• Family Conference 2023
  • DecNewsletter
  • First Legacy Guardian Peggy Davis


OUR MISSION

In our drive to find a cure for NBIA,
we provide support to families, educate the public and accelerate research with collaborators from around the world.


 

News

NBIAcure prepares to submit for FDA
approval of CoA-Z compound

 

Educational Resources

​Posters, flyers and
other educational handouts

 

CoPAN

An ultra-rare form of NBIA known as CoPAN, or COASY Protein-Associated Neurodegeneration, is caused by a mutation in the COASY (Coenzyme A Synthase) gene on chromosome 17. CoPAN is so rare that only a few cases have been identified in the world.

Onset typically occurs in childhood, presenting first with spasticity and dystonia of the lower limbs and later with dystonia of the mouth and jaw. Speech problems also occur, including stuttering and slurring words caused by dysarthria (weakened muscles needed for speaking).

Clinical Diagnosis

CoPAN can be diagnosed through an MRI (T2-weighted) scan of the brain.

Based on one individual’s scan, the MRI showed hypointensity, or a darkened area that can indicate iron deposits, in the brain’s substantia nigra and globus pallidus. In another case, the scan showed hyperintensity (white spots that highlight problematic regions) and swelling in the brain’s caudate nuclei, putamina and thalamus. A third individual’s scan showed calcifications in the globus pallidus.

The diagnosis of CoPAN can be confirmed through genetic testing of the COASY gene to identify mutations.

Evaluations Following Initial Diagnosis

To establish the extent of disease in an individual diagnosed with CoPAN, the following evaluations may be useful:

  • A neurologic examination for dystonia, rigidity, spasticity and parkinsonism, including a formal evaluation for walking (ambulation) and speech.
  • An informal developmental assessment, with a referral for more formal testing if a delay is indicated.
  • An assessment for physical therapy, occupational therapy, and/or speech therapy, as well as appropriate assistive devices.
  • A psychiatric assessment for possible obsessive-compulsive behavior.

Management

While there is no standard treatment for CoPAN, symptom management is available. Most are aimed at dystonia, which can be debilitating. Management tactics used with varying degrees of success include:

  • Oral medications: baclofen, anticholinergics, tizanidine and dantrolene
  • Botox: focal injections of botulinum toxin into muscles affected by dystonia. Botox can provide relief for several months from involuntary contractions, twisting, abnormal posture or changes in a person's voice or speech. Because each affected muscle must be injected, this is most practical when an individual has dystonia significantly affecting a specific body area, such as the hand or jaw. Resistance to Botox is a phenomenon, which may cause the treatment to lose its effectiveness over time. This occurs because the body makes antibodies to combat the toxin.
  • Intrathecal baclofen therapy: Although baclofen is usually taken orally, a baclofen pump may be an option for some individuals. An evaluation can be done to determine whether a patient would respond positively to a pump, which is surgically implanted under the skin of the abdomen.
  • DBS: deep brain stimulation. This involves placing electrodes in the brain and attaching the wires to a battery-operated neurostimulator implanted in the chest. The neurostimulator sends pulses to targeted areas in the brain and takes “offline” the part sending too many signals and causing the muscles to move in painful ways. DBS is the most commonly performed surgical treatment for Parkinson's disease.

The symptoms of parkinsonism in NBIA individuals can be treated with the same medications used in Parkinson’s disease.

More information on these therapies can be found in the Medical Information section of our website.

Notice if the individual has problems swallowing to prevent aspiration. Additionally, pay attention to diet and regularly measure height and weight in children to assure adequate nutrition. A gastrostomy tube can be placed if needed. Clinicians also recommend assessing ambulation and speech and communication needs. While the symptoms of CoPAN typically progress slowly, individuals in their 30s often have lost their ability to walk and must use a wheelchair.

Genetics

CoPAN is inherited in an autosomal recessive manner. Because most of our genes exist in pairs (one from the mother, one from the father), we normally carry two working copies of each gene. When a person has one copy of a recessive gene with a change (mutation) in it, the individual is not affected by the disease. That person is called a carrier.

Recessive diseases only occur when both parents are carriers for the same diseaseand pass their mutated genes onto their child. Statistically, there is a one in four chance that two carriers would have an affected child. The chance is two in four that two carriers would have a child who is also a carrier, and it’s one in four that the child will not have the mutation. Carrier testing for relatives and prenatal testing for at-risk pregnancies are suggested if both disease-causing mutations have been identified in an affected family member.

Prenatal Testing

When the disease-causing mutations have been identified in the family, prenatal diagnosis for pregnancies can be done. In one test, DNA is extracted from fetal cells obtained by amniocentesis, usually at 15 to 18 weeks of gestation.

Alternatively, the chorionic villus, the tiny finger-like projections on the edge of the placenta, are tested, usually at 10 to 12 weeks gestation. Embryo screening, known as preimplantation genetic diagnosis, may be an option for some families.

Natural History Studies

Researchers conduct natural history studies to understand changes in disease-affected individuals over time. The TIRCON International NBIA Registry is housed at Ludwig Maximilian University of Munich, Germany and was created under an EU grant from 2011-2015 called Treat Iron-Related Childhood-Onset Neurodegeneration.

The NBIA Alliance and other sources have provided registry funding since 2015. Clinical centers from 16 countries have provided patient clinical data. There are over 750 entries consisting of NBIA patients and controls as of September 2021. Clinical centers seeing at least five NBIA patients are eligible to participate. Clinical and natural history data are available to researchers studying NBIA disorders. Contact Anna Baur-Ulatowska at Anna.Baur@med.uni-muenchen.de for more information on this registry.

CoPan Research

The recessive mutations in CoA Synthase (COASY) that cause COPAN were first uncovered in 2013 by Sabrina Dusi of The Firc Institute of Molecular Oncology, a research center in Milan, Italy. This study used exome sequencing for individuals who had an NBIA clinical presentation and neuroimaging indications but lacked the mutations in previously discovered genes. It resulted in this publication:Exome Sequence Reveals Mutations in CoA Synthase as a Cause of Neurodegeneration with Brain Iron Accumulation

Coenzyme A (CoA) is important to all living organisms because of its involvement in numerous enzymatic reactions. It is a key molecule for metabolizing fatty acids, carbohydrates, amino acids and ketone bodies. The last two steps of its biosynthesis are carried out by CoA Synthase, where the COPAN mutation occurs. Normal Coenzyme A Synthase is crucial for healthy nervous system function.

Because CoPAN is so rare, more studies are needed to better understand it.

Idiopathic NBIA

Idiopathic NBIA is of unknown origin, although it is suspected to be genetic. For many families, the person diagnosed with NBIA is the first and only affected individual, so it is difficult to know whether there is a specific pattern of inheritance. It is thought that most of these cases are probably recessive because some families have more than one affected child and idiopathic NBIA is more common in families in which the parents are related, such as distant cousins. That would make it more likely that they share a recessive gene.

The symptoms are more varied because there are probably several different causes of neurodegeneration for individuals in this group. As with other forms of NBIA, the idiopathic form features early- and late-onset types. Currently, about five percent of those diagnosed with NBIA are believed to be in this group.

Clinical Diagnosis

Suspicion of NBIA usually arises when an MRI of the brain shows excessive iron accumulation, along with a movement disorder. Depending on symptoms, genetic testing is done to rule out other forms of NBIA or other genetic disorders. If all are negative, but the findings are characteristic of NBIA, then the diagnosis of Idiopathic NBIA is given until further evidence of the genetic origin or another cause is determined.

Evaluations Following Initial Diagnosis

  • Neurologic examination for movement disorders, including dystonia, rigidity, spasticity and choreoathetosis, which are involuntary movements that include contractions, twisting and writhing. Walking ability and speech also are assessed.
  • An eye exam to check for such problems as retinopathy (damage to the small blood vessels in the retina) and optic atrophy (the loss of some or most of the fibers of the optic nerve).
  • Developmental screening, with referral for more formal testing if delay is indicated.
  • Assessment to see if physical therapy, occupational therapy and/or speech therapy are warranted.
  • Assessment of behavior and neuropsychiatric changes.
  • Medical genetics consultation.

Management

Doctors treat patients with medication when appropriate. Also, affected individuals may need a feeding tube or other assistance to achieve adequate nutrition. Other therapies might also be warranted.

Genetics

Testing to find the gene responsible for an individual’s form of NBIA may be pursued.

Prenatal Testing

In the absence of a known genetic mutation, testing during pregnancy to assess risk is not possible.

Research

Researchers continue their search for remaining genes that cause NBIA disorders, and much progress is being made with new technology. If a new gene is found by researchers, the person’s samples will be examined to see if they have mutations and their form of NBIA identified. Affected individuals diagnosed with Idiopathic NBIA might want to participate in the TIRCON International Patient Registry.

Beta-propeller Protein-Associated Neurodegeneration

(BPAN) is caused by mutations in the WDR45 gene on the X chromosome. It is inherited in an X-linked dominant manner, meaning that a single copy of the mutated gene is enough to cause disease in both males and females. Most affected individuals identified so far have been simplex, or isolated, cases; they are the only person in their family to have the disease. The majority are females, indicating the mutations are new, or de novo, and suggesting that mutations may be lethal in most males before birth. Rarely, the disorder may also occur in a sibling. In these cases, the mutation was inherited from a mildly affected parent. BPAN is the most common NBIA disorder at 35-45% of the NBIA population, with an estimated prevalence of 2 to 3 per million individuals.

Clinical Features

BPAN has a wide phenotypic spectrum, meaning that symptom presentation and severity can vary greatly among patients. The disease progresses in two stages. The first stage occurs in childhood and is characterized by developmental delay and intellectual disability. Most children are described as clumsy with an ataxic, or unsteady, staggering gait. In addition, children with BPAN usually show expressive language delay disproportionate to their other disabilities. Consequently, most individuals with BPAN can only speak a few words.

Several other common features are sleep disorders, seizures and ocular defects, such as retinal and optic nerve disease. Individuals may present with some autistic features, such as involuntary, repetitive and seemingly meaningless hand movements and other repetitive behaviors.

Many families report that their young children with BPAN have difficulty falling asleep and staying asleep, and thus tend to sleep in spurts. The abnormal sleep patterns may be caused by unrecognized nocturnal seizure activity, dysregulation of the sleep cycle, sleep-disordered breathing, reflux or spasticity.

Seizures are a common symptom of BPAN. Beginning in infancy and early childhood, they occur in about two-thirds of children with BPAN and typically cease in adolescence. They often start as febrile, or fever, seizures in a young child. Other types of seizures in BPAN individuals include generalized seizures, focal seizures with impaired consciousness, and epileptic spasms. Focal seizures begin in one area of the brain, while generalized seizures occur in both hemispheres of the brain at the same time. Generalized seizures can be categorized in the following ways:

  • Absence: brief, sudden lapses of consciousness
  • Tonic: sudden tension or stiffness that may affect the arms, legs or body, lasting about 20 seconds and occurring typically during sleep
  • Tonic-clonic: loss of consciousness in which muscles stiffen and jerking movements last one to three minutes
  • Myoclonic: brief, shock-like jerks of a muscle or group of muscles, in which the person is usually awake and able to think clearly

Seizure patterns can be similar to those of epileptic syndromes such as West syndrome and Lennox-Gastaut syndrome. Seizures are typically worse in early childhood and lessen with age.

Typically, BPAN individuals lose brain cells and tissue in the cerebral area of the brain, a condition called generalized cerebral atrophy. During adolescence or adulthood, affected individuals experience a relatively sudden onset of progressive dystonia-parkinsonism and cognitive decline.

Parkinsonism is caused by the degeneration of nerve cells in the brain and is characterized by tremors and shaking, slow movements, stiffness in the arms, legs or trunk, instability while standing, and frozen gait, which is a pause in one’s attempt to move forward when walking.

Dystonia, a movement disorder in which a person’s muscles contract uncontrollably, also is common. The affected body part will twist involuntarily, resulting in repetitive movements. This can affect a single muscle, a muscle group or the entire body.

BPAN is progressive, meaning the symptoms worsen over time.

Clinical Diagnosis

Delayed development and other symptoms often lead to genetic testing to uncover a diagnosis. Since BPAN is very rare and often not suspected, it typically is diagnosed with Whole Exome Sequencing (WES), which looks at all the protein-coding regions of the genome. Whole Genome Sequencing (WGS) is becoming more common, as this test looks at the entire genome and is the most extensive one currently available.

As a child progresses into adolescence, evidence of the disease can be seen in a brain MRI that shows iron accumulation in the basal ganglia, specifically in the substantia nigra and globus pallidus. Early imaging findings include mild white matter volume loss, delayed myelination and a thin corpus callosum. However, a young child with BPAN may have a normal brain MRI.

One possibly unique feature of BPAN can be detected on an MRI of the brain: the presence of a bright halo seen in a certain view (T-1 weighted) in the substantia nigra and cerebral peduncles. Hyperintense signal (an abnormality that shows as bright white on an MRI) in this region seems to appear at the same time or soon after the symptoms of progressive dystonia-parkinsonism emerge. On an MRI T-2 weighted view, a hypointense signal (an abnormality that appears dark on the MRI) appears in the substantia nigra that is usually detectable early in the second decade of life and possibly sooner.

Symptom Management

Various types of treatments help manage symptoms. Because symptom severity and complexity varies widely, management should be tailored to the individual. Individuals will benefit from routine follow-up by a neurologist for medication management and interval assessment of neurological regression, ambulation, seizure activity, speech, sight and swallowing.

Anti-seizure medication is recommended for children with recurrent, unprovoked seizures. Many children will outgrow their seizures and may be taken off the medication in adolescence as directed by their doctor.

Dopaminergic drugs, which mimic the effects of dopamine, a chemical involved in movement, are sometimes given for parkinsonism. Those drugs need to be further monitored for adverse neuropsychiatric effects and disabling motor fluctuations and dyskinesias (abnormal, uncontrollable, involuntary movements).

Other therapies that can help BPAN individuals include physical therapy to address gross motor dysfunction, maximize mobility and reduce the risk of later-onset orthopedic complications. Occupational therapy is recommended for difficulty with fine motor skills that affect adaptive function, such as feeding, grooming and dressing. Feeding therapy, by an occupational or speech therapist, is recommended for difficulty feeding due to poor oral motor control.

Since most individuals are nonverbal or can only say a few words, speech therapy is recommended to help with augmentative and alternative communication. If the child also has autism symptoms, a comprehensive treatment model is recommended that may include applied behavior analysis and developmental approaches to support communication. All children with BPAN should have a formal audiological evaluation.

Developmental delays and intellectual disabilities are often managed in school through an individualized education plan (IEP).

Genetics

With genetic testing available, a BPAN diagnosis during childhood is now possible. Earlier detection also gives families an opportunity to find others with the same diagnosis for emotional support and advice on therapies. It further enhances the ability to participate in research and may enable earlier intervention for subtle symptoms of parkinsonism.

After a child is diagnosed, parental testing is recommended. Though most cases are simplex and de novo (only one individual is affected in the family because of spontaneous mutation in the germ cell), cases have been reported in which parents have another child with BPAN.

While many individuals with BPAN do not have children, mildly affected individuals that choose to conceive should seek genetic counseling. The chance to pass on a WDR45 mutation (pathogenic variant) is as great as 50%.

The main resource for the clinical information provided here is BPAN - GeneReviews. GeneReviews is primarily used by genetics professionals, so the terminology and information may be difficult for the general public.

All information is for informational purposes only. We do not endorse specific studies or clinical trials, experimental drugs, procedures, biotech or pharmaceutical companies.

Natural History Studies

BPANready

Oregon Health & Science University has a registry and conducts natural history studies that involve patients or guardians entering data remotely by phone, paper or online. It involves the retrospective use of medical records as well as listing BPAN milestones and patient (or parent) reported outcome measures.

The research team is using Latent Growth Models (LGM) in which latent growth curves model disease progression. Understanding the progression of the disease and how it manifests in different cases will increase clinical knowledge and help researchers identify important avenues for study and treatment.

BPAN families wishing to participate can find more information and register at NBIAcure.

RARE-X BPAN Data Platform

RARE-X is a nonprofit created to accelerate rare disease research, treatments, and cures by removing barriers for data collection and sharing. The Rare-X platform is designed to encourage data-sharing and thus quicken the speed of information and the pace of research into BPAN.

The platform is currently open to BPAN families that speak English, with plans to expand to other languages in the future. Those wishing to participate can access the site at https://bpan.rare-x.org.

The platform is free for families to use. It keeps health information confidential by providing only data that is not attached to individual names. BPAN families and individuals control whether to allow or deny access to their personal health information for any research project.

RARE-X BPAN asks BPAN families about their experiences with the condition through various structured and standardized surveys on various topics which can be updated by families as needed. A researcher who is studying mitochondria for example, can do a query for all relevant information on a specific symptom and might find similarities across various diseases that provide insights into new treatment pathways.

RARE-X provides support, technology, tools and resources necessary for successful data collection, and secure but open data sharing on a global scale.

The platform does not replace other forms of data collection that we have available in our NBIA community such as BPANready, Citizen and the TIRCON International NBIA Registry and Biobank. In fact, these existing projects can be connected to the RARE-X platform and expand the data available to interested researchers.

It is hoped that RARE-X BPAN will enhance our readiness for clinical trials and engage new researchers and biotech companies. It is cloud-based and researchers can query the database in many ways to find data that is brought together rather than in separate silos. The de-identified data never leaves the system. Researchers can link to it but cannot download it.

TIRCON International NBIA Registry

The TIRCON International NBIA Registry is housed at Ludwig Maximilian University of Munich, Germany, and was created under an EU grant from 2011-2015 called Treat Iron-Related Childhood-Onset Neurodegeneration.

The NBIA Alliance and other sources have provided registry funding since 2015. Clinical centers from 16 countries have provided patient clinical data. There are over 750 entries consisting of NBIA patients and controls as of September 2021. Clinical centers seeing at least five NBIA patients are eligible to participate. Clinical and natural history data are available to researchers studying NBIA disorders. Contact Anna Baur-Ulatowska at Anna.Baur@med.uni-muenchen.de for more information on this registry.

Articles


BPAN Consensus Guidelines Information

READ ARTICLE

WATCH VIDEO

Statement About Rett Syndrome Medication

READ STATEMENT

BPAN Mouse Model Now Available to Researchers

READ ARTICLE


Research

The WDR45 gene was discovered in 2012 by Dr. Tobias Haack of Helmholtz Zentrum München, a German research center. The discovery resulted from a collaborative study involving the lab of Dr. Susan Hayflick at the Oregon Health & Science University. It applied next generation sequencing to individuals suffering from NBIA with no known genetic cause. This study resulted in the finding of the WDR45 gene, causing BPAN, with the findings reported in December 2012 in the American Journal of Human Genetics entitled "Exome Sequencing Reveals De Novo WDR45 Mutations Causing a Phenotypically Distinct, X-Linked Dominant Form of NBIA."

The focus of the following research has been on finding suitable model organisms to perform pre-clinical testing. These organisms can provide important clues on the cause of disease as well as help develop and test potential treatments through drug screening that can be later translated to clinical trials. It is important that the model mimics the condition seen in patients. Below are various study models for BPAN.

  • C. elegans - roundworm
  • Drosophila - fruit flies
  • Mice
  • Pluripotent Stem Cells - skin cells from BPAN patients reprogrammed into neurons to model the disease. This enables scientists to learn more about the progression of the disease and to screen large numbers of small molecule drugs for potential treatment.

Other research areas focus on understanding the role of iron in BPAN and how a WDR45 mutation leads to neurodegeneration.

Many research efforts have indicated that BPAN causes impaired autophagic activity. Autophagy is essential for maintaining a stable environment in cells. It is the process by which cells clean out waste materials. In normal cells, waste materials are aggregated into a sac called the autophagosome and are transported to an organelle called the lysosome. The autophagosome and lysosome fuse and become an autolysosome. The waste is then broken down by enzymes that can digest the waste products and recycle them for further use in the cell.

When neural-specific depletion of genes for autophagosome formation occurs in the brain, it leads to massive neuron loss and axonal degeneration. Neurons and axons are fundamental units of the brain responsible for sending electrical signals to receive sensory information and motor commands to muscles.

Researchers want to understand the role that WDR45 plays in the incredibly intricate cascade of genes and proteins in the autophagy process. Once the precise mechanisms are determined, therapeutic drugs can be tested to target the specific pathways necessary to restore autophagy function in the cell.

Several potential pathways to treatments exist. One is testing known drugs that target neuroinflammation, oxidative stress and mitochondrial dysfunction. Another is gene therapy. This would involve providing a working copy of the WDR45 gene into cells or activating the inactive copy of WDR45 on the X chromosome. Scientists have yet to establish if these are viable options for BPAN.

The 7th International Symposium on NBIA & Related Disorders held Sept. 30 to Oct. 3, 2020, has recorded sessions #14-16 pertaining to BPAN research.

BPAN Research Articles:

Following is a list of BPAN research articles published recently and early papers after the gene discovery. Other free access BPAN articles can be found at Pub Med Central.

 

2023 - Cardiac glycosides restore autophagy flux in an iPSC-derived neuronal model of WDR45 deficiency

2023 - A burning question from the first international BPAN symposium: is restoration of autophagy a promising therapeutic strategy for BPAN?

2023 - Automated high-content imaging in iPSC-derived neuronal progenitors

2022 - Psychometric outcome measures in beta-propeller protein-associated neurodegeneration (BPAN)

2022 - Digest it all: the lysosomal turnover of cytoplasmic aggregates

2021 - A neurodegeneration gene, WDR45, links impaired ferritinophagy to iron accumulation

2021 - Consensus clinical management guideline for beta-propeller protein-associated neurodegeneration

2021 - A comprehensive phenotypic characterization of a whole-body Wdr45 knock-out mouse

2020 - WDR45 contributes to neurodegeneration through regulation of ER homeostasis and neuronal death

2020 - Phenotypic and Imaging Spectrum Associated With WDR45

2019 - WDR45 contributes to neurodegeneration through regulation of ER homeostasis and neuronal death

2019 - Role of Wdr45b in maintaining neural autophagy and cognitive function

2019 - Is WDR45 the missing link for ER stress-induced autophagy in beta-propeller associated neurodegeneration?

2018 - Autophagosome maturation: An epic journey from the ER to lysosomes

2018 - Iron overload is accompanied by mitochondrial and lysosomal dysfunction in WDR45 mutant cells

2015 - Neuropathology of Beta-propeller protein associated neurodegeneration (BPAN): a new tauopathy

2013 - De novo mutations in the autophagy gene WDR45 cause static encephalopathy of childhood with neurodegeneration in adulthood

2013 - β-Propeller protein-associated neurodegeneration: a new X-linked dominant disorder with brain iron accumulation

2013 - BPAN: The Only X-Linked Dominant NBIA Disorder

Kufor-Rakeb Syndrome, also known as Parkinson’s Disease 9 (PARK9), is an ultra-rare form of NBIA inherited in an autosomal-recessive manner. It is characterized by juvenile-onset parkinsonism and dementia.

Kufor-Rakeb is named for the village in Jordan where it was first described in 1994. In 2006, a mutation in the ATP13A2 gene was deemed responsible. Fewer than 50 affected individuals have been diagnosed with Kufor-Rakeb. They live in the US, Italy, South America, the Middle East and Asia.

Clinical Diagnosis

Brain CT and MRI scans may show diffuse, or widespread, moderate cerebral, cerebellar and brain stem atrophy. Iron accumulation in the basal ganglia affecting the putamen and caudate is present in some, but not all, individuals.

Disease onset is usually in adolescence. Parkinsonism is caused by the degeneration of nerve cells in the brain and is characterized by tremors and shaking; slow movements; stiffness in the arms, legs or trunk; instability while standing; and freezing of gait.

Key Clinical Manifestations:

  • Parkinsonism
  • Dementia (progressive cognitive decline)
  • Supranuclear gaze palsy (inability to look in a particular direction because of cerebral impairment)
  • Facial-faucial-finger myoclonus (involuntary jerking of the facial and finger muscles)
  • Visual hallucinations
  • Oculogyric dystonic spasms (involuntary intermittent or sustained deviation of the eyes in a usually upward direction)

Symptom onset often leads to genetic testing to search for a diagnosis. Because Kufor-Rakeb is so rare and not often suspected, it is often diagnosed with Whole Exome Sequencing (WES), which looks at all the protein coding regions of the genome. Whole Genome Sequencing (WGS) is becoming more common. It looks at the entire genome and is the most extensive test available at this time.

Genetics

Kufor-Rakeb is caused by a mutation in the ATP13A2 gene and is inherited in an autosomal recessive manner. Because most of our genes exist in pairs (one coming from the mother and one coming from the father), we normally carry two working copies of each gene. When one copy of a recessive gene has a change (mutation) in it, the person should still have normal health. That person is called a carrier.

Recessive diseases only occur when both parents are carriers for the same condition and pass their changed genes on to their child. Statistically, there is a one in four chance that two carriers would have an affected child. There is a two in four chance the parents will have a child who is also a carrier. The chances are one in four that the child will not have the gene mutation. Carrier testing for relatives and prenatal testing for at-risk pregnancies are suggested if both disease-causing mutations have been identified in an affected family member.

Prenatal Testing

When disease-causing mutations are identified in a family, prenatal diagnosis for pregnancies can be done. In one test, DNA is extracted from fetal cells obtained by amniocentesis, usually at 15 to 18 weeks’ gestation. Or, sampling is done of the chorionic villus, the tiny finger-like projections on the edge of the placenta, usually at 10 to 12 weeks’ gestation. Embryo screening, known as preimplantation genetic diagnosis, may be an option for some families.

Treatments

There is no cure for Kufor-Rakeb syndrome, but treatments are available to manage symptoms. As symptom severity and complexity varies widely, management should be tailored to the individual. Individuals will benefit from routine follow up by a neurologist for medication management and interval assessment of motor and cognitive functioning.

Treatment of Kufor-Rakeb syndrome often resembles treatment of typical Parkinson’s disease. Commonly, two medications, levodopa (L-DOPA) and carbidopa, are combined.

Other therapies also may benefit individuals with Kufor-Rakeb Syndrome. Physical therapy often is recommended to address gross motor dysfunction, maximize mobility and reduce risk of later onset orthopedic complications. Occupational therapy is recommended for difficulty with fine motor skills that affect adaptive function, such as feeding, grooming and dressing. Feeding therapy by an occupational or speech therapist is recommended for affected individuals who have difficulty feeding due to poor oral motor control.

Clinical Trials

Search ClinicalTrials.gov in the US and EU Clinical Trials Register in Europe for access to information on clinical studies as they become available.

Research Articles:

Following is a list of relevant published research articles. Other free access articles can be found at Pub Med Central.

2020 - Kufor-Rakeb Syndrome/Parkinson Disease Type 9

2018 - Action Myoclonus and Seizure in Kufor-Rakeb Syndrome

2015 - The role of ATP13A2 in Parkinson's disease: Clinical phenotypes and molecular mechanisms

2014 - Mutations in the ATP13A2 Gene and Parkinsonism: A Preliminary Review

2012 - Mutation of the parkinsonism gene ATP13A2 causes neuronal ceroid-lipofuscinosis | Human Molecular Genetics | Oxford Academic

2011 - Regulation of intracellular manganese homeostasis by Kufor-Rakeb syndrome-associated ATP13A2 protein

2006 - Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a lysosomal type 5 P-type ATPase

Partners

nbia alliance logo1NBIA Cure logoRare Disease Day PartnerTIRCON

Genetic Alliance logoNORDEURORDISGlobal Genes


Disclaimer    |     Privacy Policy    |     Financials    |     Contact Us

Give While You Shop!

Igive.com

G-CFB5FV5NLL