My watch list
my.bionity.com  
Login  

Huntington's disease



Huntington's disease
Classification & external resources
George Huntington's 1872 paper described the disorder.
ICD-10 G10., F02.2
ICD-9 333.4, 294.1
OMIM 143100
DiseasesDB 6060
MeSH D006816

Huntington's disease, known historically as Huntington's chorea and chorea maior, is a rare inherited neurological disorder affecting up to approximately 1 person per 10,000 people of Western European descent and 1 per 1,000,000 of Asian and African descent. It takes its name from the New York physician George Huntington who described it precisely in 1872 in his first medical paper. HD has been heavily researched in the last few decades and it was one of the first inherited genetic disorders for which an accurate test could be performed.

Huntington's disease is caused by a trinucleotide repeat expansion in the gene coding for Huntingtin (Htt) and is one of several polyglutamine diseases. This expansion produces an altered form of the Htt protein, mutant Huntingtin (mHtt), which results in neuronal cell death in select areas of the brain. Huntington's disease is a terminal illness.

Huntington's disease's most obvious symptoms are abnormal body movements called chorea and a lack of coordination, but it also affects a number of mental abilities and some aspects of personality. These physical symptoms occur in a large range of ages, with a mean occurrence in a person's late forties/early fifties. If the age of onset is below 20 years then it is known as Juvenile HD. As there is currently no proven cure, symptoms are managed with various medications and care methods.

Contents

Symptomatology and pathology

Although there is no sudden loss of affected abilities, there is a progressive decline of them. Physical signs are usually the first noticed, with cognitive and psychiatric deficits manifesting subsequently. Physical symptoms are almost always visible; cognitive symptoms exhibit differently from person to person, and psychiatric problems may not be evident.

Degeneration of neuronal cells, especially in the frontal lobes and caudate nucleus (the striatum) of the basal ganglia occurs. There is also astrogliosis and loss of medium spiny neurons. This results in the selective degeneration of the indirect (inhibitory) pathway of the basal ganglia, via the lateral pallidum and the subthalamic nucleus coupled pacemaker system.

In more detail, the neurological changes involved with Huntingtons are as follows. Inside the brain, the initiation of motion is sent down the spinal cord from the primary motor areas, which in turn have received signals from the other regions that deal with motor function. At the same time that the stimulus is being sent down the spinal cord, the subthalamic nuclei of the striatum excite the internal globus pallidus. In a normally functioning individual, the signal would fully inhibit the internal globus pallidus, which would in turn inhibit the thalamus and modulate motion. The problem in Huntington's disease is that the subthalamic nuclei no longer generate enough excitation to the internal globus pallidus. The globus pallidus thus sends an abnormally weak inhibitory signal to the thalamus. The thalamus in turn then sends a strong excitatory signal to the putamen. The end result is a lack of modulation via two pathways, direct and indirect. All signals inside the striatum are too weak to inhibit the appropriate target regions. The only exception is the external globus pallidus, which over- inhibits when signalled, and alters the flow of excitation from the subthalamic nuclei, contributing to the lowered function and loss of movement control. This creates the characteristic jerky uncontrolled movement.

Physical

Most people with HD eventually exhibit jerky, random, uncontrollable movements called chorea, although some exhibit very slow movement and stiffness (bradykinesia, dystonia). These abnormal movements are initially exhibited as general lack of coordination and an unsteady gait and gradually increase as the disease progresses; this eventually causes problems with loss of facial expression (called "masks in movement") or exaggerated facial gestures, inability to sit or stand stably, speech, chewing and swallowing (which can lead to weight loss if diet and eating methods are not adjusted accordingly[1][2]), and loss of determination. In the later stages of the disease, speaking is impaired with slurred words and uncontrollable movements of the mouth, eating and mobility are extremely difficult if not impossible, and full-time care is required.

Cognitive

Selective cognitive abilities are progressively impaired, including: executive function (planning; cognitive flexibility, abstract thinking, rule acquisition, initiating appropriate actions, and inhibiting inappropriate actions), psychomotor function (slowing of thought processes to control muscles), speech (slurring of words) and some uncontrollable movement of the lips, perceptual and spatial skills of self and surrounding environment, selection of correct methods of remembering information (but not actual memory itself), and ability to learn new skills, depending on the affected parts of the brain.

Psychopathological

Psychopathological symptoms vary more than cognitive and physical symptoms, and may include anxiety, depression, a reduced display of emotions (blunting) and decreased ability to recognize negative expressions like anger, disgust, fear or sadness in others[3], egocentrism, aggressive behavior, compulsivity which can cause addictions such as alcoholism and gambling, or hypersexuality.

Inheritance

  Huntington's disease is autosomal dominant, needing only one affected allele from either parent to inherit the disease. Although this generally means there is a one in two chance of inheriting the disorder from an affected parent, the inheritance of HD and other trinucleotide repeat disorders is more complex.

When the gene has more than 36 copies of the repeated trinucleotide sequence, the DNA replication process becomes unstable and the number of repeats can change in successive generations. This can mean that in a parent without HD but with a count close to 36, the count may increase above the threshold that causes HD.

If the gene is inherited from the mother, the count is usually similar. Paternal inheritance tends to increase the number of repeats.[4] Because of the progressive increase in length of the repeats, the disease tends to increase in severity and have an earlier onset in successive generations. This is known as anticipation.

De novo mutations (neither parent has HD) are rare.

Homozygous individuals (where both parents have HD) generally do not show an earlier onset of disease, but may have an increased rate of decline.

Causes

See also: HD (gene)

The gene involved in Huntington's disease, called the HD gene or Interesting Transcript 15 (IT15), is located on the short arm of chromosome 4 (4p16.3). In the first part (5'end) of the HD gene, there is a sequence of three DNA bases, cytosine-adenine-guanine (CAG), that is repeated multiple times (i.e. ...CAGCAGCAG...); this is called a trinucleotide repeat. CAG is the genetic code for the amino acid glutamine, thus a series of CAG forms a chain of glutamine known as polyglutamine or (polyQ).

A polyQ length of less than 36 glutamines, produces a cytoplasmic protein called huntingtin (Htt), whereas a sequence of 40 or more produces an erroneous form of Htt, mHtt (standing for mutant Htt). Counts between these two have not been fully understood, and sometimes result in HD, othertimes not.

Having mHtt instead of Htt causes certain neurons in select areas of the brain to have an increased mortality, progressively affecting neurological functioning. Observations show that generally, the greater the number of CAG repeats, the earlier the onset of symptoms.[5]

Mechanism

See also: Huntingtin

Like all proteins, Htt and mHtt are translated, perform or affect biological functioning, and are finally cleared up in a process called degradation. The exact mechanism in which mHtt causes or affects the biological processes of DNA replication and programmed cell death (apoptosis) remains unclear, so research is divided in two; identifying the normal processes of Htt, the abnormal processes of mHtt, and the effects of parts of the protein (known as aggregates) left after degradation.

Function

Htt is involved in vesicle trafficking as it interacts with HIT1, a clathrin binding protein, to mediate endocytosis, the absorption of materials into a cell.[6][7]

MHtt reduces the production of brain-derived neurotrophic factor (BDNF) which protects neurons in the striatum.[8] This loss of BDNF may contribute to striatal cell death, which does not follow apoptotic pathways as the neurons appear to die of starvation.[9]

Degradation

Both Htt and mHtt are cleaved (the first step in degradation) by Caspase-3, which removes the (amino end) N-terminal.[10] Caspase-2 then further breaks down the amino terminal fragment ( the CAG repeat part) of Htt, but cannot act upon all of the repeats of mHtt.[11] These repeats left in the cell, called aggregates or N-fragments, are able to affect polyQ dependent transcription.[12] Specifically, mHtt binds with TAFII130, a coactivator to CREB dependent transcription.[13] The aggregates also interact with SP1, thereby preventing it from binding to DNA,the normal functioning of these proteins.[14]

In transgenic mice neurodegeneration caused by mHtt is related to the caspase-6 enzyme cleaving the Htt protein, as they did not show effects of HD in experiments.[15]

In genetically altered "knockin" mice, the extended CAG repeat portion of the gene is all that is needed to cause disease.[16] Aggregates of mHtt are present in the brains of both HD patients[17] and HD mice,[18] and are most prevalent in cortical pyramidal neurones, less so in striatal medium-sized spiny neurones and almost absent in most other brain regions including hippocampus and cerebellum [19][20][21] These aggregates consist mainly of the amino terminal end of mHtt (CAG repeat), and are found in both the cytoplasm and nucleus of neurons.[22] The presence of these aggregates however does not correlate with cell death.[23] Thus mHtt acts in the nucleus but does not cause apoptosis through aggregation.[24]

Diagnosis

To determine whether initial symptoms are evident, a physical and/or psychological examination is required. The uncontrollable movements are often the symptoms which cause initial alarm and lead to diagnosis; however, the disease may begin with cognitive or emotional symptoms, which are not always recognized. Pre-symptomatic testing is possible by means of a blood test which counts the number of repetitions in the gene.

A negative blood test means that the individual does not carry the expanded copy of the gene, will never develop symptoms, and cannot pass it on to children. A positive blood test means that the individual does carry the expanded copy of the gene, will develop the disease, and has a 50% chance of passing it on to children. A pre-symptomatic positive blood test is not considered a diagnosis, because it may be decades before onset.

Because of the ramifications on the life of an at-risk individual, with no cure for the disease and no proven way of slowing it, several counseling sessions are usually required before the blood test. Unless a child shows significant symptoms or is sexually active or considered to be Gillick competent, children under eighteen will not be tested. The members of the Huntington's Disease Society of America strongly encourage these restrictions in their testing protocol. A pre-symptomatic test is a life-changing event and a very personal decision.

For those living in America, there is a list of testing centers available on the HDSA homepage[25] and embryonic genetic screening is also possible, giving mutation-positive or at-risk individuals the option of making sure their children will not inherit the disease. Expense and the ethical considerations of abortion are potential drawbacks to these procedures. A full pathological diagnosis can only be established by a neurological examination's findings and/or demonstration of cell loss, especially in the caudate nucleus, supported by a cranial CT or MRI scan findings.

It is possible to test an embryo either in the womb (prenatal diagnosis) or to ensure a child will not have HD by utilising in vitro fertilisation and testing before implantation.

Management

There is no treatment to fully arrest the progression of the disease, but symptoms can be reduced or alleviated through the use of medication and care methods. Huntington mice models exposed to better husbandry techniques, better access to water especially, lived much longer than mice who were not well cared for.

Medication

Other standard treatments to alleviate emotional symptoms include the use of antidepressants and sedatives, with antipsychotics (in low doses) for psychotic symptoms.

Nutrition

Nutrition is an important part of treatment; most HD sufferers need two to three times the calories of the average person to maintain body weight[citation needed], so a nutritionist's advice is needed (the normal population's average daily intake is approximately 2000 calories for women and 2500 for children and men).

Speech therapy can help by improving speech and swallowing methods. This advice should be sought early on, as the ability to learn is reduced as the disease progresses.

To aid swallowing, thickener can be added to drinks. The option of using a stomach PEG is available when eating becomes too hazardous or uncomfortable, this will reduce the chances of pneumonia due to aspiration of food and increase the amount of nutrients and calories that can be ingested.

EPA, an Omega-III fatty acid, slows and possibly reverses the progression of the disease.[citation needed] It is currently in FDA clinical trial, as Miraxion (LAX-101), for prescription use. Clinical trials utilize 2 grams per day of EPA. In the United States, it is available over the counter in lower concentrations in Omega-III and fish oil supplements.

Potential treatments

Trials and research are conducted on Drosophila fruit flies and mice that have been genetically modified to exhibit HD, before moving on to human trials.

Research is reviewed on various websites for HD sufferers and their families, including the Huntington's Disease Lighthouse, Hereditary Disease Foundation, and Stanford HOPES websites. Primary research can be found by searching the National Library of Medicine's PubMed. Clinical trials of various treatments are ongoing, or yet to be initiated. For example, the US registrar of trials has nine that are currently recruiting volunteers.[26]

Intrabody Therapy

Engineered intracellular antibody fragments (intrabodies) have shown efficacy in vivo as therapeutic agents against pathogenic mutant huntingtin protein in fly models of HD. An intracellularly expressed single-chain Fv against the amino-terminal end of mutant huntingtin (mHtt) has been shown to reduce mHtt aggregate formation and increase turnover of the mHtt fragments in tissue culture models of HD.[27][28] In a drosophila HD model, the expression of this anti-HD intrabody rescued fly survival through the larval and pupal stages to adult emergence. Additionally, the intrabody delayed neurodegeneration in the fly model, and significantly increased the mean adult lifespan.[29] The engineered antibody approach shows promise as a tool for drug discovery and as a potential novel therapeutic for other neurodegenerative disorders resulting from protein misfolding or abnormal protein interactions, including Parkinson’s, Alzheimer’s and prion diseases.[30]

Gene silencing

The pathology of HD has been conclusively linked to a single gene, researchers have investigated using gene knockdown of mHtt as a potential treatment. Using a mouse model of HD, siRNA therapy achieved a 60% reduction in expression of the mHtt and progression of the disease was stalled.[31] In another study, mouse models in late stages of the disease recovered motor function after expression of mHtt was shut down.[32]

Stem Cell Implants

This treatment is based on the replacement of damaged neurons by injecting stem cells (a type of cell that can form itself into a specialized cell) into the damaged area. If enough damaged neurons are replaced, symptoms should be alleviated. This treatment would not prevent further neuronal damage. Experiments have yielded some positive results in animal models. [33]

Others

Other agents and measures that have shown promise in initial experiments include dopamine receptor blockers, creatine, CoQ10, the antibiotic Minocycline, exercise, antioxidant-containing foods and nutrients, antidepressants (notably, but not exclusively, selective serotonin reuptake inhibitors SSRIs, such as sertraline, fluoxetine, and paroxetine) and select dopamine antagonists, such as tetrabenazine.

Prognosis

Development of Huntington’s disease is highly CAG repeat length-dependent. In the normal population, the CAG repeat is of between 7 and 35 repeats. Individuals carrying more than approximately 40 repeats will, however, go on to develop the disease at some point within their lifetime [34]. The age of onset (and to a degree the severity of the disease) and hence the age at death, are inversely correlated with the length of the expanded CAG repeat, such that those with longer repeats develop the disease earlier. Individuals with greater than approximately 60 CAG repeats often develop juvenile Huntington's disease [35]. There is a large variation in age of onset for any given CAG repeat length within the intermediate range (40-50 CAGs). For example, a repeat length of 40 CAGs leads to an onset ranging from 40 to 70 years of age (North American and Canadian population). This variation means that, although logarithmic algorithms have been proposed for predicting the age of onset (for example see [36],[37]), in reality predicting the precise age at which clinical signs will manifest is not practical.

Juvenile HD has been defined as having an onset younger than 20 years of age. The symptoms of juvenile HD are different from those of adult-onset HD in that they generally progress faster and are more likely to exhibit rigidity and bradykinesia instead of chorea and often include seizures[38].

Following the onset of the “classical” symptoms of the disorder, patients generally live for a further 15 to 20 years [39]. Death, however, is not caused by Huntington’s disease per se, but rather by associated complications. The most common causes of death of HD patients includes pneumonia (one third of all patients), heart failure (although heart disease, cerebrovascular disease and atherosclerosis show no increase), choking and nutritional deficiencies[40]. Suicide is an associated risk, with suicide rates of up to 7.3 per cent of all patient deaths; four times that of the general population [41]. This rather taboo subject is likely to be under-reported, with up to 27 per cent of possibly affected individuals attempting suicide at least once [42]. Likewise, suicide in the general population is likely to be underestimated; however, some reports have accounted for this and still find a significant increase in suicide rate amongst HD patients [43]

Epidemiology

The prevalence is 5 to 8 per 100,000, varying geographically.

About 10 percent of HD cases occur in people under the age of 20 years. This is referred to as Juvenile HD, "akinetic-rigid", or "Westphal variant" HD.

Ethical aspects

Whether or not to have the test for HD Genetic counseling may provide perspective for those at risk of the disease. Some choose not to undergo HD testing due to numerous concerns (for example, insurability). Testing of a descendant of a person 'at-risk', has serious ethical implications, as a positive result in a child's test automatically diagnoses the parent.

Parents and grandparents have to decide when and how to tell their children and grandchildren. The issue of disclosure also comes up when siblings are diagnosed with the disease, and especially in the case of identical twins.

For those at risk, or known to have the disease, consideration is necessary prior to having children due to the genetically dominant nature of the disease. In vitro and embryonic genetic screening now make it possible (with 99% certainty) to have an HD-free child; however, the cost of this process can easily reach tens of thousands of dollars. Another consideration regarding genetic testing is the fact that this kind of screening is a form of eugenics. Indeed, historically, Huntington's disease patients were one of the targets groups for the eugenic improvement of the human gene pool. The American scientist Charles Davenport proposed in 1910 that compulsory sterilization and immigration control be aimed at those afflicted with HD (amongst other diseases) [1]

Financial institutions are also faced with the question of whether to use genetic testing results when assessing an individual, e.g. for life insurance. Some countries' organisations have already agreed not to use this information.

Cultural references

HD has been depicted in a variety of media:

Books

  • Jacqueline Susann's 1966 American novel Valley of the Dolls (night club singer Tony Polar).
  • Kurt Vonnegut's 1985 American novel Galapagos.
  • Pål Johan Karlsen's 2002 Norwegian novel Daimler (main character Daniel Grimsgaard is afflicted).
  • Diane Tulson's 2002 American novel Saving Jasey (Trist, Jasey and their Grandfather).
  • Nancy Werlin's 2004 American novel Double Helix (Ava Samuels, Kayla Matheson and others).
  • Steven T. Seagle's autobiographical 2004 Anerican graphic-novel It's a Bird features the author coming to grips with the presence of HD in his family.
  • Ian McEwan's 2005 British novel Saturday. The character of Baxter is negatively portrayed in his affliction. [44].

Films

  • Arlo Guthrie's 1969 film Alice's Restaurant

Television

  • Dr. Samantha O'Hara from the Australian medical drama All Saints.
  • Hannah's father from the American drama Everwood, as revealed in the episode "Need to Know" (3x10).
  • Characters in the episodes "Pad'ar" (3x08) and "the Sins of the Father" (4x03) of Gene Roddenberry's Earth: Final Conflict.
  • Angie Padgett from the episode "In Which Charlotte Goes Down the Rabbit Hole" (1x06) of Private Practice.
  • The character known as "Thirteen", in the episode "You Don't Want to Know" (4x08) of House; she may have inherited the disease from her mother, who died of Huntington's, but refuses to be tested.
  • An episode of the BBC drama Waterloo Road

History

Research and discovery

  • c. 300: Along with other conditions with abnormal movements, it may have been referred to as St Vitus' dance. St Vitus is the Christian patron saint of epileptics who was martyred in 303.
  • Middle Ages: People with the condition were probably persecuted as being witches or as being possessed by spirits, and were shunned, exiled or worse. Some speculate that the "witches" in the Salem Witch Trials in 1692 had HD.[45]
  • 1860: One of the early medical descriptions of HD was made in 1860 by a Norwegian district physician, Johan Christian Lund. He noted that in Setesdalen, a remote and rather secluded area, there was a high prevalence of dementia associated with a pattern of jerking movement disorders that tended to run in families. This is the reason for the disease being commonly referred to as Setesdalsrykkja (Setesdalen=the location, rykkja=jerking movements) in Norwegian.
  • 1872: George Huntington was the third generation of a family medical practice in Long Island. With their combined experience of several generations of a family with the same symptoms, he realised their conditions were linked and set about describing it. A year after leaving medical school, in 1872, he presented his accurate definition of the disease to a medical society in Middleport, Ohio.
  • c. 1923: Smith Ely Jelliffe (1866–1945) and Frederick Tilney (1875–1938) began analyzing the history of HD sufferers in New England.
  • 1932: P. R. Vessie expanded Jelliffe and Tilney's work, tracing about a thousand people with HD back to two brothers and their families who left Bures in Essex for Suffolk bound for Boston in 1630.
  • 1979: The U.S-Venezuela Huntington's Disease Collaborative Research Project began an extensive study which gave the basis for the gene to be discovered. This was conducted in the small and isolated Venezuelan fishing villages of Barranquitas and Lagunetas. Families there have a high presence of the disease, which has proved invaluable in the research of the disease.
  • 1983: James Gusella, David Housman, P. Michael Conneally, Nancy Wexler, and their colleagues find the general location of the gene, using DNA marking methods for the first time—an important first step toward the Human Genome Project.
  • 1992: Anita Harding, et al., find that trinucleotide repeats affect disease severity[46]
  • 1993: The Huntington's Disease Collaborative Research Group isolates the precise gene at 4p16.3.
  • 1996: A transgenic mouse ([the R6 line]) was created that could be made to exhibit HD greatly advancing how much experimentation can be achieved.
  • 1997: DiFiglia M, Sapp E, Chase KO, et al., discover that mHtt aggregates (misfolds) to form nuclear inclusions.[47]
  • The full record of research is extensive.[48][49][50]

Organizations

  • 1967: Woody Guthrie's wife, Marjorie Guthrie, helped found the Committee to Combat Huntington's Disease, after his death whilst suffering from HD. This eventually became the Huntington's Disease Society of America.[51] Since then, lay organizations have been formed in many countries around the world.
  • 1968: After experiencing HD in his wife's family Dr. Milton Wexler was inspired to start the Hereditary Disease Foundation (HDF). Professor Nancy S. Wexler, Dr. Wexler's daughter, was in the research team in Venezeula and is now president of the HDF.
  • 1974: The first international meeting took place when the founders of the Canadian HD Society (Ralph Walker) and of the British HD Society (Mauveen Jones) attended the annual meeting of the American HD Society.
  • 1977: second meeting organized by the Dutch Huntington Society the "Vereniging van Huntington", representatives of six countries were present.
  • 1979: International Huntington Association (IHA) formed during international meeting in Oxford, England organized by HDA of England.
  • 1981–2001: Biennial meetings held by IHA which became the World Congress on HD.
  • 2003: The first World Congress on Huntington's Disease was held in Toronto[52]. This is a biennial meeting for associations and researchers to share ideas and research, which is held on odd-number years. The Euro-HD Network[53] was started as part of the Huntington Project,[54] funded by the High-Q Foundation.[55]

See also

References

  1. ^ Gaba AM, Zhang K, Marder K, Moskowitz CB, Werner P, and Boozer CN. Energy balance in early-stage Huntington disease. Am J Clin Nutr. 2005; 81(6):1335-41. PMID 15941884
  2. ^ Caregiver's Handbook for Advanced-Stage Huntington Disease. Booklet by the Huntington Society of Canada, retrieved 2007-04-11.
  3. ^ Johnson SA, Stout JC, Solomon AC, et al (2007). "Beyond disgust: impaired recognition of negative emotions prior to diagnosis in Huntington's disease". Brain 130 (Pt 7): 1732–44. doi:10.1093/brain/awm107. PMID 17584778.
  4. ^ RM Ridley, CD Frith, TJ Crow and PM Conneally (1988). "Anticipation in Huntington's disease is inherited through the male line but may originate in the female". Journal of Medical Genetics 25: 589-595.
  5. ^ Kieburtz K, MacDonald M, Shih C, Feigin A, Steinberg K, Bordwell K, Zimmerman C, Srinidhi J, Sotack J, Gusella J, et al. Trinucleotide Repeat Length and Progression of Illness in Huntington's Disease. J Med Genet. 1994; 31:872
  6. ^ Velier J, Kim M, Schwarz C, Kim T.W, Sapp E, Chase K, Aronin N, DiFiglia M. Wild-Type and Mutant Huntingtins Function in Vesicle Trafficking in the Secretory and Endocytic Pathways. Experimental Neurology 1998; 152:34
  7. ^ Waelter S, Scherzinger E, Hasenbank R, Nordhoff E, Lurz R, Goehler H, Gauss C, Sathasivam K, Bates G, Lehrach H, and Wanker E. The Huntingtin Interacting Protein HIP1 is a Clathrin and α–adaptin-binding Protein Involved in Receptor Mediated Endocytosis. Human Molecular Genetics 2001; 10:1807
  8. ^ Canals J, Pineda J, Torres-Peraza J, Bosch M, Martin-Ibanez R, Munoz M, Mengod G, Ernfors P, and Alberch J. Brain Derived Neurotrophic Factor Regulates the Onset and Severity of Motor Dysfunction Associated with Enkephalinergic Neuronal Degeneration in Huntington’s Disease. Neurobiology of Disease 2004; 24:7727
  9. ^ Sawa A, Nagata E, Sutcliffe S, Dulloor P, Cascio MB, Ozeki Y, Roy S, Ross CA, Snyder SH. Huntingtin is Cleaved by Caspases in the Cytoplasm and Translocated to the Nucleus via Perinuclear Sites in Huntington's Disease Patient Lymphoblasts. Neurobiol Dis 2005
  10. ^ Kim YJ, Yi Y, Sapp E, Wang Y, Cuiffo B, Kegel KB, Qin ZH, Aronin N, DiFiglia M. Caspase 3-cleaved N-terminal Fragments of Wild-type and Mutant Huntingtin are Present in Normal and Huntington's Disease Brains, Associate with Membranes, and Undergo calpain-dependent proteolysis. Proc Natl Acad Sci U S A. 2001; 98:12784
  11. ^ Hermel E, Gafni J, Propp SS, Leavitt BR, Wellington CL, Young JE, Hackam AS, Logvinova AV, Peel AL, Chen SF, Hook V, Singaraja R, Krajewski S, Goldsmith PC, Ellerby HM, Hayden MR, Bredesen DE, Ellerby LM. Specific Caspase Interactions and Amplification are Involved in Selective Neuronal Vulnerability in Huntington's Disease. Cell Death Differ 2004; 11:424
  12. ^ Freiman R, and Tjian R. A Glutamine-Rich Trail Leads to Transcription Factors. Science 2002; 296:2149
  13. ^ Bae B.I, Xu H, Igarashi S, Fujimuro M, Agrawal N, Taya Y, Hayward S.D, Moran T.H, Montell C, Ross C.A, Snyder S.H, and Sawa A. p53 Mediates Cellular Dysfunction and Behavioral Abnormalities in Huntington's Disease. Neuron 2005; 47:29
  14. ^ Dunah A, Jeong H, Griffin A, Kim Y, Standeart D, Hersch S, Mouradian M, Young A, Tanese N, and Krainc D. Sp1 and TAFII130 Transcriptional Activity Disrupted in Early Huntington’s Disease. Science 2002; 296: 2238
  15. ^ Graham RK, Deng Y, Slow EJ, et al (2006). "Cleavage at the caspase-6 site is required for neuronal dysfunction and degeneration due to mutant huntingtin". Cell 125 (6): 1179–91. doi:10.1016/j.cell.2006.04.026. PMID 16777606.
  16. ^ Murphy K, Carter R, Lione L, Mangiarini L, Mahal A, Bates G, Dunnett S, and Morton J. Abnormal Synaptic Plasticity and Impaired Spatial Cognition in Mice Transgenic for Exon 1 of the Human Huntington’s Disease Mutation. Journal of Neuroscience 2000; 20:5115
  17. ^ Difiglia M, Sapp E, Chase E, Davies K. et al. Aggregation of Huntingtin in Neuronal Intranuclear Inclusions and Dystrophic Neurites in Brain. Science 1997
  18. ^ Davies S, Turmaine M, Cozens B, Difiglia M, Sharp A, Ross C, Scherzinger E, Wanker E, Mangiarini L, and Bates G. Formation of Neuronal Intranuclear Inclusions Underlies the Neurological Dysfunction in Mice Transgenic for the HD Mutation. Cell 1997; 90:537
  19. ^ Difiglia M, Sapp E, Chase E, Davies K. et al. Aggregation of Huntingtin in Neuronal Intranuclear Inclusions and Dystrophic Neurites in Brain. Science 1997
  20. ^ Gutekunst CA, Li SH, Yi H, Mulroy JS, Kuemmerle S, Jones R, Rye D, Ferrante RJ, Hersch SM, and Li XJ. Nuclear and neuropil aggregates in Huntington's disease: Relationship to neuropathology. J Neurosci 19: 2522-2534, 1999.
  21. ^ Sieradzan KA, Mechan AO, Jones L, Wanker EE, Nukina N, and Mann DM. Huntington's disease intranuclear inclusions contain truncated, ubiquitinated huntingtin protein. Exp Neurol 156: 92-99, 1999.
  22. ^ Cooper JK, Schilling G, Peters MF, Herring WJ, Sharp AH, Kaminsky Z, Masone J, Khan FA, Delanoy M, Borchelt DR, Dawson VL, Dawson TM, Ross CA. Truncated N-terminal Fragments of Huntingtin with Expanded Glutamine Repeats Form Nuclear and Cytoplasmic Aggregates in Cell Culture. Hum Mol Genet. 1998; 7:783
  23. ^ F. R. Fusco, Q. Chen, W. J. Lamoreaux, G. Figueredo-Cardenas, Y. Jiao, J. A. Coffman, D. J. Surmeier, M. G. Honig, L. R. Carlock, and A. Reiner. Cellular Localization of Huntingtin in Striatal and Cortical Neurons in Rats: Lack of Correlation with Neuronal Vulnerability in Huntington's Disease. J. Neurosci 1999; 19: 1189
  24. ^ Saudou F, Finkbeiner S, Devys, and Greenberg M. Huntingtin Acts in the Nucleus to Induce Apoptosis but Death Does Not Correlate with the Formation of Intranuclear Inclusions. Cell. 1998; 95:55
  25. ^ www.hdsa.org
  26. ^ clinicaltrials.gov
  27. ^ Lecerf, J. M., Shirley, T. L., Zhu, Q., Kazantsev, A., Amersdorfer, P., Housman, D. E., Messer, A. & Huston, J. S. Human single-chain Fv intrabodies counteract in situ huntingtin aggregation in cellular models of Huntington's disease. (2001) Proc. Natl. Acad. Sci. USA 98, 4764-4769.
  28. ^ Miller, T. W., Zhou, C., Gines, S., MacDonald, M. E., Mazarakis, N. D., Bates, G. P., Huston, J. S. & Messer, A. A human single-chain Fv intrabody preferentially targets amino-terminal huntingtin fragments in striatal models of Huntington's disease (2005) Neurobiol. Dis. 19, 47-56.
  29. ^ Wolfgang WJ, Miller TW, Webster JM, Huston JS, Thompson LM, Marsh JL, Messer A. Suppression of Huntington's disease pathology in Drosophila by human single-chain Fv antibodies. (2005) Proc Natl Acad Sci USA 32:11563-8.
  30. ^ Miller, T. W., Messer, A. Intrabody Applications in Neurological Disorders: Progress and Future Prospects. (2005) Molecular Therapy. 12, 394-401.
  31. ^ Harper SQ, Staber PD, He X, et al (2005). "RNA interference improves motor and neuropathological abnormalities in a Huntington's disease mouse model". Proc. Natl. Acad. Sci. U.S.A. 102 (16): 5820–5. doi:10.1073/pnas.0501507102. PMID 15811941.
  32. ^ Miguel Díaz-Hernández, Jesús Torres-Peraza, Alejandro Salvatori-Abarca, María A. Morán, Pilar Gómez-Ramos, Jordi Alberch, and José J. Lucas (October 19, 2005). "Full Motor Recovery Despite Striatal Neuron Loss and Formation of Irreversible Amyloid-Like Inclusions in a Conditional Mouse Model of Huntington's Disease". The Journal of Neuroscience 25 (42): 9773-9781. Retrieved on 2006-07-16.
  33. ^ World health Article
  34. ^ Andrew SE, Goldberg YP, Kremer B, Telenius H, Theilmann J, Adam S, Starr E, Squitieri F, Lin B, Kalchman MA, Graham RK, and Hayden MR. The relationship between trinucleotide (CAG) repeat length and clinical features of Huntington's disease. Nat Genet 4: 398-403, 1993.
  35. ^ Harper PS. Huntington's disease: A clinical, genetic and molecular model for polyglutamine repeat disorders. Philos Trans R Soc Lond B Biol Sci 354: 957-961, 1999.
  36. ^ Rubinsztein DC, Leggo J, Chiano M, Dodge A, Norbury G, Rosser E, and Craufurd D. Genotypes at the GluR6 kainate receptor locus are associated with variation in the age of onset of Huntington disease. Proc Natl Acad Sci U S A 94: 3872-3876, 1997.
  37. ^ Adams P, Falek A, Arnold J (1988). "Huntington disease in Georgia: age at onset". Am. J. Hum. Genet. 43 (5): 695–704. PMID 2973230.
  38. ^ Kremer B. Clinical neurology of Huntington's disease. In: Huntington's Disease (Third ed.), edited by Bates GP, Harper PS and Jones L. Oxford: Oxford University Press, 2002, p. 28-61.
  39. ^ Roos RA, Hermans J, Vegter-van der Vlis M, van Ommen GJ, and Bruyn GW. Duration of illness in Huntington's disease is not related to age at onset. J Neurol Neurosurg Psychiatry 56: 98-100, 1993.
  40. ^ Lanska DJ, Lanska MJ, Lavine L, and Schoenberg BS. Conditions associated with Huntington's disease at death. A case-control study. Arch Neurol 45: 878-880, 1988.
  41. ^ Di Maio L, Squitieri F, Napolitano G, Campanella G, Trofatter JA, and Conneally PM. Suicide risk in Huntington's disease. J Med Genet 30: 293-295, 1993.
  42. ^ Kremer B. Clinical neurology of Huntington's disease. In: Huntington's Disease (Third ed.), edited by Bates GP, Harper PS and Jones L. Oxford: Oxford University Press, 2002, p. 28-61.
  43. ^ Schoenfeld M, Myers RH, Cupples LA, Berkman B, Sax DS, and Clark E. Increased rate of suicide among patients with Huntington's disease. J Neurol Neurosurg Psychiatry 47: 1283-1287, 1984.
  44. ^ Prejudice in a portrayal of Huntington's disease, by Nancy S Wexler and Michael D Rawlins, in The Lancet, Vol 366 September 24,2005
  45. ^ The brief history of HD on stanford.edu
  46. ^ La Spada AR, Roling DB, Harding AE, et al (1992). "Meiotic stability and genotype-phenotype correlation of the trinucleotide repeat in X-linked spinal and bulbar muscular atrophy". Nat. Genet. 2 (4): 301–4. doi:10.1038/ng1292-301. PMID 1303283.
  47. ^ DiFiglia M, Sapp E, Chase KO, et al (1997). "Aggregation of huntingtin in neuronal intranuclear inclusions and dystrophic neurites in brain". Science 277 (5334): 1990–3. PMID 9302293.
  48. ^ Achievements of Hereditary Disease Foundation
  49. ^ HDA research news—medical research into treatment & prevention on hda.org.uk
  50. ^ Bates G, Harper PS, Jones L (2002) Huntington's disease, 3rd Edition. Oxford: Oxford University Press.
  51. ^ Huntington's Disease Society of America
  52. ^ World Congress on Huntington's Disease
  53. ^ Euro-HD Network
  54. ^ Huntington Project
  55. ^ High-Q Foundation

Sources

  • Conomy, John P., M.D., J.D. Dr. George Sumner Huntington and the Disease Bearing His Name.
  • Huntington, G. (1872-04-13). "On Chorea". Medical and Surgical Reporter of Philadelphia 26 (15): 317-321.
  • Online Mendelian Inheritance in Man, Article 143100 - Huntington Disease, Johns Hopkins University
  • Online Mendelian Inheritance in Man, Article 606438 - Huntingtons Disease-Like 2, Johns Hopkins University
  • Stevenson, Charles S. (April 1934). "A Biography of George Huntington, M.D." (Microsoft Word). Bulletin of the Institute of the History of Medicine II (2). Johns Hopkins University. Retrieved on 2007-12-05.
  • Vessie, P. R. (1932). "On the transmission of Huntington's chorea for 300 years—the Bures family group". Journal of Nervous and Mental Disease, Baltimore 76: 553–573.

Bibliography

  • Bates, Gillian, Peter Harper, and Lesley Jones (2002). Huntington's Disease - Third Edition. Oxford: Oxford University Press. ISBN 0-19-851060-8. 

Support and advocacy

  • The International Huntington Association - Coordinates support organizations in 39 countries, and individual contacts in others.
  • Hereditary Disease Foundation - Spearheaded Venezuela Collaborative Huntington's Disease Project.
  • Huntington's Disease Lighthouse - Reports on the latest research and studies.
  • Huntington's Disease Advocacy Center - Provides community support through shared stories and forums.
  • Huntington's Disease Support Club
  • Huntington's Disease Society of America
  • Huntington Society of Canada - Resource materials, latest in Canadian and International HD research and description of comprehensive services portfolio to help families struggling with HD
  • European HD Network
  • Australian Huntington's Disease Association (NSW) Inc.
  • Huntington's Disease Association UK

Other

  • Mind Matters: RTÉ Radio 1 programme on Huntington's Disease, featuring a family affected from Ireland. Podcast feed: www.rte.ie/radio1/podcast/podcast_mindmatters.xml
  • Huntington's Disease Drug Works - Website covering results and progress of clinical trials and alternative treatments for HD.
  • WeAreHD.org - Online Community for those living with Huntington's Disease.


 
This article is licensed under the GNU Free Documentation License. It uses material from the Wikipedia article "Huntington's_disease". A list of authors is available in Wikipedia.
Your browser is not current. Microsoft Internet Explorer 6.0 does not support some functions on Chemie.DE