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

LRRK2 Parkinson Disease

, MD, MPH, , MS, and , MS.

Author Information and Affiliations

Initial Posting: ; Last Revision: July 6, 2023.

Estimated reading time: 46 minutes

Summary

Clinical characteristics.

LRRK2 Parkinson disease (PD) is characterized by features consistent with idiopathic PD: initial motor features of slowly progressive asymmetric tremor at rest and/or bradykinesia, cogwheel muscle rigidity, postural instability, and gait abnormalities that may include festination and freezing. Certain nonmotor symptoms in LRRK2-PD, especially REM sleep behavior disorder and cognitive decline, may occur at similar or slightly reduced frequency compared to typical idiopathic* PD. Onset is generally after age 50, although early-onset (in the 20s) and late-onset (in the 90s) disease has been described.

* Idiopathic PD refers to the presence of signs and symptoms of PD for which the etiology is currently unknown and in which there is no known family history of PD.

Diagnosis/testing.

The diagnosis of LRRK2-PD relies on clinical findings and the identification of a heterozygous pathogenic variant in LRRK2.

Management.

Treatment of manifestations: Symptomatic treatment of parkinsonism is the same as for idiopathic Parkinson disease: pharmacologic replacement of dopamine, most commonly accomplished with the precursor of dopamine, L-dopa, combined with carbi-dopa. Dopamine agonists may also be used, as well as monoamine oxidase-B (MAO-B) inhibitors, amantadine, and/or anticholinergics. Physical, occupational, and voice therapy may be beneficial. Exercise is often recommended. Treatment of nonmotor manifestations – e.g., depression, anxiety, sleep disorders, urinary issues, orthostatic hypotension – should be addressed based on individual manifestations.

Surveillance: Annual evaluation for both motor and nonmotor symptoms. Motor evaluation focuses on gait and falls, slowness of movement and dexterity, tremor, and rigidity. Evaluation for nonmotor signs and symptoms includes assessment of constipation, mood disorder, impulse control disorders, other psychiatric disorders, cognitive changes, sleep disturbance, orthostatic hypotension, and urinary frequency. In addition, at least yearly evaluation for melanoma.

Agents/circumstances to avoid: Dopamine-blocking therapies may exacerbate parkinsonism.

Genetic counseling.

LRRK2-PD is inherited in an autosomal dominant manner. However, given the reduced penetrance associated with LRRK2-PD, a high percentage of affected individuals report unaffected parents. De novo mutation may occur; its frequency is unknown. Each child of an individual with LRRK2 Parkinson disease has a 50% chance of inheriting the pathogenic variant. However, the risk of developing disease is lower than 50% because of age-related reduced penetrance. Prenatal diagnosis for pregnancies at increased risk is possible if the pathogenic variant in the family is known.

Diagnosis

While there are subtle group differences between LRRK2 Parkinson disease (LRRK2-PD) and idiopathic PD (IPD), on an individual case basis LRRK2-PD is indistinguishable from idiopathic PD. Criteria for probable and clinically established PD were developed by the International Parkinson and Movement Disorders Society [Postuma et al 2015] (see also Parkinson Disease Overview).

Note: "Idiopathic Parkinson disease" and "sporadic Parkinson disease" are terms used in the Parkinson disease medical literature to describe Parkinson disease of unknown cause diagnosed in an individual with a negative family history. Future advances in the understanding of genetic risk factors are likely to identify genetic causes / risk factors for some Parkinson disease currently considered "idiopathic" or "sporadic."

Suggestive Findings

LRRK2 Parkinson disease (LRRK2-PD) should be suspected in individuals with the following clinical features, neuroimaging findings, and family history.

Clinical features include signs of Parkinson disease (PD):

  • Bradykinesia (slowness of movement) with decrements in speed or amplitude as movements are continued
    AND
  • Rest tremor (4-6-Hz tremor in a fully resting limb) and/or rigidity.

To meet Movement Disorders Society clinical diagnostic criteria [Postuma et al 2015], at least two supportive findings in addition to the signs of PD listed above should be present:

  • Clear and dramatic beneficial response to dopaminergic therapy
  • Levodopa-induced dyskinesias
  • Rest tremor in a limb (prior or current)
  • Positive results from an ancillary diagnostic test (e.g.):
    • Cardiac metaiodobenzylguanidine (MIBG) scintigraphy
    • Olfactory abnormality

However: (1) Early and untreated individuals with PD may not meet the supportive criteria, as dopaminergic therapy may not have been tried, and thus no response or dyskinesias documented; (2) attributing olfactory abnormality to LRRK2-PD may be problematic, as the frequency of abnormal olfaction is reduced in individuals with LRRK2-PD compared with those who have IPD [Silveira-Moriyama et al 2010, Ruiz-Martínez et al 2011, Saunders-Pullman et al 2014].

Exclusion criteria. Early eye movement abnormalities, significant orthostatic hypotension, and signs and symptoms that support atypical parkinsonism are generally exclusionary criteria. However, LRRK2-PD may occasionally manifest with atypical features.

Neuroimaging findings (brain CT and MRI) are normal. DaT or PET scan may further support the diagnosis, but neither is necessary for diagnosis.

Family history for LRRK2-PD consistent with autosomal dominant inheritance. However, absence of a family history of LRRK2-PD does not preclude the diagnosis (see Genetic Counseling).

Establishing the Diagnosis

The diagnosis of LRRK2-PD is established in a proband with clinical features consistent with PD and a heterozygous pathogenic (or likely pathogenic) variant in LRRK2 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 a heterozygous LRRK2 variant of uncertain significance does not establish or rule out the diagnosis.

Gene-targeted testing requires that the clinician determine which gene(s) are likely involved, whereas genomic testing does not. Because the phenotype of LRRK2-PD overlaps with IPD and other genetic forms of PD, the best approach to understanding the genetic cause in an individual usually includes testing for LRRK2 as well as other genes that lead to PD.

Molecular genetic testing approaches can include a combination of gene-targeted testing (single-gene testing, multigene panel) and comprehensive genomic testing (exome sequencing, exome array, genome sequencing) depending on the phenotype.

Single-gene testing

  • Targeted analysis for specific pathogenic variants can be performed first in individuals with PD of certain ancestries, including Ashkenazi (Eastern European) Jewish, North African Berber, Spanish/Hispanic, Belgian, Portuguese, and Asian (see Molecular Genetics, Pathogenic Variants), or when the family-specific pathogenic variant is known. However, additional testing to evaluate for other causative genes may also be indicated. Ashkenazi Jews may occasionally have both the LRRK2 p.Gly2019Ser variant and a GBA1 (formerly GBA) Parkinson-related variant [Yahalom et al 2019].
  • Sequence analysis of LRRK2 may be performed in individuals who are not of any of the ancestries listed above if the suspicion of LRRK2-PD is high.

A multigene panel that includes LRRK2 and other genes of interest (see Differential Diagnosis) may be considered. 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; thus, clinicians need to determine which multigene panel is most likely 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. (3) Methods used in a panel may include sequence analysis, deletion/duplication analysis, and/or other non-sequencing-based tests. (4) Currently some PD panels do not include testing for variants in glucocerebrosidase resulting from pathogenic variants in GBA1, the most frequent cause of genetic PD, for which clinical trials are ongoing. Therefore, single-gene testing for GBA1 may be needed if not included on the panel.

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

Comprehensive genomic testing. When the phenotype is indistinguishable from many other inherited disorders characterized by features of Parkinson disease, comprehensive genomic testing (which does not require the clinician to determine which gene[s] are likely involved) is the best option. 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 1.

Molecular Genetic Testing Used in LRRK2 Parkinson Disease

Gene 1MethodProportion of Probands with a Pathogenic Variant 2 Detectable by Method
LRRK2 Sequence analysis 3~100% 4
Gene-targeted deletion/duplication analysis 5See footnote 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 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 interpretation of sequence analysis results, click here.

4.

LRRK2-PD is defined by the presence of a pathogenic variant in LRRK2; thus, the pathogenic variant detection rate approaches 100% for nucleotide changes, small deletions/insertions, and pathogenic variants in splice site consensus motifs.

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.

To date, no deletions or duplications involving LRRK2 have been reported to cause LRRK2 Parkinson disease [Mata et al 2005a, Di Fonzo et al 2006].

7.

Homozygous p.Gly2019Ser cases are not phenotypically different from heterozygous cases [Ishihara et al 2006, Ben Romdhan et al 2018].

Clinical Characteristics

Clinical Description

LRRK2 Parkinson disease (PD) is characterized by features consistent with idiopathic PD (IPD), including initial symptoms of slowness, decreased arm swing, rest tremor, and initial motor signs of bradykinesia, associated with rigidity and/or resting tremor. Gait abnormalities and postural instability also occur, particularly with progression of the disease [Alcalay et al 2013, Mirelman et al 2013, Trinh et al 2014b]. While indistinguishable individually from IPD, motor and cognitive features suggest that as a group, signs and symptoms of LRRK2-PD may be milder than in IPD [Kestenbaum & Alcalay 2017, Ben Romdhan et al 2018, Saunders-Pullman et al 2018] (see Phenotype Correlations by Cause).

LRRK2-PD disease onset is insidious with a slowly progressive course.

Motor features. Data conflict regarding the predominant motor features specific to LRRK2-PD. Some have reported more tremor at onset [Healy et al 2008, Marras et al 2011], while others have reported more gait-predominant features, postural instability, and rigidity at onset [Alcalay et al 2009, Gan-Or et al 2010]. Motor features are overall similar to IPD and include the following:

  • Slowness and difficulty with dexterity (bradykinesia)
  • Tremor, which may occur at rest or during action, although this sign may not be present
  • Slow walk or shuffling gait
  • Unsteadiness and falls
  • Low vocal volume, with difficulty projecting
  • Facial hypomimia (masking)

As with other forms of Parkinson disease, the disorder is slowly progressive, and generally spreads from unilateral to bilateral involvement. Disease progression varies significantly among individuals and is related to age of onset.

Nonmotor features may appear prior to the movement disorder or emerge with motor disease progression. They include the following [Healy et al 2008, Saunders-Pullman et al 2011, Alcalay et al 2013, Gaig et al 2014, Alcalay et al 2015]:

  • Cognitive decline, including mild cognitive impairment and dementia
  • Constipation
  • Hyposmia/anosmia
  • Depression, anxiety, and other neuropsychiatric features [Brockmann et al 2011, Marras et al 2011, Shanker et al 2011]
  • Seborrhea
  • Sexual dysfunction
  • Sleep complaints, including poor sleep quality, excessive daytime sleepiness, sleep fragmentation, early awakening, daytime sleepiness, and insomnia [Pont-Sunyer et al 2015]
  • Urinary frequency
  • Orthostatic hypotension

Risk of malignancy

Atypical Parkinson disease phenotypes. For a small number of individuals with a LRRK2 pathogenic variant, clinical features and/or pathology support an atypical parkinsonian syndrome. These atypical phenotypes include progressive supranuclear palsy (PSP), multiple-system atrophy, corticobasal syndrome, amyotrophic lateral sclerosis-like signs, and frontotemporal dementia [Zimprich et al 2004a, Ross et al 2006, Dächsel et al 2007, Chen-Plotkin et al 2008, Santos-Rebouças et al 2008, Lee et al 2018b]. See also Neuropathology (below).

DaT and PET scanning. In general, PET scan results in individuals with LRRK2-PD are similar to those in others with adult-onset PD from a variety of causes.

  • There is typically significant reduction in uptake of 18F-dopa (PET) and dopamine transporter (DaT-SPECT).
  • Studies of these and other tracers support abnormalities of both dopaminergic and serotonergic metabolism [Lin et al 2008, Fu et al 2018].

Infrequently, normal PET scanning early in the disease course has been observed in both LRRK2-PD and non-LRRK2-PD and does not preclude a PD diagnosis [Wile et al 2016].

Neuropathology. The hallmark pathologic features of the common idiopathic form of PD are neuronal loss and gliosis in the substantia nigra and the presence of intracytoplasmic inclusions (or Lewy bodies). The majority of individuals with LRRK2-PD exhibit these characteristics [Ross et al 2006]. However, a significant subset of individuals with LRRK2-PD, particularly those with the p.Gly2019Ser pathogenic variant, have substantia nigra dopaminergic neuronal loss and gliosis without accompanying Lewy body inclusion. Of great interest, the pathology correlates with the extent of nonmotor clinical features. Kalia et al [2015] correlated the presence of Lewy bodies with nonmotor features of cognitive impairment / dementia, anxiety, orthostatic hypotension, and the absence of Lewy bodies with a predominantly motor phenotype.

LRRK2-PD has also been documented with alternate pathologies. Of particular note, three out of six individuals with parkinsonism and a heterozygous LRRK2 pathogenic variant identified in a large brain bank series were found to have pathology suggestive of PSP or pre-PSP with tau-positive neurons, neuropil threads, and tufted astrocytes [Blauwendraat et al 2019]. Additional series include those with PSP pathology and neurofibrillary tangles, as well as ubiquitin-immunopositive inclusions (Marinesco bodies) and TDP-43 inclusions [Wszolek et al 2004, Zimprich et al 2004b, Funayama et al 2005, Ross et al 2006, Covy et al 2009, Ujiie et al 2012].

Click here (pdf) for more information on neuropathology.

Phenotype Correlations by Cause

Among the confirmed pathogenic variants in LRRK2, the pathogenic p.Gly2019Ser variant is the most common. The numerous published reports related to its associated clinical features are the focus of this section. See Genotype-Phenotype Correlations for differences associated with other LRRK2 variants.

Most studies have shown that individual clinical features are indistinguishable between individuals with LRRK2-PD and idiopathic PD (IPD) [Healy et al 2008, Alcalay et al 2013]. However, mild differences are observed in many studies. These cluster overall on a slightly milder clinical motor course for individuals with LRRK2-PD who have the p.Gly2019Ser variant compared to those with IPD. Some of the difference in nonmotor features may be attributable to neuropathologic heterogeneity, as individuals with fewer or less severe nonmotor features are more likely to have isolated nigral degeneration without associated Lewy bodies [Kalia et al 2015].

Reported Clinical Differences Between Individuals with LRRK2-PD and IPD

While LRRK2-PD typically has late-onset disease (mean of 58 years [Marder et al 2015] or 61.6 years [San Luciano et al 2017]), a subset of individuals have very early-onset disease (in the 20s). This may be related to other genes that modify the expression of LRRK2 [Trinh et al 2016].

Affected male-to-female ratio is similar, compared to 60% male and 40% female for IPD [Gan-Or et al 2015, San Luciano et al 2017].

Overall survival is longer in those with LRRK2-PD [Thaler et al 2018].

Motor features

Nonmotor features

Co-Occurring Pathogenic Variants in LRRK2 and GBA1

Among 12 Ashkenazi Jewish individuals who had both the LRRK2 pathogenic p.Gly2019Ser variant and a heterozygous pathogenic GBA1 (formerly GBA) variant, no significant differences in phenotype were observed when compared to those with just the LRRK2 variant [Yahalom et al 2019].

Genotype-Phenotype Correlations

Differences in phenotype have been observed for the rarer pathogenic LRRK2 variants as compared to the p.Gly2019Ser variant [Domingo & Klein 2018]:

  • Individuals with the p.Arg1441Gly pathogenic variant may be more likely to have excessive tremor, although they may also present with bradykinesia [Paisán-Ruíz et al 2004, Mata et al 2005b].
    However, one systematic review found more frequent tremor in individuals with the p.Gly2019Ser variant compared to those with the p.Arg1441Gly variant.
    The same review found that individuals with a pathogenic variant at residue 1441 (p.Arg1441Gly, p.Arg1441Cys, p.Arg1441His, p.Arg1441Ser) have more frequent motor fluctuations compared to individuals with the p.Gly2019Ser variant [Trinh et al 2018].
  • Individuals with a p.Gly2385Arg variant appear to have more rapidly progressive parkinsonism with greater subjective and objective scores of motor decline, as well as more motor fluctuations, compared to individuals with the p.Gly2019Ser variant [Marras et al 2016].
  • Individuals with at least one risk-factor variant (p.Gly2385Arg, p.Arg1628Pro, or p.Ser1647Thr) showed greater motor progression over a four-year period than those with the p.Gly2019Ser variant [Oosterveld et al 2015, Saunders-Pullman et al 2018].
  • Individuals with the pathogenic p.Tyr1699Cys variant may present with a variety of signs and symptoms. This variant was identified in a large Korean family who had clinical features similar to those with idiopathic PD, although interfamilial clinical heterogeneity was described [Kim et al 2012]. Atypical presentations initially reported include symptoms of dementia and amyotrophy [Zimprich et al 2004a; Family A] or behavior disorder characterized by depression and anxiety [Khan et al 2005; Lincolnshire kindred].

Penetrance

Penetrance of LRRK2 pathogenic variants is age dependent and may vary based on pathogenic variant and population ethnicity (including ancestral background and country of origin [Hentati et al 2014]). Both Marder et al [2015] and Lee et al [2017a] employed the kin-cohort method to estimate genotypes of family members whose genotypes were unknown. In these two studies penetrance in Jews and non-Jews was not statistically different. Penetrance in men and women was also not statistically different.

The most frequent variant is p.Gly2019Ser; it exists as a founder variant in both Eastern European (Ashkenazi) Jews and North African Berbers.

Other variants. High penetrance was reported for some of the rarer pathogenic LRRK2 variants [Lee et al 2017a]:

Nomenclature

Alternate nomenclature for LRRK2-PD includes the following:

  • PARK8, which refers to the chromosomal region of 12q12 linked to disease in a large Japanese PD kindred [Funayama et al 2002].
  • "Dardarin," the Basque word for "tremor," has been used to refer to LRRK2-PD caused by the pathogenic p.Arg1441Gly variant.

Prevalence

In the US, LRRK2-PD causes approximately 1% of simplex PD (i.e., single occurrences in a family) and approximately 5%-6% of familial PD.

  • A founder effect exists for certain pathogenic variants, such as p.Gly2019Ser (North African Berber and Ashkenazi Jewish populations). p.Gly2019Ser is also common in individuals of Portuguese, Brazilian, Spanish, and Italian ancestry and has been reported in Puerto Rican, British, Norwegian, Japanese, Chinese, and Indian populations.
  • A founder effect exists for the p.Arg1441Gly variant in the Basque population.
  • p.Arg1441Cys may be more common in the southern Italian and Belgian populations.

Further information about the prevalence of certain variants in different populations can be accessed here (pdf).

Differential Diagnosis

The differential diagnosis of LRRK2 Parkinson disease includes other genes associated with inherited forms of Parkinson disease, neurologic entities that commonly mimic Parkinson disease, and autosomal dominant neurologic conditions and genetic disorders in which parkinsonism can be a prominent feature (see Parkinson Disease Overview).

Management

Evaluations Following Initial Diagnosis

To establish the extent of disease and needs in an individual diagnosed with LRRK2 Parkinson disease, the evaluations below (if not performed as part of the evaluation that led to the diagnosis) are recommended:

  • Assessment of motor symptoms and signs, including motor functioning and falls
  • Assessment of nonmotor symptoms and signs, including:
    • Cognition
    • Constipation
    • Mood
    • Illusions/hallucinations
    • Sexual dysfunction
    • Pain
    • Sleep disturbance
    • Urinary difficulties, including frequency
    • Orthostatic hypotension
    • Baseline skin evaluation for evidence of melanoma
  • Consider referral to a clinical geneticist and/or genetic counselor.

Treatment of Manifestations

The treatment of individuals with LRRK2-PD does not differ from that of idiopathic PD (IPD), and should be tailored to the individual (see also Parkinson Disease Overview), with referral to a neurologist with training in movement disorders, a rehabilitation medicine specialist, psychiatrist, sleep medicine specialist, gastroenterologist, urologist, and pain medicine specialist, as indicated.

Pharmacotherapy, if needed, is symptomatic and may include the following:

  • Pharmacologic replacement of dopamine
    • Different formulations of levodopa may be considered, including l-dopa combined with cardi-dopa.
    • Multiple formulations of l-dopa (which may be immediate release or longer acting) exist, as well as multiple routes of delivery, including oral (the major one), sublingual, and enteric.
    • Utilization of l-dopa should be driven by clinical need, and the clinician should consider using the lowest dose that yields a satisfactory clinical effect [Olanow & Stocchi 2018, Bressman & Saunders-Pullman 2019].
  • Monoamine oxidate B (MAO-B) inhibitors, including selegiline and rasagiline
  • Amantadine (Symmetrel®)
  • Dopamine receptor agonists
  • Anticholinergics

Supportive treatment

  • Exercise, as safe for the particular affected individual; often recommended [Giladi et al 2016, Lee et al 2018a]
  • Physical and occupational therapy
  • Voice therapy, particularly the Lee Silverman Voice Treatment
  • Cognitive behavioral therapy; potentially beneficial either with or without pharmacologic treatment
  • For those with hallucinations, consideration of non-dopamine-blocking medications (particularly pimavanserin; see Gaig et al [2014])

Treatment of complications of levodopa therapy. With disease progression, up to approximately half of individuals with PD develop complications of levodopa therapy within the first two years of treatment [PSG 2000, Stocchi et al 2010]; complications may include the following:

  • Troubling dyskinesias
    • Dyskinesias in individuals with LRRK2-PD occur at a similar or lower frequency than in those with IPD [Healy et al 2008], with no difference in prevalence or latency to dyskinesia [Yahalom et al 2012].
    • In general, women with LRRK2-PD have a higher rate of dyskinesias than men [San Luciano et al 2017].
    • While dyskinesia is related to l-dopa dose, decisions regarding dosing should be guided by the clinician [Olanow & Stocchi 2018].
  • Significant wearing-off of doses
  • Delayed onset of medication action
  • Refractory rest tremor

Neurosurgical procedures such as deep brain stimulation (DBS) of the subthalamic nucleus (STN) / globus pallidus interna (GPi) may be considered if there is good response to but complications from l-dopa therapy.

Further, disease-modifying clinical trials are under way for LRRK2-PD (see Therapies Under Investigation).

Surveillance

Table 2.

Recommended Surveillance for Individuals with LRRK2 Parkinson Disease

System/ConcernAnnual Evaluation
Neurologic Neurologic exam: 1 incl bradykinesia, rigidity, tremor, & gait & postural stability assessment (MDS-UPDRS) 2
Gastrointestinal Assess for signs & symptoms of constipation.
Psychiatric Assess for symptoms of depression, anxiety, apathy, hallucinations, illusions, impulse control disorders, or other psychiatric condition.
Sleep disorder Assess for signs & symptoms.
Cardiovascular Assess for orthostatic hypotension.
Urologic/Gynecologic Assess for urinary frequency & sexual dysfunction.
Skin Full skin exam by dermatologist for signs of melanoma. 3
1.

To assess effect of motor therapies (including wearing off and dyskinesias) and need for symptomatic treatment

2.

Systematic assessment of history of motor and nonmotor features including associated disability as well as examination of motor features are captured in the Movement Disorder Society Sponsored Revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS) [Goetz et al 2008].

3.

Agents/Circumstances to Avoid

Dopamine-blocking therapies (both typical and atypical dopamine-blocking psychiatric medications as well as dopamine blockers for gastrointestinal causes) may exacerbate parkinsonism in LRRK2-PD and should be avoided, when possible.

Evaluation of Relatives at Risk

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

Pregnancy Management

While the data are restricted to case reports and registries, most support treatment with L-dopa during pregnancy (reviewed by Seier & Hiller [2017]).

See MotherToBaby for further information on medication use during pregnancy.

Therapies Under Investigation

The Lrrk2 protein is a fusion of Rab (Roc), COR, and kinase (MAPK) domains, and pathogenic variants are postulated to augment kinase activity [Kachergus et al 2005, West et al 2005, Gloeckner et al 2006, Greggio et al 2006]. Hence, the development of specific kinase inhibitors offers an attractive therapeutic target for neuroprotection in asymptomatic and affected LRRK2 heterozygotes, as well as for IPD [Albrecht 2005, Toft et al 2005]. A number of inhibitors are being developed; however, selectivity, specificity, and delivery into the central nervous system remain difficult issues to address [Lee et al 2010]. LRRK2 kinase inhibition had been associated with pulmonary complications in rodents and non-human primates [Herzig et al 2011, Baptista et al 2013, Fuji et al 2015]. However, more recent data suggest that this effect is reversible, and that toxicity is not significant (see bioRxiv). Initial studies of the drug DNL201 have proceeded to a Phase I clinical trial (ClinicalTrials.gov).

In addition to kinase inhibitors, antisense oligonucleotides have been evaluated preclinically [Zhao et al 2017], and may have therapeutic potential. Such therapies are reviewed in Sardi & Simuni [2019] and in Shihabuddin et al [2018].

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.

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

LRRK2 Parkinson disease (PD) is inherited in an autosomal dominant manner with reduced penetrance.

Risk to Family Members

Parents of a proband

  • To date, all individuals with LRRK2-PD inherited an LRRK2 pathogenic variant from a parent. However, because penetrance of LRRK2-PD is reduced, a high percentage of probands report unaffected parents.
  • The probability that an asymptomatic parent with a pathogenic variant will become symptomatic increases with age.
  • If the pathogenic variant found in the proband cannot be detected in the leukocyte DNA of either parent, possible explanations include germline mosaicism in a parent or a de novo pathogenic variant in the proband. (See also Related Genetic Counseling Issues, Considerations in families with an apparent de novo pathogenic variant.)
    • Although no instances of germline mosaicism have been reported, it remains a possibility.
    • Similarly, de novo mutation has not been seen, but remains a possibility. Some sites within LRRK2 may be highly mutable – notably, the arginine codon at residue 1441, in which four sometimes recurring amino acid changes have been reported to be pathogenic [Mata et al 2016].
  • The family history of some individuals diagnosed with LRRK2-PD may appear to be negative because of failure to recognize the disorder in family members, early death of the parent before the onset of symptoms, reduced penetrance, or late onset of the disease in the affected parent. Therefore, an apparently negative family history cannot be confirmed unless molecular genetic testing has been performed on the parents of the proband.

Sibs of a proband. The risk to the sibs of the proband depends on the genetic status of the proband's parents:

  • If a parent of the proband is affected or has an LRRK2 pathogenic variant, the risk to the sibs of inheriting the pathogenic variant is 50%. However, the risk to sibs of developing disease is lower than 50% because of reduced, age-related penetrance (see Penetrance).
  • The probability that an asymptomatic sib who has the pathogenic variant will become symptomatic increases with age.

Offspring of a proband

  • Each child of an individual with LRRK2-PD has a 50% chance of inheriting the pathogenic variant.
  • The probability that an offspring with a pathogenic variant will become symptomatic increases with age.

Other family members. The risk to other family members depends on the status of the proband's parents: if a parent is affected or has an LRRK2 pathogenic variant, the parent's family members may be at risk.

Related Genetic Counseling Issues

Testing of asymptomatic at-risk individuals

  • Testing for at-risk relatives is possible once the LRRK2 pathogenic variant has been identified in an affected family member. Such testing is not useful in predicting age of onset, severity, type of symptoms, or rate of progression in asymptomatic individuals.
  • Potential consequences of such testing (including but not limited to socioeconomic changes and the need for long-term follow up and evaluation arrangements for individuals with a positive test result), as well as the capabilities and limitations of such testing, should be discussed in the context of formal genetic counseling prior to testing. The Genetic Information Non-Discrimination Act (GINA) does not provide protection against genetic discrimination for life insurance, long-term insurance, or disability insurance.

Testing of asymptomatic at-risk individuals younger than age 18 years

  • Testing of asymptomatic minors for adult-onset disorders for which treatment of an asymptomatic individual does not decrease morbidity or mortality is not considered appropriate. Such testing negates the autonomy of the child with no compelling benefit. Further, concern exists regarding the potential unhealthy adverse effects that such information may have on family dynamics, the risk of discrimination and stigmatization in the future, and the anxiety that such information may cause.
  • For more information, see the National Society of Genetic Counselors position statement on genetic testing of minors for adult-onset conditions and the American Academy of Pediatrics and American College of Medical Genetics and Genomics policy statement: ethical and policy issues in genetic testing and screening of children.

In a family with an established diagnosis of LRRK2-PD, it is appropriate to consider testing of symptomatic individuals regardless of age.

Considerations in families with an apparent de novo pathogenic variant. When neither parent of a proband with an autosomal dominant condition has the pathogenic variant identified in the proband or clinical evidence of the disorder, the pathogenic variant is likely de novo. However, non-medical explanations including alternate paternity or maternity (e.g., with assisted reproduction) and undisclosed adoption could also be explored.

Family planning

  • The optimal time for determination of genetic risk and discussion of the availability of prenatal/preimplantation genetic testing is before pregnancy. Similarly, decisions about testing to determine the genetic status of at-risk asymptomatic family members are best made 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 or at risk.

Prenatal Testing and Preimplantation Genetic Testing

Once the LRRK2 pathogenic variant has 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.

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.

LRRK2 Parkinson Disease: Genes and Databases

Locus NameGeneChromosome LocusProteinLocus-Specific DatabasesHGMDClinVar
PARK8 LRRK2 12q12 Leucine-rich repeat serine/threonine-protein kinase 2 Parkinson's disease Mutation Database (LRRK2) LRRK2 LRRK2

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 LRRK2 Parkinson Disease (View All in OMIM)

168600PARKINSON DISEASE, LATE-ONSET; PD
607060PARKINSON DISEASE 8, AUTOSOMAL DOMINANT; PARK8
609007LEUCINE-RICH REPEAT KINASE 2; LRRK2

Molecular Pathogenesis

LRRK2, which comprises 144 kb and 51 exons, encodes a leucine-rich repeat serine/threonine-protein kinase 2 (LRRK2). LRRK2 is a 2,527-amino acid protein (286 kd) that shares homology with the Roco family of proteins. The six conserved domains are the following [Bosgraaf & Van Haastert 2003, Mata et al 2006]:

  • Ankyrin repeat
  • Leucine-rich repeat
  • Roc
  • COR
  • Kinase
  • WD40

See Figure 1.

Figure 1. . Schematic representation of the 144-kb LRRK2 loci on chromosome 12q12.

Figure 1.

Schematic representation of the 144-kb LRRK2 loci on chromosome 12q12. The estimated start of the Lrrk2 domain structure is indicated by the residue number (690) below the exonic-intronic (Ex1-Ex51) and domain scheme. Domains: ANK = ankyrin repeat region (more...)

Several biochemical studies have confirmed kinase activity for LRRK2 wild type protein [West et al 2005, Gloeckner et al 2006, Greggio et al 2006, Iaccarino et al 2007]. Additionally, LRRK2 has GTPase activity [Ito et al 2007, Li et al 2007, Liu et al 2016]. Given the size, domain composition, and organization of LRRK2, and the potential for protein-protein interactions, it is likely to be part of a higher molecular-weight complex involved in cellular signaling. Monomers of LRRK2 protein may also dimerize [Guaitoli et al 2016, Civiero et al 2017].

LRRK2 pathogenic variants have been reported through the gene including in the ROC, COR, kinase, and WD40 domains.

Abnormal LRRK2 has been implicated in a number of different pathways [Berwick & Harvey 2011, Berwick & Harvey 2012, Friedman et al 2012, Gómez-Suaga & Hilfiker 2012, Tong et al 2012, Bravo-San Pedro et al 2013, Howlett et al 2017, Lee et al 2017b, Pan et al 2017, Verma et al 2017], including the following:

  • Autophagy
  • Endosomal-lysosomal function
  • Mitochondrial dysfunction
  • Immune signaling microglial motility
  • Synaptic vesicle trafficking and Wnt signaling

Mechanism of disease. In general, pathogenic variants tend to result in overactivity of LRRK2 [Cookson 2015].

While the precise mechanism of action of LRRK2 pathogenic variants is unknown, most (e.g., p.Gly2019Ser) appear to disrupt kinase or GTPase activity. LRRK2 p.Gly2019Ser is within exon 41 and the "activation hinge" of the kinase domain and is associated with increased intra- and intermolecular phosphorylation [Luzón-Toro et al 2007, Anand et al 2009]. While in vitro assays have provided conflicting information on whether enhanced kinase activity represents a characteristic feature shared by all pathogenic LRRK2 variants, several well-known pathogenic variants have been shown to increase phosphorylation of known LRRK2 substrates [Greggio & Cookson 2009, Sheng et al 2012, Steger et al 2016].

LRRK2-specific laboratory technical considerations. Penetrance of LRRK2 pathogenic variants is age dependent and may vary based on pathogenic variant and population ethnicity (including ancestral background and country of origin [Hentati et al 2014]). In addition, some disease-associated LRRK2 variants are common in specific populations. Therefore, interpretation of LRRK2 variants requires careful review of all relevant literature.

Of note, reported risk-factor variants (which are not associated with mendelian disease) include p.Arg1628Pro, p.Ser1647Thr, and p.Gly2385Arg [Oosterveld et al 2015].

Table 3.

Notable LRRK2 Pathogenic Variants

Reference SequencesDNA Nucleotide ChangePredicted Protein ChangeComment [Reference]
NM_198578​.4
NP_940980​.4
c.4321C>Tp.Arg1441CysMay be more common in southern Italian & Belgian populations – see Prevalence
c.4321C>Gp.Arg1441Gly
c.4322G>Ap.Arg1441HisSee Genotype-Phenotype Correlations.
c.4321C>Ap.Arg1441Ser
c.6059T>Cp.Ile2020ThrThe change found in the original LRRK2 family [Funayama et al 2005]
c.6055G>Ap.Gly2019Ser
c.5096A>Gp.Tyr1699CysA range of clinical presentations have been reported – see Genotype-Phenotype Correlations [Kim et al 2012].

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.

Chapter Notes

Author Notes

Dr Saunders-Pullman is the Bachmann-Strauss Chair of Neurology at Mount Sinai. Her research interests focus on the clinical-genetic spectrum of Parkinson disease (PD) and dystonia, including the study of biomarkers and molecular pathways implicated in PD. Ms Raymond is a genetic counselor and clinical research manager. Ms Elango is also a genetic counselor at Mount Sinai. The authors have no financial disclosures.

Acknowledgments

Current research is supported through the National Institutes of Health (NINDS U01-NS107016-01A1U01 U01-NS094148-02), the Bigglesworth Family Foundation, and the Michael J Fox Foundation.

We are grateful to the individuals involved in our research, including the many scientists, clinicians, and especially the patients and their families.

Author History

Sonya Elango, MS (2019-present)
Matthew Farrer, PhD; University of British Columbia (2006-2019)
Ilaria Guella, PhD; University of British Columbia (2014-2019)
Deborah Raymond, MS (2019-present)
Owen A Ross, PhD; Mayo Clinic (2006-2019)
Rachel Saunders-Pullman, MD, MPH (2019-present)
Jeremy T Stone, BSc; Mayo Clinic (2006-2010)
Joanne Trinh, BSc; University of British Columbia (2014-2019)

Revision History

  • 6 July 2023 (sw) Revision: information about LRRK2 pathogenic variants p.Arg1441Gly and p.Arg1441Cys corrected in Prevalence and Molecular Genetics
  • 24 October 2019 (ma) Comprehensive update posted live
  • 11 December 2014 (me) Comprehensive update posted live
  • 13 September 2012 (me) Comprehensive update posted live
  • 29 April 2010 (me) Comprehensive update posted live
  • 2 November 2006 (me) Review posted live
  • 6 July 2006 (mf) Original submission

References

Published Guidelines / Consensus Statements

  • Committee on Bioethics, Committee on Genetics, and American College of Medical Genetics and Genomics Social, Ethical, Legal Issues Committee. Ethical and policy issues in genetic testing and screening of children. Available online. 2013. Accessed 7-3-23.
  • National Society of Genetic Counselors. Position statement on genetic testing of minors for adult-onset conditions. Available online. 2018. Accessed 7-3-23.

Literature Cited

  • Aasly JO, Toft M, Fernandez-Mata I, Kachergus J, Hulihan M, White LR, Farrer M. Clinical features of LRRK2-associated Parkinson's disease in central Norway. Ann Neurol. 2005;57:762–5. [PubMed: 15852371]
  • Agalliu I, Ortega RA, Luciano MS, Mirelman A, Pont-Sunyer C, Brockmann K, Vilas D, Tolosa E, Berg D, Warø B, Glickman A, Raymond D, Inzelberg R, Ruiz-Martinez J, Mondragon E, Friedman E, Hassin-Baer S, Alcalay RN, Mejia-Santana H, Aasly J, Foroud T, Marder K, Giladi N, Bressman S, Saunders-Pullman R. Cancer outcomes among Parkinson's disease patients with leucine rich repeat kinase 2 mutations, idiopathic Parkinson's disease patients, and nonaffected controls. Mov Disord. 2019;34:1392–8. [PMC free article: PMC6754269] [PubMed: 31348549]
  • Agalliu I, San Luciano M, Mirelman A, Giladi N, Waro B, Aasly J, Inzelberg R, Hassin-Baer S, Friedman E, Ruiz-Martinez J, Marti-Masso JF, Orr-Urtreger A, Bressman SB, Saunders-Pullman R. Higher frequency of certain cancers in LRRK2 G2019S mutation carriers with Parkinson disease: a pooled analysis. JAMA Neurology. 2015;72:58–65. [PMC free article: PMC4366130] [PubMed: 25401981]
  • Albrecht M. LRRK2 mutations and Parkinsonism. Lancet. 2005;365:1230. [PubMed: 15811455]
  • Alcalay RN, Mejia-Santana H, Mirelman A, Saunders-Pullman R, Raymond D, Palmese C, Caccappolo E, Ozelius L, Orr-Urtreger A, Clark L, Giladi N, Bressman S, Marder K, et al. Neuropsychological performance in LRRK2 G2019S carriers with Parkinson's disease. Parkinsonism Relat Disord. 2015;21:106–10. [PMC free article: PMC4306614] [PubMed: 25434972]
  • Alcalay RN, Mejia-Santana H, Tang MX, Rosado L, Verbitsky M, Kisselev S, Ross BM, Louis ED, Comella CL, Colcher A, Jennings D, Nance MA, Bressman S, Scott WK, Tanner C, Mickel SF, Andrews HF, Waters CH, Fahn S, Cote LJ, Frucht SJ, Ford B, Rezak M, Novak K, Friedman JH, Pfeiffer R, Marsh L, Hiner B, Siderowf A, Caccappolo E, Ottman R, Clark LN, Marder KS. Motor phenotype of LRRK2 G2019S carriers in early-onset Parkinson disease. Arch Neurol. 2009;66:1517–22. [PMC free article: PMC2837584] [PubMed: 20008657]
  • Alcalay RN, Mirelman A, Saunders-Pullman R, Tang MX, Mejia Santana H, Raymond D, Roos E, Orbe-Reilly M, Gurevich T, Bar Shira A, Gana Weisz M, Yasinovsky K, Zalis M, Thaler A, Deik A, Barrett MJ, Cabassa J, Groves M, Hunt AL, Lubarr N, San Luciano M, Miravite J, Palmese C, Sachdev R, Sarva H, Severt L, Shanker V, Swan MC, Soto-Valencia J, Johannes B, Ortega R, Fahn S, Cote L, Waters C, Mazzoni P, Ford B, Louis E, Levy O, Rosado L, Ruiz D, Dorovski T, Pauciulo M, Nichols W, Orr-Urtreger A, Ozelius L, Clark L, Giladi N, Bressman S, Marder KS. Parkinson disease phenotype in Ashkenazi Jews with and without LRRK2 p.Gly2019Ser mutations. Mov Disord. 2013;28:1966–71. [PMC free article: PMC3859844] [PubMed: 24243757]
  • Anand VS, Reichling LJ, Lipinski K, Stochaj W, Duan W, Kelleher K, Pungaliya P, Brown EL, Reinhart PH, Somberg R, Hirst WD, Riddle SM, Braithwaite SP. Investigation of leucine-rich repeat kinase 2: enzymological properties and novel assays. FEBS J. 2009;276:466–78. [PubMed: 19076219]
  • Artusi CA, Dwivedi A, Romagnolo A, Pal G, Kauffman M, Mata I, Patel D, Vizcarra JA, Duker A, Marsili L, Cheeran B, Woo D, Contarino MF, Verhagen L, Lopiano L, Espay AJ, Fasano A, Merola A. Association of subthalamic deep brain stimulation with motor, functional, and pharmacologic outcomes in patients with monogenic Parkinson disease: a systematic review and meta-analysis. JAMA Netw Open. 2019;2:e187800. [PMC free article: PMC6484599] [PubMed: 30707228]
  • Baptista MA, Dave KD, Frasier MA, Sherer TB, Greeley M, Beck MJ, Varsho JS, Parker GA, Moore C, Churchill MJ, Meshul CK, Fiske BK. Loss of leucine-rich repeat kinase 2 (LRRK2) in rats leads to progressive abnormal phenotypes in peripheral organs. PLoS One. 2013;8:e80705. [PMC free article: PMC3828242] [PubMed: 24244710]
  • Belarbi S, Hecham N, Lesage S, Kediha MI, Smail N, Benhassine T, Ysmail-Dahlouk F, Lohman E, Benhabyles B, Hamadouche T, Assami S, Brice A, Tazir M. LRRK2 G2019S mutation in Parkinson's disease: a neuropsychological and neuropsychiatric study in a large Algerian cohort. Parkinsonism Relat Disord. 2010;16:676–9. [PubMed: 20933457]
  • Ben Romdhan S, Farhat N, Nasri A, Lesage S, Hdiji O, Ben Djebara M, Landoulsi Z, Stevanin G, Brice A, Damak M, Gouider R, Mhiri C. LRRK2 G2019S Parkinson's disease with more benign phenotype than idiopathic. Acta Neurol Scand. 2018;138:425–31. [PubMed: 29989150]
  • Ben Sassi S, Nabli F, Hentati E, Nahdi H, Trabelsi M, Ben Ayed H, Amouri R, Duda JE, Farrer MJ, Hentati F. Cognitive dysfunction in Tunisian LRRK2 associated Parkinson's disease. Parkinsonism Relat Disord. 2012;18:243–6. [PubMed: 22056842]
  • Berwick DC, Harvey K. LRRK2 functions as a Wnt signaling scaffold, bridging cytosolic proteins and membrane-localized LRP6. Hum Mol Genet. 2012;21:4966–79. [PMC free article: PMC3709196] [PubMed: 22899650]
  • Berwick DC, Harvey K. LRRK2 signaling pathways: the key to unlocking neurodegeneration? Trends Cell Biol. 2011;21:257–65. [PubMed: 21306901]
  • Blauwendraat C, Pletnikova O, Geiger JT, Murphy NA, Abramzon Y, Rudow G, Mamais A, Sabir MS, Crain B, Ahmed S, Rosenthal LS, Bakker CC, Faghri F, Chia R, Ding J, Dawson TM, Pantelyat A, Albert MS, Nalls MA, Resnick SM, Ferrucci L, Cookson MR, Hillis AE, Troncoso JC, Scholz SW. Genetic analysis of neurodegenerative diseases in a pathology cohort. Neurobiol Aging. 2019;76:214.e1–214.e9. [PMC free article: PMC6391207] [PubMed: 30528841]
  • Bosgraaf L, Van Haastert PJ. Roc, a Ras/GTPase domain in complex proteins. Biochim Biophys Acta. 2003;1643:5–10. [PubMed: 14654223]
  • Bravo-San Pedro JM, Niso-Santano M, Gómez-Sánchez R, Pizarro-Estrella E, Aiastui-Pujana A, Gorostidi A, Climent V, López de Maturana R, Sanchez-Pernaute R, López de Munain A, Fuentes JM, González-Polo RA. The LRRK2 p.Gly2019Ser mutant exacerbates basal autophagy through activation of the MEK/ERK pathway. Cell Mol Life Sci. 2013;70:121–36. [PubMed: 22773119]
  • Breit S, Wächter T, Schmid-Bielenberg D, Weiss D, Leitner P, Nägele T, Freudenstein D, Gasser T, Krüger R. Effective long-term subthalamic stimulation in PARK8 positive Parkinson's disease. J Neurol. 2010;257:1205–7. [PubMed: 20177695]
  • Bressman S, Saunders-Pullman R. When to start Levodopa therapy for Parkinson's disease. N Engl J Med. 2019;380:389–90. [PubMed: 30673551]
  • Brockmann K, Srulijes K, Hauser AK, Schulte C, Csoti I, Gasser T, Berg D. GBA-associated PD presents with nonmotor characteristics. Neurology. 2011;77:276–80. [PubMed: 21734182]
  • Chen-Plotkin AS, Yuan W, Anderson C, McCarty Wood E, Hurtig HI, Clark CM, Miller BL, Lee VM, Trojanowski JQ, Grossman M, Van Deerlin VM. Corticobasal syndrome and primary progressive aphasia as manifestations of LRRK2 gene mutations. Neurology. 2008;70:521–7. [PMC free article: PMC3619720] [PubMed: 17914064]
  • Civiero L, Russo I, Bubacco L, Greggio E. Molecular insights and functional implication of LRRK2 dimerization. Adv Neurobiol. 2017;14:107–21. [PubMed: 28353281]
  • Cookson MR. LRRK2 pathways leading to neurodegeneration. Curr Neurol Neurosci Rep. 2015;15:42. [PMC free article: PMC5839465] [PubMed: 26008812]
  • Covy JP, Yuan W, Waxman EA, Hurtig HI, Van Deerlin VM, Giasson BI. Clinical and pathological characteristics of patients with leucine-rich repeat kinase-2 mutations. Mov Disord. 2009;24:32–9. [PMC free article: PMC2634827] [PubMed: 19006185]
  • Dächsel JC, Ross OA, Mata IF, Kachergus J, Toft M, Cannon A, Baker M, Adamson J, Hutton M, Dickson DW, Farrer MJ. Lrrk2 p.Gly2019Ser substitution in frontotemporal lobar degeneration with ubiquitin-immunoreactive neuronal inclusions. Acta Neuropathol. 2007;113:601–6. [PubMed: 17151837]
  • Dalvin LA, Damento GM, Yawn BP, Abbott BA, Hodge DO, Pulido JS. Parkinson disease and melanoma: confirming and reexamining an association. Mayo Clin Proc. 2017;92:1070–9. [PMC free article: PMC5682925] [PubMed: 28688464]
  • Di Fonzo A, Tassorelli C, De Mari M, Chien HF, Ferreira J, Rohe CF, Riboldazzi G, Antonini A, Albani G, Mauro A, Marconi R, Abbruzzese G, Lopiano L, Fincati E, Guidi M, Marini P, Stocchi F, Onofrj M, Toni V, Tinazzi M, Fabbrini G, Lamberti P, Vanacore N, Meco G, Leitner P, Uitti RJ, Wszolek ZK, Gasser T, Simons EJ, Breedveld GJ, Goldwurm S, Pezzoli G, Sampaio C, Barbosa E, Martignoni E, Oostra BA, Bonifati V. Comprehensive analysis of the LRRK2 gene in sixty families with Parkinson's disease. Eur J Hum Genet. 2006;14:322–31. [PubMed: 16333314]
  • Domingo A, Klein C. Genetics of Parkinson disease. Handb Clin Neurol. 2018;147:211–27. [PubMed: 29325612]
  • Friedman LG, Lachenmayer ML, Wang J, He L, Poulose SM, Komatsu M, Holstein GR, Yue Z. Disrupted autophagy leads to dopaminergic axon and dendrite degeneration and promotes presynaptic accumulation of α-synuclein and LRRK2 in the brain. J Neurosci. 2012;32:7585–93. [PMC free article: PMC3382107] [PubMed: 22649237]
  • Fu JF, Klyuzhin I, Liu S, Shahinfard E, Vafai N, McKenzie J, Neilson N, Mabrouk R, Sacheli MA, Wile D, McKeown MJ, Stoessl AJ, Sossi V. Investigation of serotonergic Parkinson's disease-related covariance pattern using [11C]-DASB/PET. Neuroimage Clin. 2018;19:652–60. [PMC free article: PMC6014591] [PubMed: 29946508]
  • Fuji RN, Flagella M, Baca M, Baptista MA, Brodbeck J, Chan BK, Fiske BK, Honigberg L, Jubb AM, Katavolos P, Lee DW, Lewin-Koh SC, Lin T, Liu X, Liu S, Lyssikatos JP, O'Mahony J, Reichelt M, Roose-Girma M, Sheng Z, Sherer T, Smith A, Solon M, Sweeney ZK, Tarrant J, Urkowitz A, Warming S, Yaylaoglu M, Zhang S, Zhu H, Estrada AA, Watts RJ. Effect of selective LRRK2 kinase inhibition on nonhuman primate lung. Sci Transl Med. 2015;7:273ra15. [PubMed: 25653221]
  • Funayama M, Hasegawa K, Kowa H, Saito M, Tsuji S, Obata F. A new locus for Parkinson's disease (PARK8) maps to chromosome 12p11.2-q13.1. Ann Neurol. 2002;51:296–301. [PubMed: 11891824]
  • Funayama M, Hasegawa K, Ohta E, Kawashima N, Komiyama M, Kowa H, Tsuji S, Obata F. An LRRK2 mutation as a cause for the parkinsonism in the original PARK8 family. Ann Neurol. 2005;57:918–21. [PubMed: 15880653]
  • Gan-Or Z, Bar-Shira A, Mirelman A, Gurevich T, Kedmi M, Giladi N, Orr-Urtreger A. LRRK2 and GBA mutations differentially affect the initial presentation of Parkinson disease. Neurogenetics. 2010;11:121–5. [PubMed: 19458969]
  • Gan-Or Z, Leblond CS, Mallett V, Orr-Urtreger A, Dion PA, Rouleau GA. LRRK2 mutations in Parkinson disease; a sex effect or lack thereof? A meta-analysis. Parkinsonism Relat Disord. 2015;21:778–82. [PubMed: 25962553]
  • Gaig C, Vilas D, Infante J, Sierra M, García-Gorostiaga I, Buongiorno M, Ezquerra M, Martí MJ, Valldeoriola F, Aguilar M, Calopa M, Hernandez-Vara J, Tolosa E. Nonmotor symptoms in LRRK2 G2019S associated Parkinson's disease. PLoS One. 2014;9:e108982. [PMC free article: PMC4201457] [PubMed: 25330404]
  • Giladi N, Mirelman A, Thaler A, Orr-Utreger A. A personalized approach to Parkinson's disease patients based on a founder mutation analysis. Front Neurol. 2016;7:71. [PMC free article: PMC4861838] [PubMed: 27242656]
  • Gloeckner CJ, Kinkl N, Schumacher A, Braun RJ, O'Neill E, Meitinger T, Kolch W, Prokisch H, Ueffing M. The Parkinson disease causing LRRK2 mutation I2020T is associated with increased kinase activity. Hum Mol Genet. 2006;15:223–32. [PubMed: 16321986]
  • Goetz CG, Nutt JG, Stebbins GT. The Unified Dyskinesia Rating Scale: presentation and clinimetric profile. Mov Disord. 2008;23:2398–403. [PubMed: 19025759]
  • Goldwurm S, Zini M, Di Fonzo A, De Gaspari D, Siri C, Simons EJ, van Doeselaar M, Tesei S, Antonini A, Canesi M, Zecchinelli A, Mariani C, Meucci N, Sacilotto G, Cilia R, Isaias IU, Bonetti A, Sironi F, Ricca S, Oostra BA, Bonifati V, Pezzoli G. LRRK2 p.Gly2019Ser mutation and Parkinson's disease: a clinical, neuropsychological and neuropsychiatric study in a large Italian sample. Parkinsonism Relat Disord. 2006;12:410–9. [PubMed: 16750929]
  • Goldwurm S, Zini M, Mariani L, Tesei S, Miceli R, Sironi F, Clementi M, Bonifati V, Pezzoli G. Evaluation of LRRK2 p.Gly2019Ser penetrance: relevance for genetic counseling in Parkinson disease. Neurology. 2007;68:1141–3. [PubMed: 17215492]
  • Gómez-Suaga P, Hilfiker S. LRRK2 as a modulator of lysosomal calcium homeostasis with downstream effects on autophagy. Autophagy. 2012;8:692–3. [PubMed: 22441017]
  • Greenbaum L, Israeli-Korn SD, Cohen OS, Elincx-Benizri S, Yahalom G, Kozlova E, Strauss H, Molshatzki N, Inzelberg R, Spiegelmann R, Israel Z, Hassin-Baer S. The LRRK2 G2019S mutation status does not affect the outcome of subthalamic stimulation in patients with Parkinson's disease. Parkinsonism Relat Disord. 2013;19:1053–6. [PubMed: 23932063]
  • Greggio E, Cookson MR. Leucine-rich repeat kinase 2 mutations and Parkinson's disease: three questions. ASN Neuro. 2009;1:e00002. [PMC free article: PMC2695577] [PubMed: 19570025]
  • Greggio E, Jain S, Kingsbury A, Bandopadhyay R, Lewis P, Kaganovich A, van der Brug MP, Beilina A, Blackinton J, Thomas KJ, Ahmad R, Miller DW, Kesavapany S, Singleton A, Lees A, Harvey RJ, Harvey K, Cookson MR. Kinase activity is required for the toxic effects of mutant LRRK2/dardarin. Neurobiol Dis. 2006;23:329–41. [PubMed: 16750377]
  • Guaitoli G, Raimondi F, Gilsbach BK, Gómez-Llorente Y, Deyaert E, Renzi F, Li X, Schaffner A, Jagtap PK, Boldt K, von Zweydorf F, Gotthardt K, Lorimer DD, Yue Z, Burgin A, Janjic N, Sattler M, Versées W, Ueffing M, Ubarretxena-Belandia I, Kortholt A, Gloeckner CJ. Structural model of the dimeric Parkinson's protein LRRK2 reveals a compact architecture involving distant interdomain contacts. Proc Natl Acad Sci U S A. 2016;113:E4357–66. [PMC free article: PMC4968714] [PubMed: 27357661]
  • Haugarvoll K, Rademakers R, Kachergus JM, Nuytemans K, Ross OA, Gibson JM, Tan EK, Gaig C, Tolosa E, Goldwurm S, Guidi M, Riboldazzi G, Brown L, Walter U, Benecke R, Berg D, Gasser T, Theuns J, Pals P, Cras P, De Deyn PP, Engelborghs S, Pickut B, Uitti RJ, Foroud T, Nichols WC, Hagenah J, Klein C, Samii A, Zabetian CP, Bonifati V, Van Broeckhoven C, Farrer MJ, Wszolek ZK. Lrrk2 R1441C parkinsonism is clinically similar to sporadic Parkinson disease. Neurology. 2008;70:1456–60. [PMC free article: PMC3906630] [PubMed: 18337586]
  • Healy DG, Falchi M, O'Sullivan SS, Bonifati V, Durr A, Bressman S, Brice A, Aasly J, Zabetian CP, Goldwurm S, Ferreira JJ, Tolosa E, Kay DM, Klein C, Williams DR, Marras C, Lang AE, Wszolek ZK, Berciano J, Schapira AH, Lynch T, Bhatia KP, Gasser T, Lees AJ, Wood NW. International LRRK2 Consortium. Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson's disease: a case-control study. Lancet Neurol. 2008;7:583–90. [PMC free article: PMC2832754] [PubMed: 18539534]
  • Hentati F, Trinh J, Thompson C, Nosova E, Farrer MJ, Aasly JO. LRRK2 parkinsonism in Tunisia and Norway: a comparative analysis of disease penetrance. Neurology. 2014;83:568–9. [PMC free article: PMC4142000] [PubMed: 25008396]
  • Herzig MC, Kolly C, Persohn E, Theil D, Schweizer T, Hafner T, Stemmelen C, Troxler TJ, Schmid P, Danner S, Schnell CR, Muelle M, Kinzel B, Grevot A, Bolognani F, Stirn M, Kuhn RR, Kaupmann K, van der Putten PH, Rovelli G, Shimshek DR. LRRK2 protein levels are determined by kinase function and are crucial for kidney and lung homeostasis in mice. Hum Mol Genet. 2011;20:4209–23. [PMC free article: PMC3188995] [PubMed: 21828077]
  • Howlett EH, Jensen N, Belmonte F, Zafar F, Hu X, Kluss J, Schüle B, Kaufman BA, Greenamyre JT, Sanders LH. LRRK2 p.Gly2019Ser-induced mitochondrial DNA damage is LRRK2 kinase dependent and inhibition restores mtDNA integrity in Parkinson's disease. Hum Mol Genet. 2017;26:4340–51. [PMC free article: PMC5886254] [PubMed: 28973664]
  • Huang P, Yang XD, Chen SD, Xiao Q. The association between Parkinson's disease and melanoma: a systematic review and meta-analysis. Transl Neurodegener. 2015;4:21. [PMC free article: PMC4631109] [PubMed: 26535116]
  • Hulihan MM, Ishihara-Paul L, Kachergus J, Warren L, Amouri R, Elango R, Prinjha RK, Upmanyu R, Kefi M, Zouari M, Sassi SB, Yahmed SB, El Euch-Fayeche G, Matthews PM, Middleton LT, Gibson RA, Hentati F, Farrer MJ. LRRK2 Gly2019Ser penetrance in Arab-Berber patients from Tunisia: a case-control genetic study. Lancet Neurol. 2008;7:591–4. [PubMed: 18539535]
  • Iaccarino C, Crosio C, Vitale C, Sanna G, Carrì MT, Barone P. Apoptotic mechanisms in mutant LRRK2-mediated cell death. Hum Mol Genet. 2007;16:1319–26. [PubMed: 17409193]
  • Inzelberg R, Cohen OS, Aharon-Peretz J, Schlesinger I, Gershoni-Baruch R, Djaldetti R, Nitsan Z, Ephraty L, Tunkel O, Kozlova E, Inzelberg L, Kaplan N, Fixler Mehr T, Mory A, Dagan E, Schechtman E, Friedman E, Hassin-Baer S. The LRRK2 G2019S mutation is associated with Parkinson disease and concomitant non-skin cancers. Neurology. 2012;78:781–6. [PubMed: 22323743]
  • Ishihara L, Warren L, Gibson R, Amouri R, Lesage S, Durr A, Tazir M, Wszolek ZK, Uitti RJ, Nichols WC, Griffith A, Hattori N, Leppert D, Watts R, Zabetian CP, Foroud TM, Farrer MJ, Brice A, Middleton L, Hentati F. Clinical features of Parkinson disease patients with homozygous leucine-rich repeat kinase 2 p.Gly2019Ser mutations. Arch Neurol. 2006;63:1250–4. [PubMed: 16966502]
  • Ito G, Okai T, Fujino G, Takeda K, Ichijo H, Katada T, Iwatsubo T. GTP binding is essential to the protein kinase activity of LRRK2, a causative gene product for familial Parkinson's disease. Biochemistry. 2007;46:1380–8. [PubMed: 17260967]
  • Jaleel M, Nichols RJ, Deak M, Campbell DG, Gillardon F, Knebel A, Alessi DR. LRRK2 phosphorylates moesin at threonine-558: characterization of how Parkinson's disease mutants affect kinase activity. Biochem J. 2007;405:307–17. [PMC free article: PMC1904520] [PubMed: 17447891]
  • Kachergus J, Mata IF, Hulihan M, Taylor JP, Lincoln S, Aasly J, Gibson JM, Ross OA, Lynch T, Wiley J, Payami H, Nutt J, Maraganore DM, Czyzewski K, Styczynska M, Wszolek ZK, Farrer MJ, Toft M. Identification of a novel LRRK2 mutation linked to autosomal dominant parkinsonism: evidence of a common founder across European populations. Am J Hum Genet. 2005;76:672–80. [PMC free article: PMC1199304] [PubMed: 15726496]
  • Kalia LV, Lang AE, Hazrati LN, Fujioka S, Wszolek ZK, Dickson DW, Ross OA, Van Deerlin VM, Trojanowski JQ, Hurtig HI, Alcalay RN, Marder KS, Clark LN, Gaig C, Tolosa E, Ruiz-Martínez J, Marti-Masso JF, Ferrer I, López de Munain A, Goldman SM, Schüle B, Langston JW, Aasly JO, Giordana MT, Bonifati V, Puschmann A, Canesi M, Pezzoli G, Maues De Paula A, Hasegawa K, Duyckaerts C, Brice A, Stoessl AJ, Marras C. Clinical correlations with Lewy body pathology in LRRK2-related Parkinson disease. JAMA Neurol. 2015;72:100–5. [PMC free article: PMC4399368] [PubMed: 25401511]
  • Kestenbaum M, Alcalay RN. Clinical features of LRRK2 carriers with Parkinson's disease. Adv Neurobiol. 2017;14:31–48. [PubMed: 28353277]
  • Khan NL, Jain S, Lynch JM, Pavese N, Abou-Sleiman P, Holton JL, Healy DG, Gilks WP, Sweeney MG, Ganguly M, Gibbons V, Gandhi S, Vaughan J, Eunson LH, Katzenschlager R, Gayton J, Lennox G, Revesz T, Nicholl D, Bhatia KP, Quinn N, Brooks D, Lees AJ, Davis MB, Piccini P, Singleton AB, Wood NW. Mutations in the gene LRRK2 encoding dardarin (PARK8) cause familial Parkinson's disease: clinical, pathological, olfactory and functional imaging and genetic data. Brain. 2005;128:2786–96. [PubMed: 16272164]
  • Kim JS, Cho JW, Shin H, Lee WY, Ki CS, Cho AR, Kim HT. A Korean Parkinson's disease family with the LRRK2 p.Tyr1699Cys mutation showing clinical heterogeneity. Mov Disord. 2012;27:320–4. [PubMed: 22162019]
  • Klein C, Westenberger A. Genetics of Parkinson's disease. Cold Spring Harb Perspect Med. 2012;2:a008888. [PMC free article: PMC3253033] [PubMed: 22315721]
  • Kuusimäki T, Korpela J, Pekkonen E, Martikainen MH, Antonini A, Kaasinen V. Deep brain stimulation for monogenic Parkinson disease: a systematic review. J Neurol. 2020;267:883–97. [PMC free article: PMC7109183] [PubMed: 30659355]
  • Leaver K, Miravite J, Dilli C, Sa B, Ortega RP, Moran E, Kopell B, Okun MS, Foote KD, Shanker V, Bressman SB, Saunders-Pullman RJ. Longitudinal outcomes of LRRK2 gene mutation parkinsonism undergoing either subthalamic nucleus (STN) or globus pallidus internus (GPi) deep brain stimulation (DBS): a multigroup comparison. 2019. American Academy of Neurology 2019 Annual Meeting, May 4-10.
  • Lee AJ, Wang Y, Alcalay RN, Mejia-Santana H, Saunders-Pullman R, Bressman S, Corvol JC, Brice A, Lesage S, Mangone G, Tolosa E, Pont-Sunyer C, Vilas D, Schüle B, Kausar F, Foroud T, Berg D, Brockmann K, Goldwurm S, Siri C, Asselta R, Ruiz-Martinez J, Mondragón E, Marras C, Ghate T, Giladi N, Mirelman A, Marder K. Penetrance estimate of LRRK2 p.Gly2019Ser mutation in individuals of non-Ashkenazi Jewish ancestry. Mov Disord. 2017a;32:1432–8. [PMC free article: PMC5656509] [PubMed: 28639421]
  • Lee BD, Shin JH, VanKampen J, Petrucelli L, West AB, Ko HS, Lee YI, Maguire-Zeiss KA, Bowers WJ, Federoff HJ, Dawson VL, Dawson TM. Inhibitors of leucine-rich repeat kinase-2 protect against models of Parkinson's disease. Nat Med. 2010;16:998–1000. [PMC free article: PMC2935926] [PubMed: 20729864]
  • Lee H, James WS, Cowley SA. LRRK2 in peripheral and central nervous system innate immunity: its link to Parkinson's disease. Biochem Soc Trans. 2017b;45:131–9. [PMC free article: PMC5652224] [PubMed: 28202666]
  • Lee JM, Kim TW, Park SS, Han JH, Shin MS, Lim BV, Kim SH, Baek SS, Cho YS, Kim KH. Treadmill exercise improves motor function by suppressing Purkinje cell loss in Parkinson disease rats. Int Neurourol J. 2018a;22:S147–55. [PMC free article: PMC6234730] [PubMed: 30396264]
  • Lee K, Nguyen KD, Sun C, Liu M, Zafar F, Saetern J, Flierl A, Tetrud JW, Langston JW, Dickson D, Schüle B. LRRK2 p.Ile1371Val mutation in a case with neuropathologically confirmed multi-system atrophy. J Parkinsons Dis. 2018b;8:93–100. [PubMed: 29480226]
  • Li X, Tan YC, Poulose S, Olanow CW, Huang XY, Yue Z. Leucine-rich repeat kinase 2 (LRRK2)/PARK8 possesses GTPase activity that is altered in familial Parkinson's disease R1441C/G mutants. J Neurochem. 2007;103:238–47. [PMC free article: PMC2827244] [PubMed: 17623048]
  • Lin CH, Tzen KY, Yu CY, Tai CH, Farrer MJ, Wu RM. LRRK2 mutation in familial Parkinson's disease in a Taiwanese population: clinical, PET, and functional studies. J Biomed Sci. 2008;15:661–7. [PubMed: 18523869]
  • Liu Z, Mobley JA, DeLucas LJ, Kahn RA, West AB. LRRK2 autophosphorylation enhances its GTPase activity. FASEB J. 2016;30:336–47. [PMC free article: PMC4684519] [PubMed: 26396237]
  • Luzón-Toro B, Rubio de la Torre E, Delgado A, Pérez-Tur J, Hilfiker S. Mechanistic insight into the dominant mode of the Parkinson's disease-associated G2019S LRRK2 mutation. Hum Mol Genet. 2007;16:2031–9. [PubMed: 17584768]
  • MacLeod D, Dowman J, Hammond R, Leete T, Inoue K, Abeliovich A. The familial Parkinsonism gene LRRK2 regulates neurite process morphology. Neuron. 2006;52:587–93. [PubMed: 17114044]
  • Marder K, Wang Y, Alcalay RN, Mejia-Santana H, Tang MX, Lee A, Raymond D, Mirelman A, Saunders-Pullman R, Clark L, Ozelius L, Orr-Urtreger A, Giladi N, Bressman S. Age-specific penetrance of LRRK2 p.Gly2019Ser in the Michael J. Fox Ashkenazi Jewish LRRK2 Consortium. Neurology. 2015;85:89–95. [PMC free article: PMC4501942] [PubMed: 26062626]
  • Marras C, Alcalay RN, Caspell-Garcia C, Coffey C, Chan P, Duda JE, Facheris MF, Fernández-Santiago R, Ruíz-Martínez J, Mestre T, Saunders-Pullman R, Pont-Sunyer C, Tolosa E, Waro B, et al. Motor and nonmotor heterogeneity of LRRK2-related and idiopathic Parkinson's disease. Mov Disord. 2016;31:1192–202. [PubMed: 27091104]
  • Marras C, Schüle B, Munhoz RP, Rogaeva E, Langston JW, Kasten M, Meaney C, Klein C, Wadia PM, Lim SY, Chuang RS, Zadikof C, Steeves T, Prakash KM, de Bie RM, Adeli G, Thomsen T, Johansen KK, Teive HA, Asante A, Reginold W, Lang AE. Phenotype in parkinsonian and nonparkinsonian LRRK2 G2019S mutation carriers. Neurology. 2011;77:325–33. [PMC free article: PMC3140802] [PubMed: 21753163]
  • Mata IF, Davis MY, Lopez AN, Dorschner MO, Martinez E, Yearout D, Cholerton BA, Hu SC, Edwards KL, Bird TD, Zabetian CP. The discovery of LRRK2 p.R1441S, a novel mutation for Parkinson's disease, adds to the complexity of a mutational hotspot. Am J Med Genet B Neuropsychiatr Genet. 2016;171:925–30. [PMC free article: PMC5028305] [PubMed: 27111571]
  • Mata IF, Kachergus JM, Taylor JP, Lincoln S, Aasly J, Lynch T, Hulihan MM, Cobb SA, Wu RM, Lu CS, Lahoz C, Wszolek ZK, Farrer MJ. Lrrk2 pathogenic substitutions in Parkinson's disease. Neurogenetics. 2005a;6:171–7. [PubMed: 16172858]
  • Mata IF, Taylor JP, Kachergus J, Hulihan M, Huerta C, Lahoz C, Blazquez M, Guisasola LM, Salvador C, Ribacoba R, Martinez C, Farrer M, Alvarez V. LRRK2 R1441G in Spanish patients with Parkinson's disease. Neurosci Lett. 2005b;382:309–11. [PubMed: 15925109]
  • Mata IF, Wedemeyer WJ, Farrer MJ, Taylor JP, Gallo KA. LRRK2 in Parkinson's disease: protein domains and functional insights. Trends Neurosci. 2006;29:286–93. [PubMed: 16616379]
  • Mirelman A, Heman T, Yasinovsky K, Thaler A, Gurevich T, Marder K, Bressman S, Bar-Shira A, Orr-Urtreger A, Giladi N, Hausdorff JM, et al. Fall risk and gait in Parkinson's disease: the role of the LRRK2 p.Gly2019Ser mutation. Mov Disord. 2013;28:1683–90. [PubMed: 24123150]
  • Nabli F, Ben Sassi S, Amouri R, Duda JE, Farrer MJ, Hentati F. Motor phenotype of LRRK2-associated Parkinson's disease: a Tunisian longitudinal study. Mov Disord. 2015;30:253–8. [PubMed: 25487881]
  • Olanow CW, Stocchi I. Levodopa: a new look at an old friend. Mov Disord. 2018;33:859–66. [PubMed: 29178365]
  • Oosterveld LP, Allen JC, Ng EY, Seah SH, Tay KY, Au WL, Tan EK, Tan LC. Greater motor progression in patients with Parkinson disease who carry LRRK2 variants. Neurology. 2015;85:1039–42. [PubMed: 26311745]
  • Ozelius LJ, Senthil G, Saunders-Pullman R, Ohmann E, Deligtisch A, Tagliati M, Hunt AL, Klein C, Henick B, Hailpern SM, Lipton RB, Soto-Valencia J, Risch N, Bressman SB. LRRK2 p.Gly2019Ser as a cause of Parkinson's disease in Ashkenazi Jews. N Engl J Med. 2006;354:424–5. [PubMed: 16436782]
  • Paisán-Ruíz C, Jain S, Evans EW, Gilks WP, Simon J, van der Brug M, Lopez de Munain A, Aparicio S, Gil AM, Khan N, Johnson J, Martinez JR, Nicholl D, Carrera IM, Pena AS, de Silva R, Lees A, Marti-Masso JF, Perez-Tur J, Wood NW, Singleton AB. Cloning of the gene containing mutations that cause PARK8-linked Parkinson's disease. Neuron. 2004;44:595–600. [PubMed: 15541308]
  • Pan PY, Li X, Wang J, Powell J, Wang Q, Zhang Y, Chen Z, Wicinski B, Hof P, Ryan TA, Yue Z. Parkinson's disease-associated LRRK2 hyperactive kinase mutant disrupts synaptic vesicle trafficking in ventral midbrain neurons. J Neurosci. 2017;37:11366–76. [PMC free article: PMC5700420] [PubMed: 29054882]
  • Parkinson's Study Group. Pramipexole vs levodopa as initial treatment for Parkinson disease: a randomized clinical trial. JAMA. 2000;284:1931–8. [PubMed: 11035889]
  • Pont-Sunyer C, Iranzo A, Gaig C, Fernández-Arcos A, Vilas D, Valldeoriola F, Compta Y, Fernández-Santiago R, Fernández M, Bayés A, Calopa M, Casquero P, de Fàbregues O, Jaumà S, Puente V, Salamero M, José Martí M, Santamaría J, Tolosa E. Sleep disorders in parkinsonian and nonparkinsonian LRRK2 mutation carriers. PLoS One. 2015;10:e0132368. [PMC free article: PMC4503402] [PubMed: 26177462]
  • Postuma RB, Berg D, Stern M, Poewe W, Olanow CW, Oertel W, et al. MDS clinical diagnostic criteria for Parkinson's disease. Mov Disord. 2015;30:1591–601. [PubMed: 26474316]
  • Quattrone A, Bagnato A, Annesi G, Novellino F, Morgante L, Savettieri G, Zappia M, Tarantino P, Candiano IC, Annesi F, Civitelli D, Rocca FE, D'Amelio M, Nicoletti G, Morelli M, Petrone A, Loizzo P, Condino F. Myocardial 123metaiodobenzylguanidine uptake in genetic Parkinson's disease. Mov Disord. 2008;23:21–7. [PubMed: 17975812]
  • 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]
  • Ross OA, Toft M, Whittle AJ, Johnson JL, Papapetropoulos S, Mash DC, Litvan I, Gordon MF, Wszolek ZK, Farrer MJ, Dickson DW. Lrrk2 and Lewy body disease. Ann Neurol. 2006;59:388–93. [PubMed: 16437559]
  • Ruiz-Martínez J, Gorostidi A, Goyenechea E, Alzualde A, Poza JJ, Rodríguez F, Bergareche A, Moreno F, López de Munain A, Martí Massó JF. Olfactory deficits and cardiac 123I-MIBG in Parkinson's disease related to the LRRK2 R1441G and G2019S mutations. Mov Disord. 2011;26:2026–31. [PubMed: 21611983]
  • Ruiz-Martínez J, Gorostidi A, Ibañez B, Alzualde A, Otaegui D, Moreno F, López de Munain A, Bergareche A, Gómez-Esteban JC, Martí Massó JF. Penetrance in Parkinson's disease related to the LRRK2 R1441G mutation in the Basque country (Spain). Mov Disord. 2010;25:2340–5. [PubMed: 20721916]
  • San Luciano M, Lipton RB, Wang C, Sanders A, Ozelius LO, Bressman SB, Saunders-Pullman R. Clinical expression of LRRK2 G2019S mutations in the elderly. Mov Disord. 2010;25:2571–6. [PMC free article: PMC2978804] [PubMed: 20721910]
  • San Luciano M, Wang C, Ortega R, Giladi N, Marder K, Bressman S, Saunders-Pullman R, et al. Sex differences in LRRK2 G2019S and idiopathic Parkinson's disease. Ann Clin Transl Neurol. 2017;4:801–10. [PMC free article: PMC5682117] [PubMed: 29159192]
  • Santos-Rebouças CB, Abdalla CB, Baldi FJ, Martins PA, Corrêa JC, Gonçalves AP, Cunha MS, Borges MB, Pereira JS, Laks J, Pimentel MM. Co-occurrence of sporadic parkinsonism and late-onset Alzheimer's disease in a Brazilian male with the LRRK2 p.p.Gly2019Ser mutation. Genet Test. 2008;12:471–3. [PubMed: 19072560]
  • Sardi SP, Simuni T. New era in disease modification in Parkinson's disease: review of genetically targeted therapeutics. Parkinsonism Relat Disord. 2019;59:32–8. [PubMed: 30391183]
  • Saunders-Pullman R, Barrett M, Stanley K, San Luciano M, Shanker V, Severt L, Hunt A, Raymond D, Ozelius LJ, Bressman SB. LRRK2 G2019S mutations are associated with an increased risk of cancer in Parkinson disease. Mov Disord. 2010;25:2536–41. [PMC free article: PMC2978749] [PubMed: 20818610]
  • Saunders-Pullman R, Mirelman A, Alcalay RN, Wang C, Ortega RA, Raymond D, Mejia-Santana H, Orbe-Reilly M, Johannes BA, Thaler A, Ozelius L, Orr-Urtreger A, Marder KS, Giladi N, Bressman SB, et al. Progression in the LRRK2-asssociated Parkinson disease population. JAMA Neurol. 2018;75:312–9. [PMC free article: PMC5885854] [PubMed: 29309488]
  • Saunders-Pullman R, Mirelman A, Wang C, Alcalay R, San Luciano M, Ortega R, Raymond D, Mejia-Santana H, Ozelius LO, Clark L, Orr-Utreger A, Marder K, Giladi N, Bressman SB. Olfactory identification in LRRK2 G2019S mutation carriers: a relevant marker? Ann Clin Transl Neurol. 2014;1:670–8. [PMC free article: PMC4241794] [PubMed: 25493281]
  • Saunders-Pullman R, Stanley K, Wang C, San Luciano M, Raymond D, Ozelius LJ, Lipton RB, Bressman SB. Olfactory dysfunction in LRRK2 G2019S mutation carriers. Neurology. 2011;77:319–24. [PMC free article: PMC3140803] [PubMed: 21753159]
  • Seier M, Hiller A. Parkinson's disease and pregnancy: an updated review. Parkinsonism Relat Disord. 2017;40:11–7. [PubMed: 28506531]
  • Shanker V, Groves M, Heiman G, Saunders-Pullman R, Ozelius L, Palmese C, Raymond D, Bressman SB. Mood and cognition in leucine-rich repeat kinase 2 G2019S Parkinson's disease. Mov Disord. 2011;26:1875–80. [PMC free article: PMC3972755] [PubMed: 21611978]
  • Sheng Z, Zhang S, Bustos D, Kleinheinz T, Le Pichon CE, Dominguez SL, Solanoy HO, Drummond J, Zhang X, Ding X, Cai F, Song Q, Li X, Yue Z, van der Brug MP, Burdick DJ, Gunzner-Toste J, Chen H, Liu X, Estrada AA, Sweeney ZK, Scearce-Levie K, Moffat JG, Kirkpatrick DS, Zhu H. Ser1292 autophosphorylation is an indicator of LRRK2 kinase activity and contributes to the cellular effects of PD mutations. Sci Transl Med. 2012;4:164ra161. [PubMed: 23241745]
  • Shihabuddin LS, Brundin P, Greenamyre T. New frontiers in Parkinson's disease: from genetics to the clinic. J Neurosci. 2018;38:9375–82. [PMC free article: PMC6705997] [PubMed: 30381429]
  • Silveira-Moriyama L, Munhoz RP, Carvalho M, Raskin S, Rogaeva E. de C Aguiar P, Bressan RA, Felicio AC, Barsottini OG, Andrade LA, Chien HF, Bonifati V, Barbosa ER, Teive HA, Lees AJ. Olfactory heterogeneity in LRRK2 related Parkinsonism. Mov Disord. 2010;25:2879–83. [PubMed: 20818658]
  • Smith WW, Pei Z, Jiang H, Dawson VL, Dawson TM, Ross CA. Kinase activity of mutant LRRK2 mediates neuronal toxicity. Nat Neurosci. 2006;9:1231–3. [PubMed: 16980962]
  • Somme JH, Molano Salazar A, Gonzalez A, Tijero B, Berganzo K, Lezcano E, Fernandez Martinez M, Zarranz JJ, Gómez-Esteban JC. Cognitive and behavioral symptoms in Parkinson's disease patients with G2019S and R1441G mutations of the LRRK2 gene. Parkinsonism Relat Disord. 2015;21:494–9. [PubMed: 25840672]
  • Srivatsal S, Cholerton B, Leverenz JB, Wszolek ZK, Uitti RJ, Dickson DW, Weintraub D, Trojanowski JQ, Van Deerlin VM, Quinn JF, Chung KA, Peterson AL, Factor SA, Wood-Siverio C, Goldman JG, Stebbins GT, Bernard B, Ritz B, Rausch R, Espay AJ, Revilla FJ, Devoto J, Rosenthal LS, Dawson TM, Albert MS, Mata IF, Hu SC, Montine KS, Johnson C, Montine TJ, Edwards KL, Zhang J, Zabetian CP. Cognitive profile of LRRK2-related Parkinson's disease. Mov Disord. 2015;30:728–33. [PMC free article: PMC4397146] [PubMed: 25650144]
  • Stefani A, Marzetti F, Pierantozzi M, Petrucci S, Olivola E, Galati S, Bassi MS, Imbriani P, Valente EM, Pastore FS. Successful subthalamic stimulation, but levodopa-induced dystonia, in a genetic Parkinson's disease. Neurol Sci. 2013;34:383–6. [PubMed: 22437494]
  • Steger M, Tonelli F, Ito G, Davies P, Trost M, Vetter M, Wachter S, Lorentzen E, Duddy G, Wilson S, Baptista MA, Fiske BK, Fell MJ, Morrow JA, Reith AD, Alessi DR, Mann M. Phosphoproteomics reveals that Parkinson's disease kinase LRRK2 regulates a subset of Rab GTPases. Elife. 2016;5:e12813. [PMC free article: PMC4769169] [PubMed: 26824392]
  • Stocchi F, Rascol O, Kieburtz K, Poewe W, Jankovic J, Tolosa E, Barone P, Lang AE, Olanow CW. Initiating levodopa/carbidopa therapy with and without entacapone in early Parkinson disease: the STRIDE PD study. Ann Neurol. 2010;68:18–27. [PubMed: 20582993]
  • Thaler A, Kozlovski T, Gurevich T, Bar-Shira A, Gana-Weisz M, Orr-Urtreger A, Giladi N, Mirelman A. Survival rates among Parkinson's disease patients who carry mutations in the LRRK2 and GBA genes. Mov Disord. 2018;33:1656–60. [PubMed: 30288804]
  • Tijero B, Gómez Esteban JC, Somme J, Llorens V, Lezcano E, Martinez A, Rodríguez T, Berganzo K, Zarranz JJ. Autonomic dysfunction in parkinsonian LRRK2 mutation carriers. Parkinsonism Relat Disord. 2013;19:906–9. [PubMed: 23764467]
  • Toft M, Mata IF, Kachergus JM, Ross OA, Farrer MJ. LRRK2 mutations and Parkinsonism. Lancet. 2005;365:1229–30. [PubMed: 15811454]
  • Tong Y, Giaime E, Yamaguchi H, Ichimura T, Liu Y, Si H, Cai H, Bonventre JV, Shen J. Loss of leucine-rich repeat kinase 2 causes age-dependent bi-phasic alterations of the autophagy pathway. Mol Neurodegener. 2012;7:2. [PMC free article: PMC3296570] [PubMed: 22230652]
  • Trinh J, Amouri R, Duda JE, Morley JF, Read M, Donald A, Vilarino-Guell C, Thompson C, Szu Tu C, Gustavsson EK, Sassi SB, Hentati E, Zouari M, Farhat E, Nabli F, Hentati F, Farrer MJ. A comparative study of Parkinson's disease and leucine-rich repeat kinase 2 p.p.Gly2019Ser parkinsonism. Neurobiol Aging. 2014a;35:1125–31. [PubMed: 24355527]
  • Trinh J, Guella I, Farrer M. Disease penetrance of late-onset parkinsonism: a meta-analysis. JAMA Neurol. 2014b;71:1535–9. [PubMed: 25330418]
  • Trinh J, Gustavsson EK, Vilariño-Güell C, Bortnick S, Latourelle J, McKenzie MB, Tu CS, Nosova E, Khinda J, Milnerwood A, Lesage S, Brice A, Tazir M, Aasly JO, Parkkinen L, Haytural H, Foroud T, Myers RH, Sassi SB, Hentati E, Nabli F, Farhat E, Amouri R, Hentati F, Farrer MJ. DNM3 and genetic modifiers of age of onset in LRRK2 Gly2019Ser parkinsonism: a genome-wide linkage and association study. Lancet Neurol. 2016;15:1248–56. [PubMed: 27692902]
  • Trinh J, Zeldenrust FMJ, Huang J, Kasten M, Schaake S, Petkovic S, Madoev H, Grünewald A, Almuammar S, König I, Lill CM, Lohmann K, Klein C, Marras C. Genotype-phenotype relations for the Parkinson's disease genes SNCA, LRRK2, VPS35: MDSGene systematic review. Mov Disord. 2018;33:1857–70. [PubMed: 30357936]
  • Ujiie S, Hatano T, Kubo S, Imai S, Sato S, Uchihara T, Yagishita S, Hasegawa K, Kowa H, Sakai F, Hattori N. LRRK2 I2020T mutation is associated with tau pathology. Parkinsonism Relat Disord. 2012;18:819–23. [PubMed: 22525366]
  • Verma M, Callio J, Otero PA, Sekler I, Wills ZP, Chu CT. Mitochondrial calcium dysregulation contributes to dendrite degeneration mediated by PD/LBD-associated LRRK2 mutants. J Neurosci. 2017;37:11151–65. [PMC free article: PMC5688524] [PubMed: 29038245]
  • Visanji NP, Bhudhikanok GS, Mestre TA, Ghate T, Udupa K, AlDakheel A, Connolly BS, Gasca-Salas C, Kern DS, Jain J, Slow EJ, Faust-Socher A, Kim S, Azhu Valappil R, Kausar F, Rogaeva E, William Langston J, Tanner CM, Schüle B, Lang AE, Goldman SM, Marras C. Heart rate variability in leucine-rich repeat kinase 2-associated Parkinson's disease. Mov Disord. 2017;32:610–4. [PubMed: 28071824]
  • Warø BJ, Aasly JO. Exploring cancer in LRRK2 mutation carriers and idiopathic Parkinson's disease. Brain Behav. 2017;8:e00858. [PMC free article: PMC5853627] [PubMed: 29568677]
  • West AB, Moore DJ, Biskup S, Bugayenko A, Smith WW, Ross CA, Dawson VL, Dawson TM. Parkinson's disease-associated mutations in leucine-rich repeat kinase 2 augment kinase activity. Proc Natl Acad Sci U S A. 2005;102:16842–7. [PMC free article: PMC1283829] [PubMed: 16269541]
  • Wile DJ, Dinelle K, Vafai N, McKenzie J, Tsui JK, Schaffer P, Ding YS, Farrer M, Sossi V, Stoessl AJ. A scan without evidence is not evidence of absence: scans without evidence of dopaminergic deficit in a symptomatic leucine-rich repeat kinase 2 mutation carrier. Mov Disord. 2016;31:405–9. [PMC free article: PMC4894497] [PubMed: 26685774]
  • Wszolek ZK, Pfeiffer RF, Tsuboi Y, Uitti RJ, McComb RD, Stoessl AJ, Strongosky AJ, Zimprich A, Muller-Myhsok B, Farrer MJ, Gasser T, Calne DB, Dickson DW. Autosomal dominant parkinsonism associated with variable synuclein and tau pathology. Neurology. 2004;62:1619–22. [PubMed: 15136696]
  • Yahalom G, Greenbaum L, Israeli-Korn S, Fay-Karmon T, Livneh V, Ruskey JA, Roncière L, Alam A, Gan-Or Z, Hassin-Baer S. Carriers of both GBA and LRRK2 mutations, compared to carriers of either, in Parkinson's disease: risk estimates and genotype-phenotype correlations. Parkinsonism Relat Disord. 2019;62:179–84. [PubMed: 30573413]
  • Yahalom G, Kaplan N, Vituri A, Cohen OS, Inzelberg R, Kozlova E, Korczyn AD, Rosset S, Friedman E, Hassin-Baer S. Dyskinesias in patients with Parkinson's disease: effect of the leucine-rich repeat kinase 2 (LRRK2) G2019S mutation. Parkinsonism Relat Disord. 2012;18:1039–41. [PubMed: 22703868]
  • Yahalom G, Orlev Y, Cohen OS, Kozlova E, Friedman E, Inzelberg R, Hassin-Baer S. Motor progression of Parkinson's disease with the leucine-rich repeat kinase 2 G2019S mutation. Mov Disord. 2014;29:1057–60. [PubMed: 24903616]
  • Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, Kachergus J, Hulihan M, Uitti RJ, Calne DB, Stoessl AJ, Pfeiffer RF, Patenge N, Carbajal IC, Vieregge P, Asmus F, Muller-Myhsok B, Dickson DW, Meitinger T, Strom TM, Wszolek ZK, Gasser T. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron. 2004a;44:601–7. [PubMed: 15541309]
  • Zimprich A, Müller-Myhsok B, Farrer M, Leitner P, Sharma M, Hulihan M, Lockhart P, Strongosky A, Kachergus J, Calne DB, Stoessl J, Uitti RJ, Pfeiffer RF, Trenkwalder C, Homann N, Ott E, Wenzel K, Asmus F, Hardy J, Wszolek Z, Gasser T. The PARK8 locus in autosomal dominant parkinsonism: confirmation of linkage and further delineation of the disease-containing interval. Am J Hum Genet. 2004b;74:11–9. [PMC free article: PMC1181898] [PubMed: 14691730]
  • Zhao HT, John N, Delic V, Ikeda-Lee K, Kim A, Weihofen A, Swayze EE, Kordasiewicz HB, West AB, Volpicelli-Daley LA. LRRK2 antisense oligonucleotides ameliorate α-synuclein inclusion formation in a Parkinson's disease mouse model. Mol Ther Nucleic Acids. 2017;8:508–19. [PMC free article: PMC5573879] [PubMed: 28918051]
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: NBK1208PMID: 20301387

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...