopharmacologic interventions.
  • Consider involving psychiatry specialist, psychologist, &/or neuropsychologist if needed.
Feeding/
Dysphagia/
Dysarthria
  • Feeding/nutritional assessment
  • Speech eval
  • Assess feeding/tongue protrusion dystonia.
  • Consider clinical &/or fiberoptic &/or radiologic feeding eval.
  • Nutrition is a significant issue; assess body weight regularly.
  • Assess possible dysarthria & communication skills, incl need for alternative means of communication (e.g., text-to-speech computer technology).
Cardiac Assess for cardiomyopathy, arrhythmia.
  • Perform echocardiography, EKG, & cardiac biomarker analysis (e.g., troponin T/I, pro-BNP).
  • If available, perform cardiac MRI.
Liver/Spleen Assess for hepatosplenomegaly.
  • Perform abdominal ultrasound exam.
Genetic counseling By genetics professionals 3 for facilitation of personal/medical decision making
  • Inform affected persons & families re nature of condition, MOI, implications of disease.
Family support
& resources
Assess need for:
  • Patient advocacy organization contact may be beneficial.
  • Home nursing can be considered to ↓ burden to patient & family.

ADL = activities of daily living; AFOs = ankle-foot orthoses; ALT = alanine transaminase; AST = aspartate transaminase; BNP = B-type natriuretic peptide; CK = creatine kinase; DTRs = deep tendon reflexes; EKG = electrocardiogram; EEG = electroencephalogram; EMG = electromyography; FMDRS = Fahn-Marsden Dystonia Rating Scale; LDH = lactate dehydrogenase; MDS-UPDRS = Movement Disorder Society-sponsored revision of the Unified Parkinson's Disease Rating Scale; MoCA = Montreal Cognitive Assessment; MOI = mode of inheritance; NCV = nerve conduction velocity; OCD = obsessive-compulsive disorder; OT = occupational therapist; PT = physical therapist; UDRS = Unified Dystonia Rating Scale; UHDRS-TMS = Unified Huntington Disease Rating Scale Total Motor Score

1.

Early recognition and treatment of seizures are important, as potential complications may be severe and could cause premature death [Walker et al 2019].

2.
3.

Medical geneticist, certified genetic counselor, certified advanced genetic nurse

Treatment of Manifestations

There is no cure for VPS13A disease.

Supportive care to improve quality of life, maximize function, and reduce complications is recommended, as for other disorders with similar findings. This ideally involves multidisciplinary care by specialists in relevant fields (see Table 6).

Table 6.

Treatment of Manifestations in Individuals with VPS13A Disease

Manifestation/ConcernTreatmentConsiderations/Other
Limb & trunk chorea
  • Dopamine depletors (i.e., VMAT2 inhibitors) or dopamine D2 receptor antagonists such as atypical neuroleptics should be offered.
  • Amantadine may be beneficial. 1
  • Monitor for side effects of parkinsonism & depression. 2
  • Neuroleptics can also help w/behavioral issues.
Orofacial chorea,
dystonia, & tics
  • Botulinum toxin may help ↓ the orolingual dystonia that interferes w/eating. 3
  • Orofacial chorea & tics can be ↓ by dopamine depleters & dopamine D2 receptor antagonists.
  • Keep object (e.g., handkerchief) in mouth to ↓ damage to lips & tongue from involuntary biting.
  • Use of mouth guard to prevent teeth grinding can also ↓ psychiatric manifestations. 4
Generalized dystonia
  • Standard medications for dystonia can be tried (e.g., benzodiazepines, anti-cholinergics).
  • Amantadine may be beneficial. 1
  • Physiotherapy (See Mobility, ADL, & need for adaptive devices below.)
  • Deep brain stimulation of globus pallidus pars interna may improve chorea & dystonia. 5
Consider local injections of botulinum toxin for dystonic equinovarus deformity.
Parkinsonism
  • Dopaminergic agents can be tried (w/caution due to psychotropic side effects).
  • Physiotherapy
Medications are often not effective.
Mobility, ADL, & need for adaptive devices
  • Physiatry to address need for adaptive devices to maintain/improve independence in mobility (e.g., canes, walkers, ramps to accommodate motorized chairs)
  • PT (balance exercises, gait training, muscle strengthening) to maintain mobility & function
  • OT to optimize ADL
  • Home adaptations to prevent falls (e.g., grab bars, raised toilet seats)
Dysphagia/Feeding
  • Speech/swallowing therapy
  • Gastrostomy tube placement
To be considered early to prevent weight loss
Dysarthria Speech-language therapyW/progression to mutism, eval for computer-assisted speech systems is appropriate.
Cardiac Per treating cardiologist
Seizures Phenytoin, clobazam, valproate, & levetiracetam are reported to be effective.
Cognitive decline Occupational &/or neuropsychological therapy
Behavioral/Psychiatric Use of psychiatric medications such as antidepressant or antipsychotic medications is based on conventional approaches.Behavioral compulsions, particularly those resulting in self-harm, should be aggressively treated w/antidepressant medications that target obsessive-compulsive symptoms.
Tics Levetiracetam 6 dopamine depleters & dopamine D2 receptor blockers
Family support
& resources
See Resources.

Surveillance

To monitor existing manifestations, the individual's response to supportive care, and the emergence of new manifestations, the following evaluations are recommended (see Table 7).

Table 7.

Recommended Surveillance for Individuals with VPS13A Disease

System/ConcernEvaluationFrequency
Movement disorders Assess response to & evaluate dosage of dopamine-depleting drugs (evolution into predominant parkinsonism/dystonia)At each visit, at least annually
Mobility, ADL, & need for adaptive devices Per treating physiatrist, PT, OTPer treating specialist
Dysphagia/
Feeding
  • Assess nutritional status & adaptation of diet to assure adequate caloric intake & prevent aspiration.
  • Assess need for gastrostomy tube & obtain informed consent as early as possible.
At each visit, at least annually
Dysarthria Per treating speech-language therapistPer treating speech-language therapist
Seizures Assess response to ASM.Per treating neurologist & patient response to therapy
EEGWhenever new-onset seizures are suspected; at least every other year 1
Behavioral/
Psychiatric/
Cognitive
Clinical impressionAt each visit, at least annually
Short cognitive screening test such as MoCA 2 &/or per treating specialistAt least every other year &/or per treating specialist
Neuromuscular system Measure serum CK levels to assess for possible rhabdomyolysis.At each visit, esp when under neuroleptic treatment
Cardiac W/known cardiac involvementPer treating specialist
W/o known cardiac involvement: cardiac exams (EKG, echocardiography, & cardiac biomarkers)Every 3-5 years
Family support
& resources
  • Eval of social, psychological, & financial situation
  • Assess family need for palliative/respite care, home nursing, & other local resources or follow-up genetic counseling if new questions arise (e.g., family planning).
At each visit, at least annually

ASM = anti-seizure medication; CK = creatine kinase; MoCA = Montreal Cognitive Assessment; OT = occupational therapist; PT = physical therapist

1.

Be aware of / monitor carefully seizure-provoking effects of antipsychotics/neuroleptics

2.

Agents/Circumstances to Avoid

Avoid the following:

  • Seizure-provoking circumstances (e.g., sleep deprivation, alcohol intake)
  • Anticonvulsants that may worsen involuntary movements/tics (e.g., carbamazepine, lamotrigine)

Evaluation of Relatives at Risk

It is appropriate to clarify the genetic status of apparently asymptomatic older and younger at-risk relatives of an affected individual in order to identify as early as possible those who would benefit from early recognition and treatment of potential manifestations of the disease such as seizures, as possible complications (e.g., status epilepticus, sudden unexpected death in epilepsy) may be severe.

See Genetic Counseling for issues related to testing of at-risk relatives for genetic counseling purposes.

Therapies Under Investigation

Several studies showed pathologically elevated tyrosine kinase Lyn activity in individuals with VSP13A disease and in disease models [De Franceschi et al 2011, De Franceschi et al 2012, Peikert et al 2021a, Peikert et al 2021b]. Lyn kinase inhibition was able to rescue the blood phenotype [Lupo et al 2016] and the neuronal phenotype [Stanslowsky et al 2016]. This evidence encouraged off-label treatment with the tyrosine kinase inhibitor dasatinib in three affected individuals. Although the reduction of both elevated Lyn kinase activity and accumulated autophagy markers suggested target engagement in red blood cells during treatment, clinical parameters remained essentially unchanged; of note, no clinically relevant side effects occurred. Putative biomarkers such as creatine kinase, serum neurofilament levels, and acanthocyte count failed to show consistent effects [Peikert et al 2021a]. Experimental follow up suggested failure of central nervous system targeting by orally administered dasatinib [Peikert et al 2021b]. Tyrosine kinase inhibitors with improved blood-brain barrier penetration would thus be needed. Thus, based on currently available information, tyrosine kinase inhibitors cannot be recommended to treat VPS13A disease.

VPS13A loss of function was reported to impair PI3K signaling leading to reduced store-operated Ca2+ entry (SOCE) and increased cell death [Pelzl et al 2017a, Pelzl et al 2017b]. VPS13A upregulates the serum- and glucocorticoid-inducible kinase SGK1, which targets Na+/K+-ATPase, whose capacity was shown to be reduced in the absence of VSP13A [Hosseinzadeh et al 2020]. Both phenotypes were restored in cell models by lithium treatment. Although this observation suggests that lithium treatment might be preferable to other mood stabilizers should they be indicated in an individual with VPS13A disease, no systematic studies of lithium treatment have been reported to date.

Search ClinicalTrials.gov in the US and EU Clinical Trials Register in Europe for access to information on clinical studies for a wide range of diseases and conditions. Note: There may not be clinical trials for this disorder.

Genetic Counseling

Genetic counseling is the process of providing individuals and families with information on the nature, mode(s) of inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members; it is not meant to address all personal, cultural, or ethical issues that may arise or to substitute for consultation with a genetics professional. —ED.

Mode of Inheritance

VPS13A disease is inherited in an autosomal recessive manner.

Note: Previous speculation as to possible autosomal dominant inheritance of VPS13A disease has been disproven in the respective families [Danek et al 2012].

Risk to Family Members

Parents of a proband

  • The parents of an affected child are presumed to be heterozygous for a VPS13A pathogenic variant.
  • Molecular genetic testing is recommended for the parents of the proband to confirm that both parents are heterozygous for a VPS13A pathogenic variant and to allow reliable recurrence risk assessment.
  • If a pathogenic variant is detected in only one parent and parental identity testing has confirmed biological maternity and paternity, it is possible that one of the pathogenic variants identified in the proband occurred as a de novo event in the proband or as a postzygotic de novo event in a mosaic parent [Jónsson et al 2017]. If the proband appears to have homozygous pathogenic variants (i.e., the same two pathogenic variants), additional possibilities to consider include:
  • Based on current knowledge, heterozygotes (carriers) do not have features of VPS13A disease and not at risk of developing the disorder [Walker et al 2012b].

Sibs of a proband

  • If both parents are known to be heterozygous for a VPS13A pathogenic variant, each sib of an affected individual has at conception a 25% chance of being affected, a 50% chance of being an asymptomatic carrier, and a 25% chance of being unaffected and not a carrier.
  • Significant phenotypic variability may be observed between affected sibs who inherit the same biallelic pathogenic variants [Merwick et al 2014]; different phenotypes have even been observed in monozygotic twins [Müller-Vahl et al 2007].
  • Based on current knowledge, heterozygotes (carriers) do not have features of VPS13A disease and are not at risk of developing the disorder [Walker et al 2012b].

Offspring of a proband. Unless an individual with VPS13A disease has children with an affected individual or a carrier, offspring will be obligate heterozygotes (carriers) for a pathogenic variant in VPS13A (see Related Genetic Counseling Issues, Family planning).

Other family members. Each sib of the proband's parents is at a 50% risk of being a carrier of a VPS13A pathogenic variant.

Carrier Detection

Carrier testing for at-risk relatives requires prior identification of the VPS13A pathogenic variants in the family.

Related Genetic Counseling Issues

Family planning

  • The optimal time for determination of genetic risk and discussion of the availability of prenatal/preimplantation genetic testing is before pregnancy.
  • It is appropriate to offer genetic counseling (including discussion of potential risks to offspring and reproductive options) to young adults who are affected, are carriers, or are at risk of being carriers.
  • Carrier testing should be considered for the reproductive partners of individuals known to be affected with or carriers of VPS13A disease, particularly if consanguinity is likely and/or if both partners are of the same ethnic background.

Prenatal Testing and Preimplantation Genetic Testing

Once the VPS13A pathogenic variants have been identified in an affected family member, prenatal and preimplantation genetic testing are possible.

Differences in perspective may exist among medical professionals and within families regarding the use of prenatal testing. While most centers would consider use of prenatal testing to be a personal decision, discussion of these issues may be helpful.

Resources

GeneReviews staff has selected the following disease-specific and/or umbrella support organizations and/or registries for the benefit of individuals with this disorder and their families. GeneReviews is not responsible for the information provided by other organizations. For information on selection criteria, click here.

  • Advocacy for Neuroacanthocytosis Patients
    United Kingdom
    Phone: 44 (0) 20 7460-8874
    Email: ginger@naadvocacy.org
  • Neuroacanthocytosis Advocacy USA, Inc.
    Email: susan@naadvocacyusa.org; joy@naadvocacyusa.org

Molecular Genetics

Information in the Molecular Genetics and OMIM tables may differ from that elsewhere in the GeneReview: tables may contain more recent information. —ED.

Table A.

VPS13A Disease: Genes and Databases

GeneChromosome LocusProteinLocus-Specific DatabasesHGMDClinVar
VPS13A 9q21​.2 Intermembrane lipid transfer protein VPS13A VPS13A database VPS13A VPS13A

Data are compiled from the following standard references: gene from HGNC; chromosome locus from OMIM; protein from UniProt. For a description of databases (Locus Specific, HGMD, ClinVar) to which links are provided, click here.

Table B.

OMIM Entries for VPS13A Disease (View All in OMIM)

200150CHOREOACANTHOCYTOSIS; CHAC
605978VACUOLAR PROTEIN SORTING 13 HOMOLOG A; VPS13A

Molecular Pathogenesis

VPS13A encodes the protein VPS13A (also called chorein), a member of a recently recognized superfamily of bridge-like lipid transfer proteins [Leonzino et al 2021, Neuman et al 2022]. These proteins are localized at membrane contact sites, where pairs of intracellular membranes are held in close apposition [Lang et al 2015, Park et al 2016, Kumar et al 2018, Yeshaw et al 2019].

VPS13A is one of four very similar mammalian VPS13 paralogues that have different subcellular localizations and functions in cell and organismal physiology [Kolehmainen et al 2003, Velayos-Baeza et al 2004, Lesage et al 2016, Gauthier et al 2018, Seong et al 2018]. VPS13 family proteins are characterized by a conserved structural organization: they are large, rod-shaped proteins with a hydrophobic channel that extends along the entire length of the protein [Li et al 2020, Dziurdzik & Conibear 2021, Leonzino et al 2021, Adlakha et al 2022, Cai et al 2022]. The ends of the protein interact with the two closely apposed membranes, so that the hydrophobic channel provides a conduit for the flow of lipids between them. VPS13A has been localized to contact sites between the endoplasmic reticulum (ER) and three different structures, the mitochondrion, the lipid droplet, and the plasma membrane [Kumar et al 2018, Yeshaw et al 2019, Guillén-Samander et al 2022]. It has also been found at contacts between mitochondria and endosomes [Muñoz-Braceras et al 2019]. Disruption of lipid exchange at one or more of these contacts is thought to be the basis for functional impairment of neurons and red blood cells (neurodegeneration and acanthocytosis, respectively).

It is not yet known how lipid flow through VPS13A is regulated or at which of these contact sites the loss of lipid flow is relevant to disease causation. However, the extreme C-terminal end of VPS13A (a PH domain) has been shown to bind directly to the XK protein, loss of which is responsible for McLeod neuroacanthocytosis syndrome (also referred to as McLeod syndrome or XK disease), a disorder characterized by clinical manifestations very similar to those of VPS13A disease [Ho et al 1994, Urata et al 2019, Park & Neiman 2020, Guillén-Samander et al 2022, Park et al 2022, Peikert et al 2022a]. XK is a plasma membrane-localized lipid scramblase that mediates movement of phospholipids between the two leaflets of a lipid bilayer [Suzuki et al 2014, Ryoden et al 2022]. One effect of this scramblase activity is exposure to the cell surface of phosphatidylserine, a phospholipid normally confined to the cytosolic leaflet of the plasma membrane [Suzuki et al 2014, Ryoden et al 2022]. A VPS13A-XK complex is found at ER-to-plasma membrane contact sites, where it may pair transport of lipids between the ER and plasma membrane with movement of lipids between the two plasma membrane leaflets [Guillén-Samander et al 2022]. Given the similarity of VPS13A disease to McLeod syndrome, disruption of the functional partnership between the proteins VPS13A and XK is likely to have a key role in disease causation [Peikert & Danek 2023].

Loss of VPS13A has been associated with increased Lyn kinase activity, disturbed autophagy, and alteration of the actin cytoskeleton [De Franceschi et al 2011, Föller et al 2012, Lupo et al 2016]. Indeed, inhibitors of Lyn kinase have been shown to be effective in restoring the peripheral blood cell phenotypes of individuals with VPS13A disease [Peikert et al 2021a]. However, cellular phenotypes in neuronal cells derived from individuals with VPS13A disease were only partially reverted by Lyn kinase inhibition [Stanslowsky et al 2016, Glaß et al 2018]. Given the biochemical function of VPS13A in lipid transfer, the question of how impairment of this function may cause alterations of Lyn kinase activity and of actin organization has yet to be resolved.

Vps13a-/- mice recapitulate key features of individuals with VSP13A disease, showing loss of VPS13A and presence of blood cell acanthocytosis, disturbed autophagy, neuronal loss, and neuroinflammation in the basal ganglia and motor disturbances, but the neurologic changes occurred late and appeared mild in comparison to findings in humans [Peikert et al 2021b]. Similar findings were reported in another mouse model, with exons 60-61 deletion in Vps13a [Tomemori et al 2005]. Further characterization of these mouse models is needed.

Mechanism of disease causation. Loss of function. In addition to the majority of VPS13A disease-associated variants being predicted to be loss-of-function variants, western blotting has shown that many result in loss of VPS13A expression. Occasionally individuals with VPS13A disease may express VPS13A that lacks the PH domain that binds to the XK protein [Park et al 2022].

VPS13A-specific laboratory technical considerations. Absence or marked reduction of VPS13A (also known as chorein) on western blot analysis has been shown in erythrocytes from individuals with VPS13A disease. In contrast, normal levels of VPS13A are observed in samples from individuals with Huntington disease and healthy controls [Dobson-Stone et al 2004]. Therefore, western blot analysis of VPS13A may be helpful in the following circumstances:

  • Variants of uncertain significance (VUS) in VPS13A
  • Negative molecular analysis of VPS13A in a proband whose phenotype is consistent with VPS13A disease
  • Used as a first diagnostic indicator when DNA analysis is not affordable or generally unavailable

Individuals with reduced levels of VPS13A need further diagnostic workup by molecular genetic testing [Spieler et al 2020].

Notes: (1) Some pathogenic variants in VPS13A are known to be associated with normal levels of VPS13A (e.g., some missense substitutions that do not result in misfolding and protein degradation or small deletions leading to expression of a truncated protein whose electrophoretic motility is very similar to wild type protein [Park et al 2022]); therefore, the presence of normal levels of VPS13A does not exclude the diagnosis of VPS13A disease. (2) Because reduced VPS13A immunoreactivity has been observed in individuals with McLeod neuroacanthocytosis syndrome (most likely because of destabilization of VPS13A in the absence of the protein XK, with which it forms a complex [Urata et al 2019]), immunohistochemistry of VPS13A needs to be carefully interpreted in the context of the clinical findings.

Table 8.

Notable VPS13A Pathogenic Variants

Reference SequencesDNA Nucleotide ChangePredicted Protein ChangeComment [Reference]
NM_033305​.3
NP_150648​.2
c.2343delAp.Lys781AsnfsTer8Reported in 3 families from the Jewish population of Djerba Island, Tunisia [Benninger et al 2016]
c.4411C>Tp.Arg1471TerPathogenic variants reported in several Japanese families [Nishida et al 2019]
NM_033305​.3 Deletion of exons 60-61 1--
Deletion of exons 70-73 1--Deletion reported in French Canadian families [Dobson-Stone et al 2005]

Variants listed in the table have been provided by the authors. GeneReviews staff have not independently verified the classification of variants.

GeneReviews follows the standard naming conventions of the Human Genome Variation Society (varnomen​.hgvs.org). See Quick Reference for an explanation of nomenclature.

1.

Variant designation that does not conform to current naming conventions

Chapter Notes

Author Notes

The following web pages provide descriptions of our clinical work, research interests, and contact information:

As an international community of clinicians, scientists, and families dealing with VPS13A disease (chorea-acanthocytosis), XK disease (McLeod neuroacanthocytosis syndrome), and related disorders, we initiated the bimonthly virtual "VPS13 forum." In this forum, we regularly discuss all aspects (from bench to bedside) of this emerging field [Peikert & Danek 2023]. For invitations to future VPS13 forum sessions, contact ed.kcotsor-inu.dem@trekiep.nivek.

Drs Adrian Danek (ed.uml@kenad), Ruth Walker (ude.mssm@reklaw.htur), Andreas Hermann (ed.kcotsor-inu.dem@nnamreh.saerdna), Kevin Peikert (ed.kcotsor-inu.dem@trekiep.nivek), and Hans Jung (hc.zsu@gnuj.snah) are actively involved in clinical research regarding individuals with VPS13A disease. They would be happy to communicate with persons who have any questions regarding the diagnosis of VPS13A disease or other considerations.

Drs Adrian Danek (ed.uml@kenad), Ruth Walker (ude.mssm@reklaw.htur), Andreas Hermann (ed.kcotsor-inu.dem@nnamreh.saerdna), Kevin Peikert (ed.kcotsor-inu.dem@trekiep.nivek), and Hans Jung (hc.zsu@gnuj.snah) are also interested in hearing from clinicians treating families affected by "neuroacanthocytosis" syndromes and Huntington disease-like syndromes in whom no causative variant has been identified through molecular genetic testing of the genes known to be involved in this group of disorders.

Contact Drs Gabriel Miltenberger-Miltenyi (tp.aobsilu.anicidem@iynetlimg) and/or Dr Antonio Velayos Baeza (moc.liamtoh@soyaleva; ku.ca.xo.gapd@soyalev.oinotna) to inquire about review of VPS13A variants of uncertain significance.

Western blot analysis for VPS13A is currently available on a research basis (contact ed.kcotsor-inu.dem@trekiep.nivek).

Acknowledgments

We are grateful to Glenn Irvine, who sadly passed away, and Ginger Irvine, the founders of the Advocacy for Neuroacanthocytosis Patients, and to Susan Wagner and Joy Willard-Williford as representatives of Neuroacanthocytosis Advocacy USA. In Glenn Irvine's memory a prize for research in VPS13A and XK diseases (chorea-acanthocytosis and McLeod neuroacanthocytosis syndrome) is awarded to junior investigators whose work has substantially contributed to the field.

We thank Antonio Velayos Baeza and Benedikt Bader, former coauthors of this GeneReview on chorea-acanthocytosis / VPS13A disease, for their work in the field and the highly appreciated contribution to this valuable resource. Some years ago, Profs Thomas Witt and Alexander Storch provided decisive input.

Author History

Benedikt Bader, MD; Ludwig-Maximilians-Universität (2010-2023)

Adrian Danek, MD (2002-present)

Pietro De Camilli, PhD (2023-present)

Carol Dobson-Stone, DPhil (2002-present)

Andreas Hermann, MD, PhD (2023-present)

Gabriel Miltenberger-Miltenyi, MD (2023-present)

Anthony P Monaco, MD, PhD (2002-present)

Aaron Neiman, PhD (2023-present)

Kevin Peikert, MD (2023-present)

Luca Rampoldi, PhD (2002-present)

Antonio Velayos Baeza, PhD; Wellcome Trust Centre for Human Genetics (2004-2023)

Ruth H Walker, MB, ChB, PhD (2006-present)

Revision History

  • 30 March 2023 (bp) Comprehensive update posted live
  • 18 April 2019 (avb) Revision: New information on VPS13C and VPS13D
  • 30 January 2014 (me) Comprehensive update posted live
  • 18 August 2011 (cd) Revision: prenatal testing available clinically as listed in the GeneTests Laboratory Directory
  • 6 July 2010 (me) Comprehensive update posted live
  • 13 October 2006 (me) Comprehensive update posted live
  • 10 January 2005 (ad) Revision: Differential Diagnosis; Testing
  • 16 July 2004 (me) Comprehensive update posted live
  • 14 June 2002 (me) Review posted live
  • 7 March 2002 (lr) Original submission

References

Literature Cited

  • Aasly J, Skandsen T, Rø M. Neuroacanthocytosis--the variability of presenting symptoms in two siblings. Acta Neurol Scand. 1999;100:322-5. [PubMed: 10536920]
  • Adlakha J, Hong Z, Li PQ, Reinisch KM. Structural and biochemical insights into lipid transport by VPS13 proteins. J Cell Biol. 2022;221:e202202030 [PMC free article: PMC8978259] [PubMed: 35357422]
  • Al-Asmi A, Jansen AC, Badhwar A, Dubeau F, Tampieri D, Shustik C, Mercho S, Savard G, Dobson-Stone C, Monaco AP, Andermann F, Andermann E. Familial temporal lobe epilepsy as a presenting feature of choreoacanthocytosis. Epilepsia. 2005;46:1256-63 [PubMed: 16060937]
  • Alawneh J, Baker MR, Young GR. Blood films in the investigation of chorea. Pract Neurol. 2012;12:268 [PubMed: 22869776]
  • Anheim M, Chamouard P, Rudolf G, Ellero B, Vercueil L, Goichot B, Marescaux C, Tranchant C. Unexpected combination of inherited chorea-acanthocytosis with MDR3 (ABCB4) defect mimicking Wilson's disease. Clinical genetics. 2010;78:294-5 [PubMed: 20695873]
  • Bader B, Vollmar C, Ackl N, Ebert A, la Fougère C, Noachtar S, Danek A. Bilateral temporal lobe epilepsy confirmed with intracranial EEG in chorea-acanthocytosis. Seizure. 2011;20:340-2. [PubMed: 21251854]
  • Bader B, Walker RH, Vogel M, Prosiegel M, McIntosh J, Danek A. Tongue protrusion and feeding dystonia: a hallmark of chorea-acanthocytosis. Mov Disord. 2010;25:127-9 [PubMed: 19938148]
  • Bayreuther C, Borg M, Ferrero-Vacher C, Chaussenot A, Lebrun C. [Chorea-acanthocytosis without acanthocytes]. Rev Neurol (Paris). 2010;166:100-3 [PubMed: 19497603]
  • Benninger F, Afawi Z, Korczyn AD, Oliver KL, Pendziwiat M, Nakamura M, Sano A, Helbig I, Berkovic SF, Blatt I. Seizures as presenting and prominent symptom in chorea-acanthocytosis with c.2343del VPS13A gene mutation. Epilepsia. 2016;57:549-56. [PubMed: 26813249]
  • Bohlega S, Al-Jishi A, Dobson-Stone C, Rampoldi L, Saha P, Murad H, Kareem A, Roberts G, Monaco AP. Chorea-acanthocytosis: clinical and genetic findings in three families from the Arabian peninsula. Mov Disord 2003;18:403-7 [PubMed: 12671946]
  • Borchardt CM, Jensen C, Dean CE, Tori J. Case study: childhood-onset tardive dyskinesia versus choreoacanthocytosis. J Am Acad Child Adolesc Psychiatry. 2000;39:1055-8 [PubMed: 10939235]
  • Cai S, Wu Y, Guillen-Samander A, Hancock-Cerutti W, Liu J, De Camilli P. In situ architecture of the lipid transport protein VPS13C at ER-lysosome membrane contacts. Proc Natl Acad Sci USA. 2022;119:e2203769119 [PMC free article: PMC9303930] [PubMed: 35858323]
  • Danek A, Bader B, Velayos-Baeza A, Walker RH. Autosomal recessive transmission of chorea-acanthocytosis confirmed. Acta Neuropathol. 2012;123:905-6 [PMC free article: PMC4629413] [PubMed: 22476160]
  • Danek A, Sheesley L, Tierney M, Uttner I, Grafman J. Cognitive and neuropsychiatric findings in McLeod syndrome and in chorea-acanthocytosis. In: Danek A, ed. Neuroacanthocytosis Syndromes. Dordrecht, the Netherlands: Springer; 2004:95-115.
  • Darras A, Peikert K, Rabe A, Yaya F, Simionato G, John T, Dasanna AK, Buvalyy S, Geisel J, Hermann A, Fedosov DA, Danek A, Wagner C, Kaestner L. Acanthocyte sedimentation rate as a diagnostic biomarker for neuroacanthocytosis syndromes: experimental evidence and physical justification. Cells. 2021;10:788. [PMC free article: PMC8067274] [PubMed: 33918219]
  • De Franceschi L, Scardoni G, Tomelleri C, Danek A, Walker RH, Jung HH, Bader B, Mazzucco S, Dotti MT, Siciliano A, Pantaleo A, Laudanna C. Computational identification of phospho-tyrosine sub-networks related to acanthocyte generation in neuroacanthocytosis. PLoS One. 2012;7:e31015 [PMC free article: PMC3280254] [PubMed: 22355334]
  • De Franceschi L, Tomelleri C, Matte A, Brunati AM, Bovee-Geurts PH, Bertoldi M, Lasonder E, Tibaldi E, Danek A, Walker RH, Jung HH, Bader B, Siciliano A, Ferru E, Mohandas N, Bosman GJ. Erythrocyte membrane changes of chorea-acanthocytosis are the result of altered Lyn kinase activity. Blood. 2011;118:5652-63 [PMC free article: PMC3217364] [PubMed: 21951684]
  • Dobson-Stone C, Danek A, Rampoldi L, Hardie RJ, Chalmers RM, Wood NW, Bohlega S, Dotti MT, Federico A, Shizuka M, Tanaka M, Watanabe M, Ikeda Y, Brin M, Goldfarb LG, Karp BI, Mohiddin S, Fananapazir L, Storch A, Fryer AE, Maddison P, Sibon I, Trevisol-Bittencourt PC, Singer C, Caballero IR, Aasly JO, Schmierer K, Dengler R, Hiersemenzel LP, Zeviani M, Meiner V, Lossos A, Johnson S, Mercado FC, Sorrentino G, Dupre N, Rouleau GA, Volkmann J, Arpa J, Lees A, Geraud G, Chouinard S, Nemeth A, Monaco AP. Mutational spectrum of the CHAC gene in patients with chorea-acanthocytosis. Eur J Hum Genet. 2002;10:773-81 [PubMed: 12404112]
  • Dobson-Stone C, Velayos-Baeza A, Filippone LA, Westbury S, Storch A, Erdmann T, Wroe SJ, Leenders KL, Lang AE, Dotti MT, Federico A, Mohiddin SA, Fananapazir L, Daniels G, Danek A, Monaco AP. Chorein detection for the diagnosis of chorea-acanthocytosis. Ann Neurol. 2004;56:299-302 [PubMed: 15293285]
  • Dobson-Stone C, Velayos-Baeza A, Jansen A, Andermann F, Dubeau F, Robert F, Summers A, Lang AE, Chouinard S, Danek A, Andermann E, Monaco AP. Identification of a VPS13A founder mutation in French Canadian families with chorea-acanthocytosis. Neurogenetics. 2005;6:151-8 [PubMed: 15918062]
  • Dolenc-Groselj L, Jazbec J, Kobal J. Sleep features in chorea-acanthocytosis. In: Danek A, ed. Neuroacanthocytosis Syndromes. Dordrecht, the Netherlands: Springer; 2004:123-5.
  • Dziurdzik SK, Conibear E. The Vps13 family of lipid transporters and its role at membrane contact sites. Int J Mol Sci. 2021;22: 2905. [PMC free article: PMC7999203] [PubMed: 33809364]
  • Ehrlich DJ, Walker RH. Functional neuroimaging and chorea: a systematic review. J Clin Mov Disord. 2017;4:8 [PMC free article: PMC5479019] [PubMed: 28649394]
  • Föller M, Hermann A, Gu S, Alesutan I, Qadri SM, Borst O, Schmidt EM, Schiele F, vom Hagen JM, Saft C, Schols L, Lerche H, Stournaras C, Storch A, Lang F. Chorein-sensitive polymerization of cortical actin and suicidal cell death in chorea-acanthocytosis. FASEB J. 2012;26:1526-34 [PubMed: 22227296]
  • Fontenelle LF, Leite MA. Treatment-resistant self-mutilation, tics, and obsessive-compulsive disorder in neuroacanthocytosis: a mouth guard as a therapeutic approach. J Clin Psychiatry. 2008;69:1186-7 [PubMed: 18687019]
  • Glaß H, Pal A, Reinhardt P, Sterneckert J, Wegner F, Storch A, Hermann A. Defective mitochondrial and lysosomal trafficking in chorea-acanthocytosis is independent of Src kinase signaling. Mol Cell Neurosci. 2018;92:137 [PubMed: 30081151]
  • Gauthier J, Meijer IA, Lessel D, Mencacci NE, Krainc D, Hempel M, Tsiakas K, Prokisch H, Rossignol E, Helm MH, Rodan LH, Karamchandani J, Carecchio M, Lubbe SJ, Telegrafi A, Henderson LB, Lorenzo K, Wallace SE, Glass IA, Hamdan FF, Michaud JL, Rouleau GA, Campeau PM. Recessive mutations in VPS13D cause childhood onset movement disorders. Ann Neurol. 2018;83:1089-95. [PubMed: 29518281]
  • Gradstein L, Danek A, Grafman J, Fitzgibbon EJ. Eye movements in chorea-acanthocytosis. Invest Ophthalmol Vis Sci. 2005;46:1979-87 [PubMed: 15914612]
  • Guillén-Samander A, Wu Y, Pineda SS, García FJ, Eisen J, Leonzino M, Uğur B, Kellis M, Heiman M and De Camilli P. A partnership of the lipid scramblase XK and of the lipid transfer protein VPS13A at the plasma membrane. Proc Natl Acad Sci USA. 2022;119:e2205425119 [PMC free article: PMC9436381] [PubMed: 35994651]
  • He W, Li C, Dong H, Shao L, Yin B, Li D, Ye L, Hu P, Zhang C, Yi W. Pallidus stimulation for chorea-acanthocytosis: a systematic review and meta-analysis of individual data. J Mov Disord. 2022;15:197-205. [PMC free article: PMC9536914] [PubMed: 35880382]
  • Ho M, Chelly J, Carter N, Danek A, Crocker P, Monaco AP. Isolation of the gene for McLeod syndrome that encodes a novel membrane transport protein. Cell. 1994;77:869-80. [PubMed: 8004674]
  • Hosseinzadeh Z, Hauser S, Singh Y, Pelzl L, Schuster S, Sharma Y, Höflinger P, Zacharopoulou N, Stournaras C, Rathbun DL, Zrenner E, Schöls L, Lang F. Decreased Na+/K+ ATPase expression and depolarized cell membrane in neurons differentiated from chorea-acanthocytosis patients. Sci Rep. 2020;10:8391. [PMC free article: PMC7242441] [PubMed: 32439941]
  • Huppertz HJ, Kroll-Seger J, Danek A, Weber B, Dorn T, Kassubek J. Automatic striatal volumetry allows for identification of patients with chorea-acanthocytosis at single subject level. J Neural Transm. 2008;115:1393-400 [PubMed: 18648728]
  • Ishikawa S, Tachibana N, Tabata KI, Fujimori N, Hayashi RI, Takahashi J, Ikeda SI, Hanyu N. Muscle CT scan findings in McLeod syndrome and chorea-acanthocytosis. Muscle Nerve. 2000;23:1113-6 [PubMed: 10883007]
  • Jiang Y, Wang Y, Zhang H. 4 cases report of cerebellar ataxia-acanthocytosis. Parkinsonism Relat Disord 2012;18:S75-S76
  • Jónsson H, Sulem P, Kehr B, Kristmundsdottir S, Zink F, Hjartarson E, Hardarson MT, Hjorleifsson KE, Eggertsson HP, Gudjonsson SA, Ward LD, Arnadottir GA, Helgason EA, Helgason H, Gylfason A, Jonasdottir A, Jonasdottir A, Rafnar T, Frigge M, Stacey SN, Th Magnusson O, Thorsteinsdottir U, Masson G, Kong A, Halldorsson BV, Helgason A, Gudbjartsson DF, Stefansson K. Parental influence on human germline de novo mutations in 1,548 trios from Iceland. Nature. 2017;549:519-22. [PubMed: 28959963]
  • Jung HH, Danek A, Walker RH. Neuroacanthocytosis syndromes. Orphanet J Rare Dis. 2011;6:68. [PMC free article: PMC3212896] [PubMed: 22027213]
  • Kageyama Y, Matsumoto K, Ichikawa K, Ueno S, Ichiba M, Nakamura M, Sano A. A new phenotype of chorea-acanthocytosis with dilated cardiomyopathy and myopathy. Mov Disord. 2007;22:1669-70 [PubMed: 17516458]
  • Katsube T, Shimono T, Ashikaga R, Hosono M, Kitagaki H, Murakami T. Demonstration of cerebellar atrophy in neuroacanthocytosis of 2 siblings. AJNR Am J Neuroradiol. 2009;30:386-8. [PMC free article: PMC7051402] [PubMed: 18945802]
  • Kaul B, Goyal V, Shukla G, Srivastava A, Garg A, Bader B, Danek A, Hayflick S, Behari M. Mineral deposition on magnetic resonance imaging in chorea-acanthocytosis: a pathogenic link with pantothenate kinase-associated neurodegeneration? Neurol India. 2013;61:169-70 [PubMed: 23644319]
  • Kihara M, Nakashima H, Taki M, Takahashi M, Kawamura Y. A case of chorea-acanthocytosis with dysautonomia; quantitative autonomic deficits using CASS. Auton Neurosci. 2002;97:42-4 [PubMed: 12036185]
  • Kolehmainen J, Black GC, Saarinen A, Chandler K, Clayton-Smith J, Traskelin AL, Perveen R, Kivitie-Kallio S, Norio R, Warburg M, Fryns JP, de la Chapelle A, Lehesjoki AE. Cohen syndrome is caused by mutations in a novel gene, COH1, encoding a transmembrane protein with a presumed role in vesicle-mediated sorting and intracellular protein transport. Am J Hum Genet. 2003;72:1359-69 [PMC free article: PMC1180298] [PubMed: 12730828]
  • Kumar N, Leonzino M, Hancock-Cerutti W, Horenkamp FA, Li P, Lees JA, Wheeler H, Reinisch KM, De Camilli P. VPS13A and VPS13C are lipid transport proteins differentially localized at ER contact sites. J Cell Biol 2018;217:3625–39 [PMC free article: PMC6168267] [PubMed: 30093493]
  • Lang AB, Peter ATJ, Walter P, Kornmann B. ER-mitochondrial junctions can be bypassed by dominant mutations in the endosomal protein VPS13. J Cell Biol. 2015;210:883-90. [PMC free article: PMC4576869] [PubMed: 26370498]
  • Lee JH, Lee SM, Baik SK. Demonstration of striatopallidal iron deposition in chorea-acanthocytosis by susceptibility-weighted imaging. J Neurol. 2011;258:321-2 [PubMed: 20798951]
  • Leonzino M, Reinisch KM, De Camilli P. Insights into VPS13 properties and function reveal a new mechanism of eukaryotic lipid transport. Biochim Biophys Acta Mol Cell Biol Lipids. 2021;1866:159003 [PMC free article: PMC8325632] [PubMed: 34216812]
  • Lesage S, Drouet V, Majounie E, Deramecourt V, Jacoupy M, Nicolas A, Cormier-Dequaire F, Hassoun SM, Pujol C, Ciura S, Erpapazoglou Z, Usenko T, Maurage CA, Sahbatou M, Liebau S, Ding J, Bilgic B, Emre M, Erginel-Unaltuna N, Guven G, Tison F, Tranchant C, Vidailhet M, Corvol JC, Krack P, Leutenegger AL, Nalls MA, Hernandez DG, Heutink P, Gibbs JR, Hardy J, Wood NW, Gasser T, Durr A, Deleuze JF, Tazir M, Destée A, Lohmann E, Kabashi E, Singleton A, Corti O, Brice A, et al. Loss of VPS13C function in autosomal-recessive parkinsonism causes mitochondrial dysfunction and increases PINK1/parkin-dependent mitophagy. Am J Hum Genet. 2016;98:500-13. [PMC free article: PMC4800038] [PubMed: 26942284]
  • Li P, Lees JA, Lusk CP, Reinisch KM. Cryo-EM reconstruction of a VPS13 fragment reveals a long groove to channel lipids between membranes. J Cell Biol. 2020;219: 3593 [PMC free article: PMC7199853] [PubMed: 32182622]
  • Lin FC, Wei LJ, Shih PY. Effect of levetiracetam on truncal tic in neuroacanthocytosis. Acta Neurol Taiwan 2006;15:38-42 [PubMed: 16599284]
  • Lossos A, Dobson-Stone C, Monaco AP, Soffer D, Rahamim E, Newman JP, Mohiddin S, Fananapazir L, Lerer I, Linetsky E, Reches A, Argov Z, Abramsky O, Gadoth N, Sadeh M, Gomori JM, Boher M, Meiner V. Early clinical heterogeneity in choreoacanthocytosis. Arch Neurol. 2005;62:611-4 [PubMed: 15824261]
  • Lupo F, Tibaldi E, Matte A, Sharma AK, Brunati AM, Alper SL, Zancanaro C, Benati D, Siciliano A, Bertoldi M, Zonta F, Storch A, Walker RH, Danek A, Bader B, Hermann A, De Franceschi L. A new molecular link between defective autophagy and erythroid abnormalities in chorea-acanthocytosis. Blood. 2016;128 :2976-87 [PMC free article: PMC5179337] [PubMed: 27742708]
  • Mente K, Kim SA, Grunseich C, Hefti MM, Crary JF, Danek A, Karp BI, Walker RH. Hippocampal sclerosis and mesial temporal lobe epilepsy in chorea-acanthocytosis: a case with clinical, pathologic and genetic evaluation. Neuropathol Appl Neurobiol. 2017;43: 542–6 [PMC free article: PMC5597461] [PubMed: 28398599]
  • Merwick Á, Mok T, McNamara B, Parfrey NA, Moore H, Sweeney BJ, Hand CK, Ryan AM. Phenotypic variation in a Caucasian kindred with chorea-acanthocytosis. Mov Disord Clin Pract. 2014;2:86-9. [PMC free article: PMC6353409] [PubMed: 30713887]
  • Miki Y, Nishie M, Ichiba M, Nakamura M, Mori F, Ogawa M, Kaimori M, Sano A, Wakabayashi K. Chorea-acanthocytosis with upper motor neuron degeneration and 3419_3420 delCA and 3970_3973 delAGTC VPS13A mutations. Acta Neuropathol. 2010;119:271-3 [PubMed: 19949804]
  • Miquel M, Spampinato U, Latxague C, Aviles-Olmos I, Bader B, Bertram K, Bhatia K, Burbaud P, Burghaus L, Cho JW, Cuny E, Danek A, Foltynie T, Garcia Ruiz PJ, Gimenez-Roldan S, Guehl D, Guridi J, Hariz M, Jarman P, Kefalopoulou ZM, Limousin P, Lipsman N, Lozano AM, Moro E, Ngy D, Rodriguez-Oroz MC, Shang H, Shin H, Walker RH, Yokochi F, Zrinzo L, Tison F. Short and long term outcome of bilateral pallidal stimulation in chorea-acanthocytosis. PLoS One. 2013;8:e79241 [PMC free article: PMC3818425] [PubMed: 24223913]
  • Müller-Vahl KR, Berding G, Emrich HM, Peschel T. Chorea-acanthocytosis in monozygotic twins: clinical findings and neuropathological changes as detected by diffusion tensor imaging, FDG-PET and (123)I-beta-CIT-SPECT. J Neurol. 2007;254:1081-8 [PubMed: 17294064]
  • Muñoz-Braceras S, Tornero-Écija AR, Vincent O, Escalante R. VPS13A is closely associated with mitochondria and is required for efficient lysosomal degradation. Dis Model Mech. 2019;12:dmm036681. [PMC free article: PMC6398486] [PubMed: 30709847]
  • Nasreddine ZS, Phillips NA, Bédirian V, Charbonneau S, Whitehead V, Collin I, Cummings JL, Chertkow H. The Montreal Cognitive Assessment, MoCA: a brief screening tool for mild cognitive impairment. J Am Geriatr Soc. 2005;53:695-9. [PubMed: 15817019]
  • Neuman SD, Levine TP, Bashirullah A. A novel superfamily of bridge-like lipid transfer proteins. Trends Cell Biol. 2022;32:962-74 [PMC free article: PMC9588498] [PubMed: 35491307]
  • Neutel D, Miltenberger-Miltenyi G, Silva I, de Carvalho M. Chorea-acanthocytosis presenting as motor neuron disease. Muscle Nerve. 2012;45:293-5 [PubMed: 22246890]
  • Niemelä V, Salih A, Solea D, Lindvall B, Weinberg J, Miltenberger G, Granberg T, Tzovla A, Nordin L, Danfors T, Savitcheva I, Dahl N, Paucar M. Phenotypic variability in chorea-acanthocytosis associated with novel VPS13A mutations. Neurol Genet. 2020;6:e426 [PMC free article: PMC7217656] [PubMed: 32494755]
  • Nishida Y, Nakamura M, Urata Y, Kasamo K, Hiwatashi H, Yokoyama I, Mizobuchi M, Sakurai K, Osaki Y, Morita Y, Watanabe M, Yoshida K, Yamane K, Miyakoshi N, Okiyama R, Ueda T, Wakasugi N, Saitoh Y, Sakamoto T, Takahashi Y, Shibano K, Tokuoka H, Hara A, Monma K, Ogata K, Kakuda K, Mochizuki H, Arai T, Araki M, Fujii T, Tsukita K, Sakamaki-Tsukita H, Sano A. Novel pathogenic VPS13A gene mutations in Japanese patients with chorea-acanthocytosis. Neurol Genet. 2019;5:e332 [PMC free article: PMC6515943] [PubMed: 31192303]
  • Park JS, Hu Y, Hollingsworth NM, Miltenberger-Miltenyi G, Neiman AM. Interaction between VPS13A and the XK scramblase is important for VPS13A function in humans. J Cell Sci. 2022;135:jcs260227 [PMC free article: PMC9482346] [PubMed: 35950506]
  • Park JS, Neiman AM. XK is a partner for VPS13A: a molecular link between chorea-acanthocytosis and McLeod syndrome. Mol Biol Cell. 2020;31:2425-36. [PMC free article: PMC7851852] [PubMed: 32845802]
  • Park JS, Thorsness MK, Policastro R, McGoldrick LL, Hollingsworth NM, Thorsness PE, Neiman AM. Yeast Vps13 promotes mitochondrial function and is localized at membrane contact sites. Mol Biol Cell. 2016;27:2435. [PMC free article: PMC4966984] [PubMed: 27280386]
  • Paucar M, Lindestad PÅ, Walker RH, Svenningsson P. Teaching video neuroimages: feeding dystonia in chorea-acanthocytosis. Neurology. 2015;85:e143 [PMC free article: PMC4653107] [PubMed: 26553947]
  • Paucar M, Pajak A, Freyer C, Bergendal Å, Döry M, Laffita-Mesa JM, Stranneheim H, Lagerstedt-Robinson K, Savitcheva I, Walker RH, Wedell A, Wredenberg A, Svenningsson P. Chorea, psychosis, acanthocytosis, and prolonged survival associated with ELAC2 mutations. Neurology. 2018;91:710-12. [PMC free article: PMC6177277] [PubMed: 30217939]
  • Peikert K, Akgün K, Beste C, Ziemssen T, Buhmann C, Danek A, Hermann A. Neurofilament light chain in serum is significantly increased in chorea-acanthocytosis. Parkinsonism Relat Disord. 2020;80:28-31 [PubMed: 32932025]
  • Peikert K, Danek A. VPS13 forum proceedings: XK, XK-related and VPS13 proteins in membrane lipid dynamics. Contact. 2023;6:25152564231156994. [PMC free article: PMC10243564] [PubMed: 37366410]
  • Peikert K, Federti E, Matte A, Constantin G, Pietronigro EC, Fabene PF, Defilippi P, Turco E, Del Gallo F, Pucci P, Amoresano A, Illiano A, Cozzolino F, Monti M, Garello F, Terreno E, Alper SL, Glaß H, Pelzl L, Akgün K, Ziemssen T, Ordemann R, Lang F, Brunati AM, Tibaldi E, Andolfo I, Iolascon A, Bertini G, Buffelli M, Zancanaro C, Lorenzetto E, Siciliano A, Bonifacio M, Danek A, Walker RH, Hermann A, De Franceschi L. Therapeutic targeting of Lyn kinase to treat chorea-acanthocytosis. Acta Neuropathol Commun. 2021b;9:81 [PMC free article: PMC8091687] [PubMed: 33941276]
  • Peikert K, Glass H, Federti E, Matte A, Pelzl L, Akgun K, Ziemssen T, Ordemann R, Lang F, De Franceschi L, Hermann A, et al. Targeting Lyn kinase in chorea acanthocytosis: a translational treatment approach in a rare disease. J Pers Med. 2021a;11:392. [PMC free article: PMC8150322] [PubMed: 34068769]
  • Peikert K, Hermann A, Danek A. XK-associated McLeod syndrome: nonhematological manifestations and relation to VPS13A disease. Transfus Med Hemother. 2022a;49:4-12. [PMC free article: PMC8832239] [PubMed: 35221863]
  • Peikert K, Storch A, Hermann A, Landwehrmeyer GB, Walker RH, Simionato G, Kaestner L, Danek A. Commentary: acanthocytes identified in Huntington's disease. Front Neurosci. 2022b;16:1049676 [PMC free article: PMC9673475] [PubMed: 36408380]
  • Pelzl L, Elsir B, Sahu I, Bissinger R, Singh Y, Sukkar B, Honisch S, Schoels L, Jemaà M, Lang E, Storch A, Hermann A, Stournaras C, Lang F. Lithium sensitivity of store operated Ca2+ entry and survival of fibroblasts isolated from chorea-acanthocytosis patients. Cell Physiol Biochem. 2017a;42:2066-77. [PubMed: 28803243]
  • Pelzl L, Hauser S, Elsir B, Sukkar B, Sahu I, Singh Y, Höflinger P, Bissinger R, Jemaà M, Stournaras C, Schöls L, Lang F. Lithium sensitive ORAI1 expression, store operated Ca2+ entry and suicidal death of neurons in chorea-acanthocytosis. Sci Rep. 2017b;7:6457 [PMC free article: PMC5526875] [PubMed: 28743945]
  • Quick S, Heidrich FM, Winkler MV, Winkler AH, Ibrahim K, Linke A, Speiser U, Grabmaier U, Buhmann C, Marxreiter F, Saft C, Danek A, Hermann A, Peikert K. Cardiac manifestation is evident in chorea-acanthocytosis but different from McLeod syndrome. Parkinsonism Relat Disord. 2021;88:90–5. [PubMed: 34153885]
  • Rampoldi L, Danek A, Monaco AP. Clinical features and molecular bases of neuroacanthocytosis. J Mol Med. 2002;80:475-91 [PubMed: 12185448]
  • Rampoldi L, Dobson-Stone C, Rubio JP, Danek A, Chalmers RM, Wood NW, Verellen C, Ferrer X, Malandrini A, Fabrizi GM, Brown R, Vance J, Pericak-Vance M, Rudolf G, Carre S, Alonso E, Manfredi M, Nemeth AH, Monaco AP. A conserved sorting-associated protein is mutant in chorea-acanthocytosis. Nat Genet. 2001;28:119-20 [PubMed: 11381253]
  • Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Grody WW, Hegde M, Lyon E, Spector E, Voelkerding K, Rehm HL, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17:405-24. [PMC free article: PMC4544753] [PubMed: 25741868]
  • Ryoden Y, Segawaa K, Nagata S. Requirement of Xk and Vps13a for the P2X7-mediated phospholipid scrambling and cell lysis in mouse T cells. Proc Natl Acad Sci USA. 2022;119:e2119286119 [PMC free article: PMC8851519] [PubMed: 35140185]
  • Saiki S, Hirose G, Sakai K, Matsunari I, Higashi K, Saiki M, Kataoka S, Hori A, Shimazaki K. Chorea-acanthocytosis associated with Tourettism. Mov Disord. 2004;19:833-6 [PubMed: 15254946]
  • Scheid R, Bader B, Ott DV, Merkenschlager A, Danek A. Development of mesial temporal lobe epilepsy in chorea-acanthocytosis. Neurology. 2009;73:1419-22 [PubMed: 19858465]
  • Schneider SA, Aggarwal A, Bhatt M, Dupont E, Tisch S, Limousin P, Lee P, Quinn N, Bhatia KP. Severe tongue protrusion dystonia: clinical syndromes and possible treatment. Neurology. 2006;67:940-3. [PubMed: 17000958]
  • Schneider SA, Lang AE, Moro E, Bader B, Danek A, Bhatia KP. Characteristic head drops and axial extension in advanced chorea-acanthocytosis. Mov Disord. 2010;25:1487-91. [PubMed: 20544815]
  • Seong E, Insolera R, Dulovic M, Kamsteeg EJ, Trinh J, Brüggemann N, Sandford E, Li S, Ozel AB, Li JZ, Jewett T, Kievit AJA, Münchau A, Shakkottai V, Klein C, Collins CA, Lohmann K, van de Warrenburg BP, Burmeister M. Mutations in VPS13D lead to a new recessive ataxia with spasticity and mitochondrial defects. Ann Neurol. 2018;83:1075-88. [PMC free article: PMC6105379] [PubMed: 29604224]
  • Sharma C, Nath K, Acharya M, Kumawat BL, Khandelwal D, Jain D. Cerebellar atrophy in neuroacanthocytosis. BMJ Case Rep. 2014:bcr2014205232. [PMC free article: PMC4054104] [PubMed: 24907220]
  • Sibon I, Ghorayeb I, Arne P, Tison F. Distressing belching and neuroacanthocytosis. Mov Disord. 2004;19:856-9 [PubMed: 15254955]
  • Simionato G, Hinkelmann K, Chachanidze R, Bianchi P, Fermo E, van Wijk R, Leonetti M, Wagner C, Kaestner L, Quint S. Red blood cell phenotyping from 3D confocal images using artificial neural networks. PLoS Comput Biol. 2021;17:e1008934. [PMC free article: PMC8118337] [PubMed: 33983926]
  • Sokolov E, Schneider SA, Bain PG. Chorea-acanthocytosis. Pract Neurol. 2012;12:40-3 [PubMed: 22258171]
  • Sorrentino G, De Renzo A, Miniello S, Nori O, Bonavita V. Late appearance of acanthocytes during the course of chorea-acanthocytosis. J Neurol Sci. 1999;163:175-8 [PubMed: 10371080]
  • Spieler D, Velayos-Baeza A, Mühlbäck A, Castrop F, Maegerlein C, Slotta-Huspenina J, Bader B, Haslinger B, Danek A. Identification of two compound heterozygous VPS13A large deletions in chorea-acanthocytosis only by protein and quantitative DNA analysis. Mol Genet Genomic Med. 2020;8:e1179. [PMC free article: PMC7507471] [PubMed: 32056394]
  • Stanslowsky N, Reinhardt P, Glass H, Kalmbach N, Naujock M, Hensel N, Lübben V, Pal A, Venneri A, Lupo F, De Franceschi L, Claus P, Sterneckert J, Storch A, Hermann A, Wegner F. Neuronal dysfunction in iPSC-derived medium spiny neurons from chorea-acanthocytosis patients is reversed by src kinase inhibition and F-actin stabilization. J Neurosci. 2016;36:12027 [PMC free article: PMC6604921] [PubMed: 27881786]
  • Stenson PD, Mort M, Ball EV, Chapman M, Evans K, Azevedo L, Hayden M, Heywood S, Millar DS, Phillips AD, Cooper DN. The Human Gene Mutation Database (HGMD®): optimizing its use in a clinical diagnostic or research setting. Hum Genet. 2020;139:1197-207. [PMC free article: PMC7497289] [PubMed: 32596782]
  • Storch A, Kornhass M, Schwarz J. Testing for acanthocytosis A prospective reader-blinded study in movement disorder patients. J Neurol. 2005;252:84-90 [PubMed: 15654559]
  • Suzuki J, Imanishi E, Nagata S. Exposure of phosphatidylserine by Xk-related protein family members during apoptosis. J Biol Chem. 2014;289:30257-67. [PMC free article: PMC4215210] [PubMed: 25231987]
  • Tamura Y, Matsui K, Yaguchi H, Hashimoto M, Inoue K. Nemaline rods in chorea-acanthocytosis. Muscle Nerve. 2005;31:516-9 [PubMed: 15660376]
  • Termsarasab P, Frucht SJ. The "stutter-step": a peculiar gait feature in advanced Huntington's disease and chorea-acanthocytosis. Mov Disord Clin Pract. 2018;5:223-4. [PMC free article: PMC6336370] [PubMed: 30746406]
  • Thomas M, Jankovic J. Neuroacanthocytosis. In: Noseworthy J, ed. Neurological Therapeutics: Principles and Practice. Abingdon, UK: CRC Press. 2003;2882-9.
  • Tomemori Y, Ichiba M, Kusumoto A, Mizuno E, Sato D, Muroya S, Nakamura M, Kawaguchi H, Yoshida H, Ueno S, Nakao K, Nakamura K, Aiba A, Katsuki M, Sano A. A gene-targeted mouse model for chorea-acanthocytosis. J Neurochem 2005;92:759-66 [PubMed: 15686477]
  • Tomiyasu A, Nakamura M, Ichiba M, Ueno S, Saiki S, Morimoto M, Kobal J, Kageyama Y, Inui T, Wakabayashi K, Yamada T, Kanemori Y, Jung HH, Tanaka H, Orimo S, Afawi Z, Blatt I, Aasly J, Ujike H, Babovic-Vuksanovic D, Josephs KA, Tohge R, Rodrigues GR, Dupré N, Yamada H, Yokochi F, Kotschet K, Takei T, Rudzińska M, Szczudlik A, Penco S, Fujiwara M, Tojo K, Sano A. Novel pathogenic mutations and copy number variations in the VPS13A gene in patients with chorea-acanthocytosis. Am J Med Genet B Neuropsychiatr Genet. 2011;156B:620-31. [PubMed: 21598378]
  • Tsai CH, Chen RS, Chang HC., Lu CS, Liao KK. Acanthocytosis and spinocerebellar degeneration: a new association? Mov Disord. 1997;12:456–9 [PubMed: 9159749]
  • Ueno S, Maruki Y, Nakamura M, Tomemori Y, Kamae K, Tanabe H, Yamashita Y, Matsuda S, Kaneko S, Sano A. The gene encoding a newly discovered protein, chorein, is mutated in chorea-acanthocytosis. Nat Genet 2001;28:121-2 [PubMed: 11381254]
  • Urata Y, Nakamura M, Sasaki N, Shiokawa N, Nishida Y, Arai K, Hiwatashi H, Yokoyama I, Narumi S, Terayama Y, Murakami T, Ugawa Y, Sakamoto H, Kaneko S, Nakazawa Y, Yamasaki R, Sadashima S, Sakai T, Arai H, Sano A. Novel pathogenic XK mutations in McLeod syndrome and interaction between XK protein and chorein. Neurol Genet. 2019;5:e328. [PMC free article: PMC6481271] [PubMed: 31086825]
  • Vaisfeld A, Bruno G, Petracca M, Bentivoglio AR, Servidei S, Vita MG. Neuroacanthocytosis syndromes in an Italian cohort: clinical spectrum, high genetic variability and muscle involvement. Genes (Basel). 2021;12:344 [PMC free article: PMC7996727] [PubMed: 33652783]
  • Velayos-Baeza A, Holinski-Feder E, Neitzel B, Bader B, Critchley EM, Monaco AP, Danek A, Walker RH. Chorea-acanthocytosis genotype in the original Critchley Kentucky neuroacanthocytosis kindred. Arch Neurol 2011;68:1330-3 [PMC free article: PMC4615612] [PubMed: 21987550]
  • Velayos-Baeza A, Vettori A, Copley RR, Dobson-Stone C, Monaco AP. Analysis of the human VPS13 gene family. Genomics. 2004;84:536-49 [PubMed: 15498460]
  • Walker RH. Management of neuroacanthocytosis syndromes. Tremor Other Hyperkinet Mov (N Y). 2015;5:346. [PMC free article: PMC4613733] [PubMed: 26504667]
  • Walker RH, Danek A. "Neuroacanthocytosis" - overdue for a taxonomic update. Tremor Other Hyperkinet Mov (N Y) 2021;11:1. [PMC free article: PMC7805383] [PubMed: 33510935]
  • Walker RH, Liu Q, Ichiba M, Muroya S, Nakamura M, Sano A, Kennedy CA, Sclar G. Self-mutilation in chorea-acanthocytosis: Manifestation of movement disorder or psychopathology? Mov Disord. 2006;21:2268-9. [PubMed: 17044067]
  • Walker RH, Miranda M, Jung HH, Danek A. Life expectancy and mortality in chorea-acanthocytosis and McLeod syndrome. Parkinsonism Relat Disord. 2019;60:158–61 [PubMed: 30245172]
  • Walker RH, Velayos-Baeza A, Bader B, Danek A, Saiki S. Mutation in the CHAC gene in a family of autosomal dominant chorea-acanthocytosis. Neurology. 2012b;79:198-9 [PubMed: 22778235]
  • Walterfang M, Evans A, Looi JC, Jung HH, Danek A, Walker RH, Velakoulis D. The neuropsychiatry of neuroacanthocytosis syndromes. Neurosci Biobehav Rev. 2011a;35:1275-83. [PubMed: 21237198]
  • Walterfang M, Looi JC, Styner M, Walker RH, Danek A, Niethammer M, Evans A, Kotschet K, Rodrigues GR, Hughes A, Velakoulis D. Shape alterations in the striatum in chorea-acanthocytosis. Psychiatry Res. 2011b;192:29-36 [PMC free article: PMC3324182] [PubMed: 21377843]
  • Walterfang M, Yucel M, Walker R, Evans A, Bader B, Ng A, Danek A, Mocellin R, Velakoulis D. Adolescent obsessive compulsive disorder heralding chorea-acanthocytosis. Mov Disord. 2008;23:422-5 [PubMed: 18058950]
  • Wihl G, Volkmann J, Allert N, Lehrke R, Sturm V, Freund HJ. Deep brain stimulation of the internal pallidum did not improve chorea in a patient with neuro-acanthocytosis. Mov Disord. 2001;16:572-5. [PubMed: 11391763]
  • Yamamoto T, Hirose G, Shimazaki K, Takado S, Kosoegawa H, Saeki M. Movement disorders of familial neuroacanthocytosis syndrome. Arch Neurol. 1982;39:298-301 [PubMed: 7073550]
  • Yeshaw WM, Zwaag M van der, Pinto F, Lahaye LL, Faber AI, Gómez-Sánchez R, Dolga AM, Poland C, Monaco AP, IJzendoorn SC van, Grzeschik NA, Velayos-Baeza A, Sibon OC. Human VPS13A is associated with multiple organelles and influences mitochondrial morphology and lipid droplet motility. eLife. 2019;8: 728. [PMC free article: PMC6389287] [PubMed: 30741634]
  • Zayas LE, Walker RH. Amantadine treatment for hyperkinetic movements in chorea-acanthocytosis. Mov Disord Clin Pract. 2022;10:346-7 [PMC free article: PMC9941915] [PubMed: 36825061]
Copyright © 1993-2024, University of Washington, Seattle. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.

GeneReviews® chapters are owned by the University of Washington. Permission is hereby granted to reproduce, distribute, and translate copies of content materials for noncommercial research purposes only, provided that (i) credit for source (http://www.genereviews.org/) and copyright (© 1993-2024 University of Washington) are included with each copy; (ii) a link to the original material is provided whenever the material is published elsewhere on the Web; and (iii) reproducers, distributors, and/or translators comply with the GeneReviews® Copyright Notice and Usage Disclaimer. No further modifications are allowed. For clarity, excerpts of GeneReviews chapters for use in lab reports and clinic notes are a permitted use.

For more information, see the GeneReviews® Copyright Notice and Usage Disclaimer.

For questions regarding permissions or whether a specified use is allowed, contact: ude.wu@tssamda.

Bookshelf ID: NBK1387PMID: 20301561

Views

Tests in GTR by Gene

Related information

  • MedGen
    Related information in MedGen
  • OMIM
    Related OMIM records
  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed
  • Gene
    Locus Links

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...