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

PINK1 Type of Young-Onset Parkinson Disease

Synonym: PARK-PINK1

, MD and , MD.

Author Information and Affiliations

Initial Posting: ; Last Update: April 25, 2024.

Estimated reading time: 31 minutes

Summary

Clinical characteristics.

PINK1 type of young-onset Parkinson disease is characterized by early onset (median age at onset 32 years) of tremor, bradykinesia, and rigidity that are often indistinguishable from other causes of Parkinson disease. Lower-limb dystonia may be a presenting sign. Postural instability, hyperreflexia, abnormal behavior, and psychiatric manifestations have been described. The disease is usually slowly progressive. Individuals have a marked and sustained response to oral administration of levodopa (L-dopa), frequently associated with L-dopa-induced fluctuations and dyskinesias.

Diagnosis/testing.

The diagnosis of PINK1 type of young-onset Parkinson disease is established by the identification of biallelic PINK1 pathogenic variants on molecular genetic testing.

Management.

Treatment of manifestations: PINK1 type of young-onset Parkinson disease usually responds well to L-dopa and/or other dopamine agonists, which may be used in combination with catechol-O-methyltransferase inhibitors or monoamine oxidase-B inhibitors, anticholinergics, and amantadine. Physical therapy and/or occupational therapy to improve and/or maintain gross motor and fine motor skills as well as speech therapy. Invasive therapies include intrajejunal L-dopa-carbidopa pump, subcutaneous apomorphine pump, or deep brain stimulation. L-dopa-induced dyskinesias can be treated by reducing L-dopa dose, switching to dopamine receptor agonists, deep brain stimulation, or continuous treatment with L-dopa or apomorphine. Atypical neuroleptic agents can be used for neuropsychiatric manifestations; standard treatments for depression. Cholinesterase inhibitors can be used to treat dementia. Consider droxidopa, midodrine, fludrocortisone, and/or supportive measures for orthostasis. Symptomatic treatment for constipation.

Surveillance: Neurologic evaluation every three to 12 months to assess motor and non-motor manifestations and treatment efficacy; in addition, assess for atypical manifestations and therapy needs at each visit. Neuropsychiatric evaluation in those with mood disorder / psychotic symptoms or as needed. Cognitive assessment annually or as needed. At each visit, assess nutrition and safety of feeding; assess for symptoms of orthostasis and measure supine and standing blood pressure and pulse; assess for constipation, urinary urgency, or urge incontinence; and assess family needs.

Agents/circumstances to avoid: Neuroleptic treatment may exacerbate parkinsonism.

Genetic counseling.

PINK1 type of young-onset Parkinson disease is inherited in an autosomal recessive manner. If both parents are known to be heterozygous for a PINK1 pathogenic variant, each sib of an affected individual has at conception a 25% chance of being affected, a 50% chance of being heterozygous, and a 25% chance of inheriting neither of the familial PINK1 pathogenic variants. Once the PINK1 pathogenic variants have been identified in an affected family member, heterozygote testing for at-risk relatives and prenatal and preimplantation genetic testing are possible.

Diagnosis

Updated guidelines on the molecular diagnosis of Parkinson disease were provided in a joint effort by the European Federation of Neurological Societies (EFNS), the European Section of the International Parkinson and Movement Disorders Society (MDS-ES), and the European Neurological Society (ENS) [Berardelli et al 2013]. Some national societies have also published guidelines on the molecular diagnosis of Parkinson disease, such as the German Society for Neurology.

In addition, new diagnostic criteria using a biological classification referred to as SynNeurGe have been published [Höglinger et al 2024]. This classification has three components: (1) the presence or absence of pathologic alpha-synuclein (Syn) in tissues or cerebrospinal fluid (CSF), (2) evidence of underlying neurodegeneration defined by neuroimaging (Neur), and (3) identification of pathogenic variants that cause or strongly predispose to Parkinson disease (Ge).

Suggestive Findings

PINK1 type of young-onset Parkinson disease should be suspected in individuals with the following clinical and family history findings.

Clinical findings

  • Early onset. Onset is age <40 years in 57%; late onset (27%) and juvenile onset (16%) can also occur.
  • Parkinsonism (bradykinesia, resting tremor, rigidity)
  • Dyskinesia
  • Motor fluctuations
  • Dystonia
  • Cognitive decline
  • Psychiatric manifestations
  • Good response to levodopa (L-dopa) treatment (in 98% of individuals with a reported L-dopa response)

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.

Establishing the Diagnosis

The diagnosis of PINK1 type of young-onset Parkinson disease is established in a proband with suggestive findings and biallelic pathogenic (or likely pathogenic) variants in PINK1 identified by molecular genetic testing (see Table 1).

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 PINK1 variants of uncertain significance (or of one known PINK1 pathogenic variant and one PINK1 variant of uncertain significance) does not establish or rule out the diagnosis.

Because the phenotype of PINK1 type of young-onset Parkinson disease is indistinguishable from many other inherited causes of Parkinson disease, recommended molecular genetic testing approaches include the use of a multigene panel or comprehensive genomic testing (exome sequencing, genome sequencing). A multigene panel requires that the clinician determine which genes are likely involved (see Option 1), whereas comprehensive genomic testing does not (see Option 2).

Note: (1) PINK1 single-exon and multiexon deletions/duplications have been identified in individuals with PINK1 type of young-onset Parkinson disease; molecular genetic testing should include deletion/duplication analysis (see Table 1). (2) Single-gene testing (sequence analysis of PINK1, followed by gene-targeted deletion/duplication analysis) is rarely useful and typically NOT recommended.

Option 1

A multigene panel that includes PINK1, PRKN, PARK7, and other genes of interest (see Differential Diagnosis) is most likely to identify the genetic cause of the condition while limiting the 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

When the diagnosis of PINK1 type of young-onset Parkinson disease is not considered because an individual has atypical phenotypic features, comprehensive genomic testing does not require the clinician to determine which genes are likely involved. Exome sequencing is most commonly used, genome sequencing can also be considered.

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

Table 1.

Molecular Genetic Testing Used in PINK1 Type of Young-Onset Parkinson Disease

Gene 1MethodProportion of Pathogenic Variants 2 Identified by Method
PINK1 Sequence analysis 3>88% 4
Gene-targeted deletion/duplication analysis 5<12% 4
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 missense, nonsense, and splice site variants and small intragenic deletions/insertions; typically, exon or whole-gene deletions/duplications are not detected. For issues to consider in the interpretation of sequence analysis results, click here.

4.

Data derived from the subscription-based professional view of the 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. Exome and genome sequencing may be able to detect deletions/duplications using breakpoint detection or read depth; however, sensitivity can be lower than gene-targeted deletion/duplication analysis.

Clinical Characteristics

Clinical Description

PINK1 type of young-onset Parkinson disease is characterized by typically early-onset Parkinson disease that is often clinically indistinguishable from other genetic causes of Parkinson disease or idiopathic Parkinson disease. Typical features include bradykinesia, rigidity, dyskinesia, motor fluctuations, and dystonia. Some individuals have cognitive decline and psychiatric manifestations. The following information is based on a systematic review of published reports including 205 individuals with PINK1 type of young-onset Parkinson disease from 136 families (see www.mdsgene.org and references therein).

Table 2.

PINK1 Type of Young-Onset Parkinson Disease: Frequency of Select Features

Feature% of Persons w/Feature 1Comment
Bradykinesia 95%
Rigidity 90%
Resting tremor 78%
Dyskinesia 67%
Motor fluctuations 79%
Dystonia 48%Often of the lower limbs
Cognitive decline 29%Including mild cognitive impairment & dementia
Autonomic dysfunction 48%Most commonly including urinary urgency or urge incontinence & orthostatic hypotension
Psychiatric manifestations 33%-56%Depression in 51%, anxiety in 56%, psychotic symptoms in 33%
1.

Data is based on 205 reported individuals identified through the MDSGene PINK1 Review, previously published by Kasten et al [2018] and updated on the MDSGene website in February 2024 (from www​.mdsgene.org; last accessed 2-29-24). Note: Clinical information was often incomplete (e.g., missing data ranged from 7%-79% for the items reported above). Numbers displayed indicate the percentages of individuals with the respective clinical feature out of all individuals for whom this feature was reported (valid percentages); missing data was not taken into account for calculation.

Onset. The majority of individuals (57%) have early onset (age at onset <40 years), 27% have late onset, and 16% have juvenile onset (age at onset <21 years). Note: The age of onset was not specified in 10% of individuals. The median age of onset is 32 years (interquartile range: age 24-40 years; range: age 9-67 years) (see www.mdsgene.org).

Parkinsonian features are usually asymmetric; in some individuals the manifestations at onset are symmetric. PINK1 type of Parkinson disease clinically resembles other monogenic causes of Parkinson disease, especially those with recessive inheritance, and Parkinson disease of unknown cause. Individuals frequently present with all the typical features of Parkinson disease, including bradykinesia, rigidity, tremor, and postural instability. Tremor and bradykinesia are the most common presenting signs. The disease is slowly progressive. Rigidity and postural instability (60%) frequently occur with disease progression. Sleep benefit was reported for 40% of individuals (see www.mdsgene.org). A clinical presentation resembling atypical parkinsonism is very rare.

Non-motor Parkinson features are frequently reported (92%) in individuals with PINK1 type of young-onset Parkinson disease and commonly include psychiatric involvement, cognitive impairment, and autonomic dysfunction.

Psychiatric involvement. Abnormal behavior and/or psychiatric manifestations – in particular, depression (51%), anxiety (56%), and psychotic symptoms (33%) (see www.mdsgene.org) – can occur in affected individuals. Related sleep impairment (e.g., falling and staying asleep) is also common in individuals with PINK1 type of young-onset Parkinson disease [Ricciardi et al 2014].

Cognitive decline, ranging from mild cognitive impairment to dementia, has been reported for a subset of individuals.

Information on the occurrence of hyposmia is scarce (www.mdsgene.org; only available for 24 individuals in total and present in ten).

Autonomic dysfunction was reported for 48% of individuals (see www.mdsgene.org) and most commonly include urinary urgency, urge incontinence, constipation, and orthostatic hypotension.

Other additional neurologic manifestations. Dystonia (often of the lower limbs) and hyperreflexia may also be present or develop as the disease progresses [Bonifati et al 2005]. Dystonia has also been reported as the initial sign in a subset of individuals (18% of individuals with available data).

Neuroimaging. CT and MRI neuroimaging of individuals with PINK1 type of young-onset Parkinson disease is usually normal. Imaging of dopamine function (using DaTscan) generally demonstrated relatively symmetric loss of radioligand uptake in the striatum, similar to the pattern seen in the Parkin type of young-onset Parkinson disease, LRRK2 (Gly2019Ser) type of Parkinson disease, and SNCA type of Parkinson disease [McNeill et al 2013].

Response to treatment. In general, the vast majority of individuals with PINK1 type of young-onset Parkinson disease have a good response to L-dopa therapy [Over et al 2021]; it was suggested to be even better than in Parkinson disease of unknown cause [Valente & Ferraris 2010]. Usually, this positive response persists over time, although no systematic longitudinal assessment is available. L-dopa primarily addresses motor features of the disease; additional medication might be needed to address non-motor features, especially psychiatric involvement. Side effects of L-dopa are common and most frequently include L-dopa-induced dyskinesias; motor fluctuations are also reported, and rarely dystonia. Response to non-L-dopa therapies including dopamine agonists, catechol-O-methyltransferase (COMT) inhibitors, monoamine oxidase-B (MAO-B) inhibitors, anticholinergics, and/or amantadine is also good, but information is limited [Over et al 2021]. Surgical therapies, including deep brain stimulation and thalamotomy, have only been reported in a small number of individuals with usually a moderate-to-good response; however, the sample size is too small to draw meaningful conclusions.

Prognosis. Progression is slower compared to Parkinson disease of unknown cause [Kasten et al 2018]. However, detailed and systematic data on disease progression is limited [Kasten et al 2018]. Large, multicenter studies with longitudinal and systematic data collection are needed. In general, the expected life span in individuals with PINK1 type of young-onset Parkinson disease is similar to Parkinson disease of unknown cause. Common causes of an earlier death include complications related to Parkinson disease (e.g., pneumonia due to severe dysphagia and aspiration, falls resulting in serious injuries due to gait difficulties, or immobility).

Heterozygotes

The evidence of heterozygous PINK1 variants causing or acting as a risk factor for Parkinson disease remains controversial. Individuals with a heterozygous PINK1 pathogenic variant usually remain asymptomatic but may show subtle subclinical alterations (e.g., a latent nigrostriatal dopaminergic deficit on functional imaging, premotor-motor excitability changes detected by transcranial magnetic stimulation, reduced arm swing, hyposmia, and/or diminished color discrimination) [Eggers et al 2010, Nürnberger et al 2015, Weissbach et al 2017]. In asymptomatic heterozygotes, voxel-based morphometry revealed an increase of putaminal and pallidal gray matter volume, findings similar to those in the Parkin type of young-onset Parkinson disease [Binkofski et al 2007, Reetz et al 2010].

Several individuals with Parkinson disease and a heterozygous PINK1 pathogenic variant have been identified, including individuals with young-onset Parkinson disease [Bonifati et al 2005, Abou-Sleiman et al 2006, Hayashida et al 2021]. These studies suggest that age at onset might differ between individuals with biallelic and heterozygous PINK1 pathogenic variants, with the age at onset of biallelic individuals being slightly younger. The clinical phenotype was similar in both groups, except some features (e.g., dystonia, hyperreflexia, and sleep benefit) seemed to be less common in heterozygous individuals. In one case-control study, heterozygous PINK1 variant p.Gly411Ser was significantly associated with a markedly increased risk for Parkinson disease (odds ratio = 2.92, P=0.032), a finding that was supported by functional analyses [Puschmann et al 2017]. However, the results of another more recent large-scale meta-analysis suggested that PINK1 variant p.Gly411Ser is likely benign, that other PINK1 heterozygous pathogenic variants are not likely associated with an increased risk for Parkinson disease, and that heterozygosity for a PINK1 pathogenic variant is not a "robust risk factor" for Parkinson disease [Krohn et al 2020].

Genotype-Phenotype Correlations

No correlation between the type of variant and age at onset, clinical presentation, or disease progression has yet been observed.

Modifiers. Recently, it has been shown that somatic mitochondrial variant load is a disease-onset modifier for PINK1 (and Parkin) type of young-onset Parkinson disease. By investigating mitochondrial DNA (mtDNA) integrity in individuals with biallelic (n=84) and heterozygous (n=170) PINK1 or PRKN pathogenic variants compared to individuals with Parkinson disease of unknown cause (n=67) and controls (n=90), it was shown that affected and unaffected individuals with a heterozygous PINK1 or PRKN pathogenic variant can be distinguished by heteroplasmic mtDNA variant load; and individuals with biallelic PINK1 or PRKN pathogenic variants contain more heteroplasmic mtDNA variants in blood than individuals with a heterozygous PINK1 or PRKN variant [Trinh et al 2023].

Further, one individual with biallelic PINK1 pathogenic variants and homoplasmy for pathogenic variants in two mitochondrial genes encoding subunits of complex I (MT-ND5 and MT-ND6) was reported with very early-onset Parkinson disease [Piccoli et al 2008]; thus, it was hypothesized that the combination of pathogenic variants in MT-ND5 and MT-ND6 accelerated the disease onset.

Penetrance

Biallelic PINK1 pathogenic variants are considered fully penetrant [Höglinger et al 2024]. Penetrance of heterozygous variants is under debate (see Heterozygotes).

Nomenclature

Based on the International Parkinson and Movement Disorder Society Task Force for Nomenclature of Genetic Movement Disorders, the recommended name for Parkinson disease caused by PINK1 pathogenic variants is "PARK-PINK1" [Marras et al 2016, Lange et al 2022].

Prevalence

The prevalence is not known. PINK1 pathogenic variants are a rare cause of early-onset Parkinson disease but are the second most common form of autosomal recessive Parkinson disease [Jia et al 2022].

The proportion of women among individuals with PINK1 type of young-onset Parkinson disease is 54%. The majority of families are of Asian (36%), White/European (31%), or mixed (21%) ancestry; reports of individuals of Arab, African, or Indian ancestry are rare (see www.mdsgene.org).

Differential Diagnosis

Clinically, PINK1 type of young-onset Parkinson disease and idiopathic Parkinson disease are difficult to differentiate (see Parkinson Disease Overview). More than 80% of individuals with Parkinson disease have no family history of the disorder. A monogenic cause of Parkinson disease can be identified in some individuals with a positive family history or a young age at disease onset.

Early-onset autosomal recessive Parkinson disease. PINK1 type of young-onset Parkinson disease and early-onset Parkinson disease of other etiologies (see Table 3) are difficult to distinguish by clinical examination.

Table 3.

Genes Associated with Early-Onset Autosomal Recessive Parkinson Disease in the Differential Diagnosis of PINK1 Type of Young-Onset Parkinson Disease

GenePD Designation 1Median Age at Onset
(Range) 2
Selected Features
PRKN Parkin type of early-onset Parkinson disease (PARK-Parkin)31 yrs
(3-81)
  • Most common cause of EOPD
  • PARK-PINK1 & PARK-Parkin are clinically indistinguishable.
PARK7 (DJ1) PARK-PARK7 (OMIM 606324)27 yrs
(15-40)
  • Phenotype similar to PARK-Parkin
  • IDD &/or seizures occasionally
DNAJC6 PARK-DNAJC6 11 yrs
(7-42)
  • Pyramidal signs
  • IDD / early cognitive impairment
  • Early & vivid hallucinations on intake of dopamine agonists
  • Early falls
  • Saccadic abnormalities
  • Pyramidal signs
FBXO7 PARK-FBXO7 (OMIM 260300)17 yrs
(10-52)
  • IDD / early cognitive impairment
  • Early & vivid hallucinations & behavioral abnormalities on intake of dopamine agonists
  • Early falls
  • Saccadic abnormalities
  • Gaze palsy
  • Oculogyric spasms
  • Pyramidal signs
  • Autonomic dysfunction
SYNJ1 PARK-SYNJ1 (OMIM 615530)21 yrs
(12-31)
  • Early cognitive impairment
  • Early falls
  • Saccadic abnormalities
  • Gaze palsy
  • Pyramidal signs
  • Ataxia
  • Autonomic dysfunction
VPS13C PARK-VPS13C (OMIM 616840)29 yrs
(0-70)
  • Early cognitive impairment
  • Early falls
  • Pyramidal signs
  • Autonomic dysfunction

EOPD = early-onset Parkinson disease; IDD = intellectual developmental disorder

1.

Nomenclature based on Marras et al [2016] and Lange et al [2022].

2.

Data based on Kasten et al [2018], Wittke et al [2021], and the MDSGene website (www​.mdsgene.org).

Levodopa-responsive dystonia. For individuals with juvenile-onset Parkinson disease, especially those with prominent dystonia, L-dopa-responsive dystonia should be considered, including:

In general, the phenotype of tyrosine hydroxylase-deficient dopa-responsive dystonia and sepiapterin reductase-deficient dopa-responsive dystonia is much more severe.

Management

No clinical practice guidelines specifically for PINK1 type of young-onset Parkinson disease have been published. Some national societies (e.g., the German Society for Neurology) recommend treating PINK1 type of young-onset Parkinson disease similar to Parkinson disease of unknown cause. In the absence of internationally published guidelines, the following recommendations are based on the authors' personal experience managing individuals with this disorder.

Evaluations Following Initial Diagnosis

To establish the extent of disease and needs in an individual diagnosed with PINK1 type of young-onset Parkinson disease, the evaluations summarized in Table 4 (if not performed as part of the evaluation that led to the diagnosis) are recommended.

Table 4.

PINK1 Type of Young-Onset Parkinson Disease: Recommended Evaluations Following Initial Diagnosis

System/ConcernEvaluationComment
Neurologic Detailed neurologic exam focusing on presence & extent of movement disorderUsing the Unified Parkinson Disease Rating Scale 1
Assessment for presence/severity of atypical manifestations
Eval by PT, OT, & SLP if required
Autonomic Assessment for presence/severity of urinary urgency, urge incontinence, constipation, & orthostatic hypotension
Neuropsychiatric Formal neuropsychiatric assessment w/neuropsychiatrist
Cognition Formal cognitive testing
Sleep Assessment for presence/severity of sleep disturbances, e.g., difficulties falling asleep, staying asleep, or rapid eye movement sleep behavior disorder (RBD)Consider polysomnography for persons w/suspected RBD.
Smell Assessment for presence/severity of hyposmia
Nutrition/
Gastrointestinal
  • Nutritional eval by dietician to monitor & ensure adequate caloric intake
  • SLP assessment of safety of feeding
Genetic counseling By genetics professionals 2To inform affected persons & their families re nature, MOI, & implications of PINK1 type of young-onset Parkinson disease to facilitate medical & personal decision making
Family support
& resources
By clinicians, wider care team, & family support organizationsAssessment of family & social structure to determine need for:

MOI = mode of inheritance; OT = occupational therapist; PT = physical therapist; SLP = speech-language therapist

1.

Goetz et al2008]

2.

Medical geneticist, certified genetic counselor, certified advanced genetic nurse

Treatment of Manifestations

To date, the treatment of PINK1 type of young-onset Parkinson disease does not differ from that of Parkinson disease of other etiologies (e.g., other monogenic forms, Parkinson disease of unknown etiology), and no gene-specific guidelines or recommendations have been developed. Updated treatment guidelines for Parkinson disease have been published by national and international neurologic societies, such as the European or American Academy of Neurology (EAN or AAN), often in cooperation with the Movement Disorder Society (MDS).

To date, no therapy can slow or stop the progression of Parkinson disease; available treatment options are purely symptomatic. In general, optimal management should begin at diagnosis and involve a multidisciplinary team approach, including pharmacologic and non-pharmacologic interventions [Bloem et al 2021].

Table 5.

PINK1 Type of Young-Onset Parkinson Disease: Treatment of Manifestations

Manifestation/ConcernTreatmentConsiderations/Other
Neurologic Medication
Levodopa (L-dopa) in combination w/peripheral dopa decarboxylase inhibitor (carbidopa, benserazide):
1.

Immediate-release (IR) tablets

2.

Disintegrating tablets

3.

Controlled-release (CR) tablets

4.

Extended-release (ER) capsules

5.

Inhalation powder

Dopamine agonists:
1.

IR & CR tablets

2.

Subcutaneous injections (apomorphine)

3.

Transdermal patch (rotigotine)

Other drugs used in combination w/L-dopa & dopamine agonists: COMT inhibitors or MAO-B inhibitors, anticholinergics, & amantadine.
  • Response to L-dopa is usually significant & sustained for low doses even after long disease duration in persons w/PINK1 type of young-onset Parkinson disease.
  • The major problem is the early occurrence of severe L-dopa-induced dyskinesias (abnormal involuntary movements) & fluctuations. Fluctuations can be ↓ by a combination of dopamine therapies (e.g., dopamine agonists), adding COMT inhibitors, & keeping the doses of L-dopa as low as possible.
Supportive non-pharmacologic options
  • PT &/or OT to improve &/or maintain gross motor & fine motor skills.
  • Speech therapy
Invasive therapies
  • Intrajejunal L-dopa-carbidopa pump
  • Subcutaneous apomorphine pump
  • DBS (preferably STN-DBS)
  • Can be considered for advanced stages w/fluctuations not satisfactorily controlled w/oral medications. 1
  • STN-DBS improves motor symptoms & quality of life. 1
  • The use of DBS was reported to be successful in 4/5 persons w/PINK1 type of young-onset Parkinson disease. 2
Dyskinesias Treatment options:
  • Reduction of L-dopa dose
  • Dopamine receptor agonists
  • DBS
  • Continuous application of L-dopa or apomorphine (pump therapies)
Neuropsychiatric manifestations
  • Atypical neuroleptic agents such as low-dose clozapine, quetiapine, or pimavanserin & reduction of dopaminergic therapy can ↓ delusions & hallucinations.
  • Standard treatments for depression
Dementia Consider treatment w/cholinesterase inhibitor (rivastigmine).
Orthostasis Consider treatment w/droxidopa, midodrine, fludrocortisone.Consider supportive measures (e.g., compression stockings, abdominal belts).
Constipation Symptomatic treatmentConsider dietary adjustments before using medication.

COMT = catechol-O-methyltransferase; DBS = deep brain stimulation; MAO-B = monoamine oxidase-B; OT = occupational therapy; PT = physical therapy; STN = subthalamic nucleus

1.

Treatment recommendations on invasive therapies based on the recent EAS/MDS-ES guidelines [Deuschl et al 2022].

2.

According to the most recent review [Kuusimäki et al 2020]

Surveillance

Table 6.

PINK1 Type of Young-Onset Parkinson Disease: Recommended Surveillance

System/ConcernEvaluationFrequency
Neurologic Neurologic eval to assess motor & non-motor manifestations & treatment efficacyEvery 3-12 mos to modify treatment as needed
  • Assessment of presence/severity of atypical manifestations
  • Assessment of need for PT, OT, & speech therapy
At each visit
Psychiatric Neuropsychiatric evalIn those w/mood disorder or psychotic symptoms, or as needed
Cognition Cognitive assessmentAnnually or as needed
Nutrition/
Gastrointestinal
  • Nutritional eval by dietician to monitor & ensure adequate caloric intake
  • SLP assessment of safety of feeding
As needed
Autonomic dysfunction
  • Assess for symptoms of orthostasis.
  • Measure supine & standing blood pressure & pulse.
  • Assess for constipation, urinary urgency, or urge incontinence.
At each visit
Family support
& resources
Assess need for:
  • Community or online resources
  • Social work support;
  • Home nursing referral.

OT = occupational therapy; PT = physical therapy

Agents/Circumstances to Avoid

Neuroleptic treatment may exacerbate parkinsonism.

Evaluation of Relatives at Risk

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

Pregnancy Management

There is only a single case report of a woman with biallelic PINK1 pathogenic variants who became pregnant [Li et al 2021]. During pregnancy, symptoms neither worsened nor improved, and the individual continued to take very low doses of L-dopa. The pregnancy and birth were without complications.

In general, data on pregnancy in individuals with Parkinson disease is rare, since age of onset is often after the childbearing years. Historically, it was reported that parkinsonian symptoms may exacerbate during pregnancy, but this may be because antiparkinsonian medications were not recommended or were underdosed [Seier & Hiller 2017]. Reviewing data from case reports and drug registries, L-dopa seems to be the safest option in pregnancy. Amantadine should be avoided in women who are pregnant or trying to become pregnant [Seier & Hiller 2017]. Data on other pharmacologic and surgical treatments is limited. To date, there is no evidence that women with Parkinson disease have higher rates of birth or fetal complications [Seier & Hiller 2017].

See MotherToBaby for further information on medication use during pregnancy.

Therapies Under Investigation

Search ClinicalTrials.gov in the US and EU Clinical Trials Register in Europe for 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

PINK1 type of young-onset Parkinson disease is inherited in an autosomal recessive manner.

Risk to Family Members

Parents of a proband

  • The parents of an affected individual are presumed to be heterozygous for a PINK1 pathogenic variant.
  • Molecular genetic testing is recommended for the parents of a proband to confirm that both parents are heterozygous for a PINK1 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:
  • The risk to heterozygotes of developing symptoms is not yet determined; however, several individuals with parkinsonism who have a single PINK1 pathogenic variant have been reported (see Clinical Description, Heterozygotes).

Sibs of a proband

  • If both parents are known to be heterozygous for a PINK1 pathogenic variant, each sib of an affected individual has at conception a 25% chance of being affected, a 50% chance of being heterozygous, and a 25% chance of inheriting neither of the familial PINK1 pathogenic variants.
  • The risk to heterozygotes of developing symptoms is not yet determined; however, several individuals with parkinsonism who have a single PINK1 pathogenic variant have been reported (see Clinical Description, Heterozygotes).

Offspring of a proband. The offspring of an individual with PINK1 type of young-onset Parkinson disease are obligate heterozygotes for a pathogenic variant in PINK1.

Other family members. Each sib of the proband's parents is at a 50% risk of being heterozygous for a PINK1 pathogenic variant.

Heterozygote Detection

Heterozygote testing for at-risk relatives requires prior identification of the PINK1 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 heterozygous, or are at risk of being heterozygous.

Prenatal Testing and Preimplantation Genetic Testing

Once the PINK1 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 decisions 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.

  • American Parkinson Disease Association (APDA)
    Phone: 800-223-2732
    Fax: 718-981-4399
    Email: apda@apdaparkinson.org
  • MedlinePlus
  • Michael J. Fox Foundation for Parkinson's Research
    Phone: 800-708-7644 (toll-free)
    Email: info@michaeljfox.org
  • Parkinson's Foundation
    Phone: 800-4PD-INFO (473-4636)
    Email: contact@parkinson.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.

PINK1 Type of Young-Onset Parkinson Disease : Genes and Databases

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 PINK1 Type of Young-Onset Parkinson Disease (View All in OMIM)

605909PARKINSON DISEASE 6, AUTOSOMAL RECESSIVE EARLY-ONSET; PARK6
608309PTEN-INDUCED KINASE 1; PINK1

Molecular Pathogenesis

PINK1 encodes serine/threoninep-protein kinase PINK1, mitochondrial (also called PTEN-induced putative kinase 1, or PINK1). PINK1 spans the outer mitochondrial membrane, with the C-terminal kinase domain facing the cytoplasm and the N-terminal inside the mitochondria. PINK1 presumably exerts its neuroprotective effect by phosphorylating specific mitochondrial proteins and modulating their functions [Sim et al 2006]. PINK1 and E3 ubiquitin-protein ligase parkin (parkin; see Parkinson Disease Overview) have been mapped to a shared pathway, with PINK1 acting upstream of parkin. PINK1 detects mitochondrial dysfunction, initiates the translocation of parkin to mitochondria, and signals parkin to ubiquitinate the damaged mitochondria, leading to their removal by autophagy [Pickrell & Youle 2015]. Thus, PINK1 and parkin acting together constitute a mitochondrial quality control function.

Most of the known pathogenic variants are localized within the serine/threonine kinase domain of PINK1, as expected [Valente et al 2004] (see www.mdsgene.org). PINK1 pathogenic variants or PINK1 silencing result in reduced mitochondrial DNA (mtDNA) levels, defective ATP production, impaired mitochondrial calcium handling, and increased free radical generation. This in turn results in a reduction of mitochondrial membrane potential and an increased susceptibility to apoptosis in neuronal cells, animal models, and patient-derived fibroblasts [Valente et al 2004, Gegg et al 2009, Abramov et al 2011].

Overexpression of parkin can rescue the effects of a PINK1 pathogenic variant in Drosophila and mammalian cells [Pickrell & Youle 2015]. Studies in fibroblasts from individuals with Parkinson disease revealed impaired ubiquitination of mitofusins and confirmed the link between the PINK1 and parkin pathways [Rakovic et al 2010, Rakovic et al 2011, Seibler et al 2011, Rakovic et al 2013, Koyano et al 2014]. A link with leucine-rich repeat serine/threonine-protein kinase 2 (LRRK2) [Azkona et al 2018] and alpha-synuclein levels [Chung et al 2016] has also been demonstrated. PINK1-patient-specific induced pluripotent stem cell-derived midbrain dopamine neurons exhibit mitochondrial dysfunction with increased susceptibility to mitochondrial toxins [Chung et al 2016]. Animal studies of Pink1 knockout mice showed defects in mitochondrial depolarization and synaptic transmission that were rescued by phosphomimetic NdufA10 in knockout mouse cells and in pinkB9-null mutated Drosophila [Morais et al 2014]. Complex I deficits and adenosine triphosphate synthesis were also rescued in cells derived from individuals with PINK1 type of young-onset Parkinson disease [Morais et al 2014]. Heterozygous Pink1 knockout (Pink+/-) mice show subtle alterations of dopamine-dependent striatal synaptic plasticity [Madeo et al 2014]. While chronic exposure to low doses of rotenone was not sufficient to alter mitochondrial integrity and ATP production in this model, the rotenone led to profound impairment in expression of long-term plasticity at corticostriatal synapses, suggesting that disruption of synaptic plasticity may represent an early feature of a premanifesting state of the disease [Martella et al 2016]. Findings of possible translational and treatment relevance include the role of vitamin K2 as a mitochondrial electron carrier rescuing PINK1 deficiency [Vos et al 2012] and the observation that cardiolipin promotes electron transport between ubiquinone and complex I to rescue PINK1 deficiency [Vos et al 2017].

Mechanism of disease causation. Loss of function

Neuropathology. Data in individuals with PINK1 type of young-onset Parkinson disease is limited [Samaranch et al 2010, Poulopoulos et al 2012]. Brain autopsy data are available for four individuals with biallelic PINK1 pathogenic variants [Samaranch et al 2010, Poulopoulos et al 2012, Steele et al 2015, Takanashi et al 2016, Nybø et al 2020], all of which had characteristic neuronal loss in the substantia nigra. Lewy bodies were present in on histopathology in three of four individuals, although distribution varied. One individual had Lewy bodies in the substantia nigra, the temporal cortex, and locus coeruleus [Nybø et al 2020]; the second in the reticular nuclei in the brain stem, substantia nigra, and basal nucleus of Meynert, with sparing of the locus coeruleus and amygdala [Samaranch et al 2010]; and the third only in the amygdala, with only minimal neurites in the substantia nigra [Steele et al 2015]. A fourth individual had no Lewy body pathology [Takanashi et al 2016]. In addition, tau depositions were present in two individuals [Steele et al 2015, Nybø et al 2020]. In a Parkinson disease brain bank study, four individuals with Parkinson disease and heterozygous PINK1 pathogenic variants had pathologic findings consistent with typical Parkinson disease, with Lewy bodies distributed in the brain stem and cortical areas, neuronal loss affecting the substantia nigra pars compacta, and neurofibrillary tangles stage I to V [Gandhi et al 2006].

Chapter Notes

Author Notes

Lara M Lange, MD (ed.kcebeul-inu@egnal.al), and Christine Klein, MD (ed.kcebeul-inu@nielk.enitsirhc), are actively involved in clinical and genetic research regarding individuals with Parkinson disease. They would be happy to communicate with persons who have any questions regarding the diagnosis of Parkinson disease or other considerations.

Lara M Lange and Christine Klein are also interested in hearing from clinicians treating families affected by Parkinson disease 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 Dr Lara M Lange or Prof Christine Klein to inquire about the review of PINK1 variants of uncertain significance.

Acknowledgments

Lara M Lange and Christine Klein would like to thank Susanne Schneider for her previous engagement in the PINK1 GeneReviews and Johanna Junker and Maria Leila M Doquenia for their work on the MDSGene PINK1 update.

Lara M Lange acknowledges support from the Bachmann-Strauss Dystonia and Parkinson Foundation.

Christine Klein acknowledges support from the Aligning Science Across Parkinson's (ASAP) Global Parkinson's Genetics Program (GP2), The Michael J Fox Foundation (MJFF), and the German Research Foundation (FOR2488).

Author History

Christine Klein, MD (2010-present)
Lara M Lange, MD (2024-present)
Susanne A Schneider, MD, PhD; Ludwig Maximilian University Munich (2010-2024)

Revision History

  • 25 April 2024 (sw) Comprehensive update posted live
  • 24 May 2018 (sw) Comprehensive update posted live
  • 18 September 2014 (me) Comprehensive update posted live
  • 6 September 2012 (me) Comprehensive update posted live
  • 16 March 2010 (me) Review posted live
  • 1 December 2009 (ck) Original submission

References

Literature Cited

  • Abou-Sleiman PM, Muqit MM, McDonald NQ, Yang YX, Gandhi S, Healy DG, Harvey K, Harvey RJ, Deas E, Bhatia K, Quinn N, Lees A, Latchman DS, Wood NW. A heterozygous effect for PINK1 mutations in Parkinson's disease? Ann Neurol. 2006;60:414-9. [PubMed: 16969854]
  • Abramov AY, Gegg M, Grunewald A, Wood NW, Klein C, Schapira AH. Bioenergetic consequences of PINK1 mutations in Parkinson disease. PLoS One. 2011;6:e25622. [PMC free article: PMC3197155] [PubMed: 22043288]
  • Azkona G, López de Maturana R, Del Rio P, Sousa A, Vazquez N, Zubiarrain A, Jimenez-Blasco D, Bolaños JP, Morales B, Auburger G, Arbelo JM, Sánchez-Pernaute R. LRRK2 expression is deregulated in fibroblasts and neurons from Parkinson patients with mutations in PINK1. Mol Neurobiol. 2018;55:506–16. [PMC free article: PMC5808058] [PubMed: 27975167]
  • Berardelli A, Wenning GK, Antonini A, Berg D, Bloem BR, Bonifati V, Brooks D, Burn DJ, Colosimo C, Fanciulli A, Ferreira J, Gasser T, Grandas F, Kanovsky P, Kostic V, Kulisevsky J, Oertel W, Poewe W, Reese JP, Relja M, Ruzicka E, Schrag A, Seppi K, Taba P, Vidailhet M. EFNS/MDS-ES/ENS [corrected] recommendations for the diagnosis of Parkinson's disease. Eur J Neurol. 2013;20:16–34. [PubMed: 23279440]
  • Binkofski F, Reetz K, Gaser C, Hilker R, Hagenah J, Hedrich K, van Eimeren T, Thiel A, Büchel C, Pramstaller PP, Siebner HR, Klein C. Morphometric fingerprint of asymptomatic Parkin and PINK1 mutation carriers in the basal ganglia. Neurology. 2007;69:842–50. [PubMed: 17724286]
  • Bloem BR, Okun MS, Klein C. Parkinson's disease. Lancet. 2021;397:2284-303. [PubMed: 33848468]
  • Bonifati V, Rohé CF, Breedveld GJ, Fabrizio E, De Mari M, Tassorelli C, Tavella A, Marconi R, Nicholl DJ, Chien HF, Fincati E, Abbruzzese G, Marini P, De Gaetano A, Horstink MW, Maat-Kievit JA, Sampaio C, Antonini A, Stocchi F, Montagna P, Toni V, Guidi M, Dalla Libera A, Tinazzi M, De Pandis F, Fabbrini G, Goldwurm S, de Klein A, Barbosa E, Lopiano L, Martignoni E, Lamberti P, Vanacore N, Meco G, Oostra BA, et al. Early-onset parkinsonism associated with PINK1 mutations: frequency, genotypes, and phenotypes. Neurology. 2005;65:87–95. [PubMed: 16009891]
  • Chung SY, Kishinevsky S, Mazzulli JR, Graziotto J, Mrejeru A, Mosharov EV, Puspita L, Valiulahi P, Sulzer D, Milner TA, Taldone T, Krainc D, Studer L, Shim JW. Parkin and PINK1 patient iPSC-derived midbrain dopamine neurons exhibit mitochondrial dysfunction and α-synuclein accumulation. Stem Cell Reports. 2016;7:664–77. [PMC free article: PMC5063469] [PubMed: 27641647]
  • Deuschl G, Antonini A, Costa J, Śmiłowska K, Berg D, Corvol JC, Fabbrini G, Ferreira J, Foltynie T, Mir P, Schrag A, Seppi K, Taba P, Ruzicka E, Selikhova M, Henschke N, Villanueva G, Moro E. European Academy of Neurology/Movement Disorder Society - European Section guideline on the treatment of Parkinson's disease: I. Invasive therapies. Eur J Neurol. 2022;29:2580-95. [PubMed: 35791766]
  • Eggers C, Schmidt A, Hagenah J, Brüggemann N, Klein JC, Tadic V, Kertelge L, Kasten M, Binkofski F, Siebner H, Neumaier B, Fink GR, Hilker R, Klein C. Progression of subtle motor signs in PINK1 mutation carriers with mild dopaminergic deficit. Neurology. 2010;74:1798–805. [PubMed: 20513816]
  • Gandhi S, Muqit MM, Stanyer L, Healy DG, Abou-Sleiman PM, Hargreaves I, Heales S, Ganguly M, Parsons L, Lees AJ, Latchman DS, Holton JL, Wood NW, Revesz T. PINK1 protein in normal human brain and Parkinson's disease. Brain. 2006;129:1720–31. [PubMed: 16702191]
  • Gegg ME, Cooper JM, Schapira AH, Taanman JW. Silencing of PINK1 expression affects mitochondrial DNA and oxidative phosphorylation in dopaminergic cells. PLoS One. 2009;4:e4756. [PMC free article: PMC2649444] [PubMed: 19270741]
  • Hayashida A, Li Y, Yoshino H, Daida K, Ikeda A, Ogaki K, Fuse A, Mori A, Takanashi M, Nakahara T, Yoritaka A, Tomizawa Y, Furukawa Y, Kanai K, Nakayama Y, Ito H, Ogino M, Hattori Y, Hattori T, Ichinose Y, Takiyama Y, Saito T, Kimura T, Aizawa H, Shoji H, Mizuno Y, Matsushita T, Sato M, Sekijima Y, Morita M, Iwasaki A, Kusaka H, Tada M, Tanaka F, Sakiyama Y, Fujimoto T, Nagara Y, Kashihara K, Todo H, Nakao K, Tsuruta K, Yoshikawa M, Hara H, Yokote H, Murase N, Nakamagoe K, Tamaoka A, Takamiya M, Morimoto N, Nokura K, Kako T, Funayama M, Nishioka K, Hattori N. The identified clinical features of Parkinson's disease in homo-, heterozygous and digenic variants of PINK1. Neurobiol Aging. 2021;97:146.e1-146.e13. [PubMed: 32713623]
  • Höglinger GU, Adler CH, Berg D, Klein C, Outeiro TF, Poewe W, Postuma R, Stoessl AJ, Lang AE. A biological classification of Parkinson's disease: the SynNeurGe research diagnostic criteria. Lancet Neurol. 2024;23:191-204. [PubMed: 38267191]
  • Jia F, Fellner A, Kumar KR. Monogenic Parkinson's Disease: Genotype, Phenotype, Pathophysiology, and Genetic Testing. Genes (Basel). 2022;13:471. [PMC free article: PMC8950888] [PubMed: 35328025]
  • 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]
  • Kasten M, Hartmann C, Hampf J, Schaake S, Westenberger A, Vollstedt EJ, Balck A, Domingo A, Vulinovic F, Dulovic M, Zorn I, Madoev H, Zehnle H, Lembeck CM, Schawe L, Reginold J, Huang J, König IR, Bertram L, Marras C, Lohmann K, Lill CM, Klein C. Genotype-phenotype relations for the Parkinson's disease genes Parkin, PINK1, DJ1: MDSGene Systematic Review. Mov Disord. 2018;33:730–41. [PubMed: 29644727]
  • Koyano F, Okatsu K, Kosako H, Tamura Y, Go E, Kimura M, Kimura Y, Tsuchiya H, Yoshihara H, Hirokawa T, Endo T, Fon EA, Trempe JF, Saeki Y, Tanaka K, Matsuda N. Ubiquitin is phosphorylated by PINK1 to activate parkin. Nature. 2014;510:162–6. [PubMed: 24784582]
  • Krohn L, Grenn FP, Makarious MB, Kim JJ, Bandres-Ciga S, Roosen DA, Gan-Or Z, Nalls MA, Singleton AB, Blauwendraat C; International Parkinson's Disease Genomics Consortium (IPDGC). Comprehensive assessment of PINK1 variants in Parkinson's disease. Neurobiol Aging. 2020 Jul;91:168.e1-168.e5. [PMC free article: PMC7236133] [PubMed: 32249012]
  • Kuusimäki T, Korpela J, Pekkonen E, Martikainen MH, Antonini A, Kaasinen V. Deep brain stimulation for monogenic Parkinson's disease: a systematic review. J Neurol. 2020;267:883-97. [PMC free article: PMC7109183] [PubMed: 30659355]
  • Lange LM, Gonzalez-Latapi P, Rajalingam R, Tijssen MAJ, Ebrahimi-Fakhari D, Gabbert C, Ganos C, Ghosh R, Kumar KR, Lang AE, Rossi M, van der Veen S, van de Warrenburg B, Warner T, Lohmann K, Klein C, Marras C; on behalf of the Task Force on Genetic Nomenclature in Movement Disorders. Nomenclature of Genetic Movement Disorders: Recommendations of the International Parkinson and Movement Disorder Society Task Force - An Update. Mov Disord. 2022;37:905-35. [PubMed: 35481685]
  • Li JY, Li NN, Wang L, Peng JX, Duan LR, Chen CL, Peng R. A compound heterozygous PINK1-associated juvenile Parkinson's disease with pregnancy in Chinese. J Neurol. 2021;268:2223-7. [PubMed: 33491134]
  • Madeo G, Schirinzi T, Martella G, Latagliata EC, Puglisi F, Shen J, Valente EM, Federici M, Mercuri NB, Puglisi-Allegra S, Bonsi P, Pisani A. PINK1 heterozygous mutations induce subtle alterations in dopamine-dependent synaptic plasticity. Mov Disord. 2014;29:41–53. [PMC free article: PMC4022284] [PubMed: 24167038]
  • Marras C, Lang A, van de Warrenburg BP, Sue CM, Tabrizi SJ, Bertram L, Mercimek-Mahmutoglu S, Ebrahimi-Fakhari D, Warner TT, Durr A, Assmann B, Lohmann K, Kostic V, Klein C. Nomenclature of genetic movement disorders: recommendations of the International Parkinson and Movement Disorder Society task force. Mov Disord. 2016;31:436–57. [PubMed: 27079681]
  • Martella G, Madeo G, Maltese M, Vanni V, Puglisi F, Ferraro E, Schirinzi T, Valente EM, Bonanni L, Shen J, Mandolesi G, Mercuri NB, Bonsi P, Pisani A. Exposure to low-dose rotenone precipitates synaptic plasticity alterations in PINK1 heterozygous knockout mice. Neurobiol Dis. 2016;91:21–36. [PubMed: 26916954]
  • McNeill A, Wu RM, Tzen KY, Aguiar PC, Arbelo JM, Barone P, Bhatia K, Barsottini O, Bonifati V, Bostantjopoulou S, Bressan R, Cossu G, Cortelli P, Felicio A, Ferraz HB, Herrera J, Houlden H, Hoexter M, Isla C, Lees A, Lorenzo-Betancor O, Mencacci NE, Pastor P, Pappata S, Pellecchia MT, Silveria-Moriyama L, Varrone A, Foltynie T, Schapira AH. Dopaminergic neuronal imaging in genetic Parkinson's disease: insights into pathogenesis. PLoS One. 2013;8:e69190. [PMC free article: PMC3720622] [PubMed: 23935950]
  • Morais VA, Haddad D, Craessaerts K, De Bock PJ, Swerts J, Vilain S, Aerts L, Overbergh L, Grünewald A, Seibler P, Klein C, Gevaert K, Verstreken P, De Strooper B. PINK1 loss-of-function mutations affect mitochondrial complex I activity via NdufA10 ubiquinone uncoupling. Science. 2014;344:203–7. [PubMed: 24652937]
  • Nürnberger L, Klein C, Baudrexel S, Roggendorf J, Hildner M, Chen S, Kang JS, Hilker R, Hagenah J. Ultrasound-based motion analysis demonstrates bilateral arm hypokinesia during gait in heterozygous PINK1 mutation carriers. Mov Disord. 2015;30:386–92. [PubMed: 25545816]
  • Nybø CJ, Gustavsson EK, Farrer MJ, Aasly JO. Neuropathological findings in PINK1-associated Parkinson's disease. Parkinsonism Relat Disord. 2020;78:105-8. [PubMed: 32814227]
  • Over L, Brüggemann N, Lohmann K. Therapies for Genetic Forms of Parkinson's Disease: Systematic Literature Review. J Neuromuscul Dis. 2021;8:341-56. [PMC free article: PMC8203229] [PubMed: 33459660]
  • Piccoli C, Ripoli M, Quarato G, Scrima R, D'Aprile A, Boffoli D, Margaglione M, Criscuolo C, De Michele G, Sardanelli A, Papa S, Capitanio N. Coexistence of mutations in PINK1 and mitochondrial DNA in early onset parkinsonism. J Med Genet. 2008;45:596–602. [PubMed: 18524835]
  • Pickrell AM, Youle RJ. The roles of PINK1, parkin, and mitochondrial fidelity in Parkinson's disease. Neuron. 2015;85:257-73. [PMC free article: PMC4764997] [PubMed: 25611507]
  • Poulopoulos M, Levy OA, Alcalay RN. The neuropathology of genetic Parkinson's disease. Mov Disord. 2012;27:831–42. [PMC free article: PMC3383342] [PubMed: 22451330]
  • Puschmann A, Fiesel FC, Caulfield TR, Hudec R, Ando M, Truban D, Hou X, Ogaki K, Heckman MG, James ED, Swanberg M, Jimenez-Ferrer I, Hansson O, Opala G, Siuda J, Boczarska-Jedynak M, Friedman A, Koziorowski D, Aasly JO, Lynch T, Mellick GD, Mohan M, Silburn PA, Sanotsky Y, Vilariño-Güell C, Farrer MJ, Chen L, Dawson VL, Dawson TM, Wszolek ZK, Ross OA, Springer W. Heterozygous PINK1 p.G411S increases risk of Parkinson's disease via a dominant-negative mechanism. Brain. 2017;140:98-117. [PMC free article: PMC5379862] [PubMed: 27807026]
  • Rakovic A, Grünewald A, Kottwitz J, Brüggemann N, Pramstaller PP, Lohmann K, Klein C. Mutations in PINK1 and Parkin impair ubiquitination of mitofusins in human fibroblasts. PLoS One. 2011;6:e16746. [PMC free article: PMC3050809] [PubMed: 21408142]
  • Rakovic A, Grünewald A, Seibler P, Ramirez A, Kock N, Orolicki S, Lohmann K, Klein C. Effect of endogenous mutant and wild-type PINK1 on Parkin in fibroblasts from Parkinson disease patients. Hum Mol Genet. 2010;19:3124–37. [PubMed: 20508036]
  • Rakovic A, Shurkewitsch K, Seibler P, Grünewald A, Zanon A, Hagenah J, Krainc D, Klein C. Phosphatase and tensin homolog (PTEN)-induced putative kinase 1 (PINK1)-dependent ubiquitination of endogenous Parkin attenuates mitophagy: study in human primary fibroblasts and induced pluripotent stem cell-derived neurons. J Biol Chem. 2013;288:2223–37. [PMC free article: PMC3554895] [PubMed: 23212910]
  • Reetz K, Tadic V, Kasten M, Brüggemann N, Schmidt A, Hagenah J, Pramstaller PP, Ramirez A, Behrens MI, Siebner HR, Klein C, Binkofski F. Structural imaging in the presymptomatic stage of genetically determined parkinsonism. Neurobiol Dis. 2010;39:402–8. [PubMed: 20483373]
  • Ricciardi L, Petrucci S, Guidubaldi A, Ialongo T, Serra L, Ferraris A, Spanò B, Bozzali M, Valente EM, Bentivoglio AR. Phenotypic variability of PINK1 expression: 12 years' clinical follow-up of two Italian families. Mov Disord. 2014;29:1561–6. [PubMed: 25164310]
  • 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; ACMG Laboratory Quality Assurance Committee. 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]
  • Samaranch L, Lorenzo-Betancor O, Arbelo JM, Ferrer I, Lorenzo E, Irigoyen J, Pastor MA, Marrero C, Isla C, Herrera-Henriquez J, Pastor P. PINK1-linked parkinsonism is associated with Lewy body pathology. Brain. 2010;133:1128–42. [PubMed: 20356854]
  • Seibler P, Graziotto J, Jeong H, Simunovic F, Klein C, Krainc D. Mitochondrial Parkin recruitment is impaired in neurons derived from mutant PINK1induced pluripotent stem cells. J Neurosci. 2011;31:5970–6. [PMC free article: PMC3091622] [PubMed: 21508222]
  • Seier M, Hiller A. Parkinson's disease and pregnancy: An updated review. Parkinsonism Relat Disord. 2017;40:11-7. [PubMed: 28506531]
  • Sim CH, Lio DS, Mok SS, Masters CL, Hill AF, Culvenor JG, Cheng HC. C-terminal truncation and Parkinson's disease-associated mutations down-regulate the protein serine/threonine kinase activity of PTEN-induced kinase-1. Hum Mol Genet. 2006;15:3251–62. [PubMed: 17000703]
  • Steele JC, Guella I, Szu-Tu C, Lin MK, Thompson C, Evans DM, Sherman HE, Vilariño-Güell C, Gwinn K, Morris H, Dickson DW, Farrer MJ. Defining neurodegeneration on Guam by targeted genomic sequencing. Ann Neurol. 2015;77:458-68. [PubMed: 25558820]
  • 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]
  • Takanashi M, Li Y, Hattori N. Absence of Lewy pathology associated with PINK1 homozygous mutation. Neurology. 2016;86:2212-3. [PubMed: 27164705]
  • Trinh J, Hicks AA, König IR, Delcambre S, Lüth T, Schaake S, Wasner K, Ghelfi J, Borsche M, Vilariño-Güell C, Hentati F, Germer EL, Bauer P, Takanashi M, Kostić V, Lang AE, Brüggemann N, Pramstaller PP, Pichler I, Rajput A, Hattori N, Farrer MJ, Lohmann K, Weissensteiner H, May P, Klein C, Grünewald A. Mitochondrial DNA heteroplasmy distinguishes disease manifestation in PINK1/PRKN-linked Parkinson's disease. Brain. 2023;146:2753-65. [PMC free article: PMC10316771] [PubMed: 36478228]
  • Valente E, Ferraris A. Pink1 (PARK6) and Parkinson’s disease. In: Schapira A, Lang AE, Fahn S. Saunders, eds. Movement Disorders 4. Blue Books of Neurology. Philadelphia, PA: Elsevier; 2010:66-82.
  • Valente EM, Abou-Sleiman PM, Caputo V, Muqit MM, Harvey K, Gispert S, Ali Z, Del Turco D, Bentivoglio AR, Healy DG, Albanese A, Nussbaum R, González-Maldonado R, Deller T, Salvi S, Cortelli P, Gilks WP, Latchman DS, Harvey RJ, Dallapiccola B, Auburger G, Wood NW. Hereditary early-onset Parkinson's disease caused by mutations in PINK1. Science. 2004;304:1158–60. [PubMed: 15087508]
  • Vos M, Esposito G, Edirisinghe JN, Vilain S, Haddad DM, Slabbaert JR, Van Meensel S, Schaap O, De Strooper B, Meganathan R, Morais VA, Verstreken P. Vitamin K2 is a mitochondrial electron carrier that rescues pink1 deficiency. Science. 2012;336:1306–10. [PubMed: 22582012]
  • Vos M, Geens A, Böhm C, Deaulmerie L, Swerts J, Rossi M, Craessaerts K, Leites EP, Seibler P, Rakovic A, Lohnau T, De Strooper B, Fendt SM, Morais VA, Klein C, Verstreken P. Cardiolipin promotes electron transport between ubiquinone and complex I to rescue PINK1 deficiency. J Cell Biol. 2017;216:695–708. [PMC free article: PMC5346965] [PubMed: 28137779]
  • Weissbach A, Bäumer T, Pramstaller PP, Brüggemann N, Tadic V, Chen R, Klein C, Münchau A. Abnormal premotor-motor interaction in heterozygous Parkin- and Pink1 mutation carriers. Clin Neurophysiol. 2017;128:275–80. [PubMed: 27843055]
  • Wittke C, Petkovic S, Dobricic V, Schaake S, Respondek G, Weissbach A, Madoev H, Trinh J, Vollstedt EJ, Kuhnke N, Lohmann K, Dulovic Mahlow M, Marras C, König IR, Stamelou M, Bonifati V, Lill CM, Kasten M, Huppertz HJ, Höglinger G, Klein C, et al. Genotype-phenotype relations for the atypical parkinsonism genes: MDSGene systematic review. Mov Disord. 2021;36:1499-510. [PMC free article: PMC9070562] [PubMed: 34396589]
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: NBK26472PMID: 20301792

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...