Spinal and bulbar muscular atrophy

Last updated
Spinal and bulbar muscular atrophy
Other namesKennedy's disease (KD), spinobulbar muscular atrophy, bulbo-spinal atrophy, X-linked bulbospinal neuropathy (XBSN), X-linked spinal muscular atrophy type 1 (SMAX1), and many other names [1]
Impacts of SBMA2.svg
This disorder is inherited via X-linked recessive manner
Specialty Neurology   OOjs UI icon edit-ltr-progressive.svg
Symptoms Weakness of limb and bulbar muscles, tremor, fasciculations, muscle cramps, dysarthria and dysphagia
CausesMutation in the AR gene
Diagnostic method Number of CAG repeats in the AR gene
TreatmentSupportive Care

Spinal and bulbar muscular atrophy (SBMA), popularly known as Kennedy's disease, is a rare, adult-onset, X-linked recessive lower motor neuron disease caused by trinucleotide CAG repeat expansions in exon 1 of the androgen receptor (AR) gene, which results in both loss of AR function and toxic gain of function. [2] [3]

Contents

In men, the disease slowly progresses over decades with bulbar and lower motor neuron loss, muscle denervation, and direct skeletal muscle involvement. [4] [5]  The disease causes progressive muscle loss with weakness, fasciculations, and cramps.  Weakness of the bulbar muscles follows causing difficulties in speech (dysarthria) and swallowing (dysphagia). Female carriers do not show symptoms.  Although there is no cure, supportive intervention can improve mobility and reduce complications. The prevalence of SBMA has been estimated at 2.6:100,000 males. [6]

There is no known cure for SBMA. [7] [8] [9] Supportive care is focused on preventing disease complications and maintaining independence. 

Signs and symptoms

Neuromuscular symptoms include muscle weakness and wasting of the limb, bulbar and respiratory muscles, tremor, fasciculations, muscle cramps, speech and swallowing difficulties, decreased or absent deep tendon reflexes, and sensory neuropathy. Other manifestations of SBMA include androgen insensitivity (gynecomastia, erectile dysfunction, reduced fertility, testicular atrophy), and metabolic impacts (glucose resistance, hyperlipidemia, fatty liver disease). [3] [5] [10]

Neuromuscular effects

SBMA patients develop limb weakness which often begins in the pelvic or shoulder regions between 30 and 50 years of age. [11] Muscle strength declines slowly, at a rate of approximately 2% per year based on quantitative muscle assessment. [11] [12] Muscle weakness often begins in proximal muscles, with most patients first noticing weakness in their lower limbs. [3] [13]

Tremor, fasciculations, and cramps are common early symptoms of SBMA. Tremor is an involuntary, somewhat rhythmic, muscle contraction and relaxation involving oscillations or twitching movements. In SBMA patients, tremor is most common in the hands, but also occur in the head, voice and lower limbs, and may be observed ten years prior to muscle weakness. [11] [3] Fasciculation, or fleeting muscle twitches visible under the skin, is a spontaneous, involuntary muscle contraction and relaxation, and is especially noticeable in the face and tongue of SBMA patients. Lingual atrophy occurs later in the course of the disease, but the tongue may develop an unusual shape due to coexisting denervation and reinnervation. A cramp is a sudden, involuntary, painful skeletal muscle contraction of skeletal muscle, and common in motor neuron disorders.

Bulbar symptoms (weakness of the facial and tongue muscles) typically follow limb manifestations and may start with difficulty with speech articulation (dysarthria) before swallowing difficulty (dysphagia). [3] [5] [11] Dysarthria is common, with hypernasality due to incomplete lifting of soft palate, but typically remains relatively mild and seldom leads to the loss of oral communication. [3] Signs of dysphagia include difficulty controlling solids, liquids or saliva in the mouth, coughing and choking. As the disease progresses, there is increased risk of aspiration pneumonia, which is the leading cause of death in SBMA patients. [3] Up to half of all patients experience laryngospasm, an uncontrolled contraction (spasm) of the vocal folds, with a sense of choking, and feel that the air cannot enter or exit the airways for long seconds. Laryngospasms are often followed by a high-pitched breathing sound (stridor) due to rapid and vigorous contraction of the laryngeal sphincters. This is a frightening and hugely distressing experience, but rarely escalates to prolonged or serious episodes. [3]

In SBMA, deep tendon reflexes are diminished or absent. Sensory involvement results in degeneration of dorsal root ganglion cells with reduced vibratory sensation (distally in the legs), neuropathic pain, and numbness. [11] There are sporadic reports of certain psychological traits (lack of self-confidence, emotional flattening, and poor concentration), but detailed neuropsychological examination of 64 SBMA patients did not detect any abnormalities. [3] [14]

Androgen insensitivity

Loss of AR function in SBMA patients results in partial androgen insensitivity, including gynecomastia, erectile dysfunction, decreased libido, infertility and testicular atrophy. [3] [10] Patients with complete androgen insensitivity syndrome do not show symptoms of SBMA, indicating neuromuscular symptoms are not caused by a loss of AR function. [11]

Gynecomastia, excessive enlargement of male breasts, is observed in about three quarters of SBMA patients. It typically becomes apparent after puberty, and is often the first evidence of this disease. [10] Erectile dysfunction, decreased libido, infertility and testicular atrophy are common. [15] One of the most disease-specific endocrine indices of SBMA is the androgen sensitivity index (luteinizing hormone / testosterone) which is elevated in 64% of cases, indicating both endocrine and exocrine testicular dysfunction. [3] [15]

Metabolic disturbances

Metabolic disturbances have also been reported in SBMA patients, with increased risk of insulin resistance, fatty liver disease, hyperlipidemia, and electrocardiogram (ECG) abnormalities. Impaired glucose homeostasis is a common feature of SBMA, and recent study found a significant correlation between insulin resistance and motor dysfunction in SBMA. [11] [16] In a group of 22 patients with SBMA, evidence of fatty liver disease was detected in all individuals by magnetic resonance spectroscopy. In a second group, liver dome magnetic resonance spectroscopy measurements were increased in participants with SBMA relative to age- and sex-matched controls. [3] [17] SBMA patients may have higher frequency of Brugada syndrome and other electrocardiogram (ECG) abnormalities, which if not detected, can lead to sudden death. There are no reports of cardiomyopathy. However, there are indications that SBMA patients may be more likely to have high blood pressure and elevated total cholesterol and triglycerides. [11] [3]

Natural history

Progression of muscle weakness is slow, with an approximately 2% decrease in muscle strength by quantitative muscle testing (QMT) per year. [11] [12]

The most extensive dataset on disease progression is a study of 223 Japanese patients where milestones in nine activities of daily living (ADL) was observed for up to 20 years. [3] [10] [11] [18] Muscle weakness was first noticed in the lower extremities (71%), upper extremities in (31%), bulbar symptoms (11%), and facial weakness (2%), with some patients observing initial muscle weakness simultaneously in two locations. [18] Hand tremor was noticed first (median age: 33 years), followed by muscular weakness (median = 44 years), need for handrail to ascend stairs (49 years), dysarthria (50 years), dysphagia (54 years), use of a cane (59 years), and wheelchair use (61 years). [18] Twenty-one patients developed pneumonia (median age: 62 years) and 15 of them died (median = 65 years). The most common cause of death in these cases was pneumonia and respiratory failure. [18] There was considerable variation in the age of onset for each milestone, often varying by 25 to 30 years between the 10th to 90th percentile ranges. The ages at onset of each ADL milestone were strongly correlated with the length of CAG repeats in the AR gene. However CAG-repeat length did not correlate with the time intervals between each ADL milestone, suggesting that the rate of disease progression does not correlate with CAG-repeat length. [18] The average time interval between muscle weakness and handrail use was 5–6 years, between weakness and cane use was 9–11 years, between weakness and wheelchair use was 12–13 years, and between weakness and death was 10–20 years. [18]

Mechanism

Androgen receptor 2AM9.png
Androgen receptor

Molecular mechanism

SBMA is caused by a trinucleotide repeat expansion in the first exon of the androgen receptor (AR) gene. [9] [8] The AR gene, located in the X chromosome, contains a CAG repeat that encodes a polyglutamine tract in the androgen receptor protein. [19] [9] The tract normally varies from about 11 to 33 repeats; however, in SBMA patients, the tract contains 38 to 68 CAG repeats. The expanded series of CAG repeats in SBMA results in production of a toxic androgen receptor protein with an expanded polyglutamine tract called polyQ-AR. The repeat expansion likely causes a toxic gain of function in the receptor protein, since loss of receptor function in androgen insensitivity syndrome does not cause motor neuron degeneration. [20]

Multiple studies have demonstrated that in SBMA, CAG repeat length inversely correlates with the age of symptom onset, but not with the rate of disease progression. [3] [11] [21] [18] [22] Individuals with longer CAG expansions reach ADL milestones earlier (handrail, cane, wheelchair-bound, death) and eventually develop more severe disease manifestations. [8] CAG repeat numbers account for about 60% of the observed variation in motor disability, indicating other factors may play important roles disease progression. [21]

The mechanisms which lead to neurodegeneration in SBMA are not fully understood. [8] In SBMA, polyQ-AR misfolds and aggregates, leading to formation of nuclear inclusions (NIs) and diffuse accumulation of AR in the nucleus.  This aggregation of the mutant protein is dependent on the presence of testosterone, the ligand for the androgen receptor.  Female carriers of SBMA express polyQ-AR are generally asymptomatic or manifest only mild symptoms, due to lower levels of testosterone. Similarly, in animal models of SBMA castration dramatically reduces disease phenotype. [11] Toxicity is believed to occur through multiple cellular mechanisms including transcriptional dysregulation, mitochondrial dysfunction, disruption of protein homeostasis and cellular signaling pathways, as well as autophagy. [11]

SBMA is member of a group of diseases caused by polyglutamine expansion, including Huntington's disease and six types of spinocerebellar ataxia.   Common characteristics of polyglutamine (or polyQ) diseases is the accumulation of polyQ proteins and the progressive degeneration of nerve cells, usually affecting people later in life. However different polyQ-containing proteins damage different subsets of neurons, leading to different symptoms. [23]

Role of skeletal muscle in disease pathophysiology

SBMA has traditionally been considered primarily a disease of the motor neuron. Motor neurons degeneration is seen in the anterior horn of the spinal cord and brainstem, and electrophysiology studies show evidence of motor neuron dysfunction. However, several studies have suggested skeletal muscle plays an important role in SBMA pathophysiology. [24] Myogenic abnormalities in patient muscle include atrophic and morphologically abnormal muscle fibers, fiber-type grouping, and centralized nuclei. [11] [3] Serum creatine kinase (CK) levels are much higher than would be expected for a purely neurogenic disease. [11]

In animal studies, m ice which express the polyglutamine androgen receptor in all tissues were shown to develop progressive neuromuscular degeneration mimicking SBMA. However, when the mice were genetically manipulated to express the protein in all tissues except skeletal muscle, muscle atrophy, neuromuscular degeneration, and survival were significantly improved. [24] Further, treatment of mouse models of SBMA with antisense oligonucleotides targeting the polyglutamine androgen receptor reduced disease burden when administered subcutaneously though they could not cross the blood brain barrier. [24] However, when administered intrathecally into the CNS, disease was not rescued. [24]

Inheritance patterns in SBMA X-linked recessive (2).svg
Inheritance patterns in SBMA

Inheritance

SBMA is a hereditary syndrome, inherited in an X-linked recessive manner. [4] It is transmitted to offspring through the expanded (mutant) CAG repeat in the AR gene of the X chromosome. If the person with the mutant AR gene is a father, he cannot pass the gene on to his sons. However, he will pass the mutant gene on to all daughters. If the person with the mutant gene is female, she is a carrier. With a carrier, there is a 50% chance that the mutant gene will be passed to any offspring, male or female. [3]

Genetic founder effects are likely to be responsible for the higher prevalence of SBMA observed in certain geographic regions. [25]

Diagnosis

Diagnosis of SBMA is established by genetic testing that identifies a CAG trinucleotide repeat expansion in the AR gene. [5] If more than 38 CAG repeats are identified, it is usually considered confirmatory of SBMA. [3] However, there is evidence for reduced penetrance with 36-37 CAG repeats. [26]

Management

There is no known cure for SBMA. [7] [8] [9] Supportive care is focused on preventing disease complications (falls, fractures, aspiration) and maintaining independence.  Early interventions include occupational therapy to maintain mobility and activities of daily living.  Common mobility aids include canes, mobility scooters, power wheelchairs, ramps, and stair lifts.  As the disease progresses, swallowing difficulties (dysphagia) become more common and patients are at increased risk of aspiration pneumoniaSwallowing interventions include dietary modifications (adjust food texture and consistency), postural techniques, and swallowing maneuvers. Respiratory therapy may be helpful to reduce respiratory tract infections and mobilize phlegm. [3]

Exercise

Exercise has been shown to provide a variety of benefits in several neuromuscular diseases. However, in SBMA patients, exercise can have both positive and negative impacts. [27] In a study with 8 SBMA patients, moderate-intensity exercise was not well tolerated, and exercise frequency was decreased prior to the end of the training program.  Levels of creatine kinase (CK), a biomarker of muscle degeneration, were found to increase during intense exercise, indicating primary myopathy. [28] [29] In a study of home-based functional exercise with 50 SBMA patients, both the low-intensity exercise and stretching control groups tolerated exercise well, but there was no significant difference in primary and secondary outcomes between the two groups. [11] [29] However, subgroup analysis did show benefit for a low-functioning group, suggesting there may be a role for exercise in these patients. [11] In a third study, high-intensity interval training (HIIT) for less than one hour per week, improved both VO2max and performed workload, without increasing CK levels, self-rated fatigue and pain. [27] [30] Overall, these results suggest that exercise programs should be individually tailored, and SBMA patients must be carefully monitored for maladaptive biomarkers (increasing CK levels) to prevent muscle damage and worsening of disease phenotypes. [27]

History

SBMA was first described in Japanese literature in 1897 by Hiroshi Kawahara in a case report detailing progressive bulbar palsy in two brothers. [31] [9] Information on the clinical course, X-linked inheritance patterns, and key pathologic features was later documented by William R. Kennedy in 1968. [4] Onset of disease in mid-life and lack of symptoms in heterozygous female carriers was further described by Anita Harding in 1982. [9] In 1986, the causative gene of SBMA was shown to be present on the proximal arm of the X chromosome by Kurt Fischbeck, [31] though the exact gene causing SBMA had not yet been characterized. In 1991, it was discovered that the AR gene is involved in the disease process, and that expansion of a CAG repeat in the AR gene causes disease. [5]

Research directions

Research in SBMA is broad, and covers a number of aspects of the disorder. Below is a summary of a few areas of ongoing research in SBMA:

Aggregation

The polyglutamine androgen receptor does not fold properly, and subsequently forms protein aggregates with other proteins. This is a gain of new function conferred by the polyglutamine tract, as the non-polyglutamine expanded androgen receptor does not form these aggregates. [24] Several proteins key to normal cellular function have been found to be sequestered[ clarification needed ] within these aggregates, including CREB-BP, Hsp70, Hsp40, and components of the ubiquitin proteasome system. [11] It is thought that loss of sufficient supply of these and other key proteins contributes to the pathogenesis of the disease, though further research is ongoing. [24]

Post-translational modifications

Following its transcription and translation, the androgen receptor is modified with a number of post-translational modifications, including phosphorylation, methylation, acetylation, and SUMOylation. The polyglutamine androgen receptor has been found to have different levels of some these post-translational modifications. [11] Further, altering the levels of certain post-translational modifications of the mutant androgen receptor has altered the degree of toxicity in cellular and animal models, suggesting they may be a target for further research and therapeutic development. [11]

Clinical trials

Leuprorelin

Leuprorelin, a GnRH agonist which blocks the synthesis of testosterone when given continuously, was initially shown to be effective at improving motor function in mouse models of SBMA. [11] A small pilot study was performed in which five SBMA patients were given subcutaneous injections of leuprorelin every four weeks for six months, with serial scrotal skin biopsies performed. [28] Nuclear accumulation of polyglutamine androgen receptor was significantly reduced in patient scrotal biopsies. Additionally, serum CK, a marker of muscle deterioration, and testosterone levels were both reduced in patients receiving leuprorelin. [28] Notably, in this study patients were not randomized to treatment groups or placebo controlled. [28]

A subsequent larger study consisted of fifty SBMA patients randomized to either leuprorelin treatment or placebo. This trial spanned 48 weeks of treatment with treatment occurring every four weeks initially. Following treatment, thirty-four patients had an open-label follow-up spanning an additional ninety-six weeks with treatment continuing every twelve weeks. [11] [10] At the forty-eight week mark, there was no significant difference in the ALS functional rating scale, the primary outcome measure of the study, between placebo and leuprorelin treated groups. [28] [10] There was improvement in swallowing a barium contrast marker, a secondary endpoint of the study, at forty-eight weeks. [28] Further, there was improvement in the ALS functional rating scale at the 96 and 144 week marks, suggesting a longer period may be needed to see effects of leuprorelin. [28]

A larger, multi-center, placebo-controlled, double blind study was then conducted which contained 199 SBMA patients who were randomized to either placebo or leuprorelin treatment. The study spanned forty-eight weeks with leuprorelin treatment every 12 weeks, with ability to swallow a barium contrast marker as the primary endpoint. [28] [11] In this study, there was not a significant difference in barium swallow at forty-eight weeks. [28] [10] There was also no difference in other video fluorography measurements, supporting the lack of improvement in swallow function in the treatment group. [28] Other secondary measures, such as number of AR positive scrotal cells and serum CK level were significantly different in the treatment group. [28] Though the primary endpoint of the study did not show an effect of leuprorelin on SBMA patients, a subgroup analysis performed on patients who had symptoms for ten years or less did show improved swallowing function, and it was suggested that treatment may be more effective in patients who have shown symptoms for shorter periods of time or with less advanced disease. [28]

A recent follow-up open-label study compared thirty-six patients treated with leuprorelin to nontreated controls over eighty four months, and found that the treated group showed slower decline in motor function than the non-treated group. [11] Significantly differing endpoints of this study included the ALS Functional Rating Scale, the Limb Norris Score, and the Norris Bulbar Score. [11]

Dutasteride

Dutasteride is a 5α-reductase inhibitor which blocks the conversion of testosterone into dihydrotestosterone, reducing overall androgen receptor activation. Fifty patients were recruited to a randomized, placebo controlled trial spanning two years, with a primary endpoint of quantitative muscle assessment. [28] No significant difference was found in quantitative muscle assessment between the placebo and dutasteride groups at the two year mark. [28] Secondary outcomes including barium swallow and manual muscle testing also showed no significant difference between groups. [11]

BVS857

IGF-1, a signaling molecule downstream of growth hormone, has well established functions promoting skeletal muscle growth. [11] In preclinical studies on mouse models of amyotrophic lateral sclerosis, IGF-1 was shown to be protective against motor neuron death. [11] A double blind, placebo-controlled clinical trial was performed with eighteen SBMA patients receiving BVS857, a mimetic of IGF-1, with nine placebo controls. [11] The study found increased thigh muscle volume improved lean body mass following 12 weeks of treatment in the BVS857 group compared to placebo. [11] However, eleven of eighteen patients were found to have an immune response against BVS857, with five patients developing neutralizing antibodies, posing a challenge for long-term treatment. [11]

Society and culture

In 2000, the Kennedy's Disease Association [32] [33] [34] was founded by Susanne and Terry Waite and Patrick Griffin to help find effective treatments and a cure for SBMA. The organization is managed and operated by an all-volunteer board of directors, and grown to over 1,600 associates from over 50 countries. Donations from patients and families provide funding to support research (90%), education (5%) and operating expenses (5%). [34] KDA has awarded grants and fellowships to researchers in the United States, Canada, Britain, Italy, and Japan to support both basic research and clinical trials on the causes and potential treatments for SBMA.  [35] The KDA website states that they focus on providing "seed-money" to post-doc and other young researchers to start working on SBMA, collecting preliminary data that can be used to support larger proposals to governmental and philanthropic organizations.  In 2022, the KDA Waite-Griffin Fellowship was established to encourage young researchers to include research on SBMA in their future career plans. [35]

See also

Related Research Articles

<span class="mw-page-title-main">Motor neuron diseases</span> Group of neurological disorders affecting motor neurons

Motor neuron diseases or motor neurone diseases (MNDs) are a group of rare neurodegenerative disorders that selectively affect motor neurons, the cells which control voluntary muscles of the body. They include amyotrophic lateral sclerosis (ALS), progressive bulbar palsy (PBP), pseudobulbar palsy, progressive muscular atrophy (PMA), primary lateral sclerosis (PLS), spinal muscular atrophy (SMA) and monomelic amyotrophy (MMA), as well as some rarer variants resembling ALS.

<span class="mw-page-title-main">Charcot–Marie–Tooth disease</span> Neuromuscular disease

Charcot–Marie–Tooth disease (CMT) is a hereditary motor and sensory neuropathy of the peripheral nervous system characterized by progressive loss of muscle tissue and touch sensation across various parts of the body. This disease is the most commonly inherited neurological disorder, affecting about one in 2,500 people. It is named after those who classically described it: the Frenchman Jean-Martin Charcot (1825–1893), his pupil Pierre Marie (1853–1940), and the Briton Howard Henry Tooth (1856–1925).

<span class="mw-page-title-main">Androgen insensitivity syndrome</span> Medical condition

Androgen insensitivity syndrome (AIS) is a difference in sex development involving hormonal resistance due to androgen receptor dysfunction.

Hypotonia is a state of low muscle tone, often involving reduced muscle strength. Hypotonia is not a specific medical disorder, but a potential manifestation of many different diseases and disorders that affect motor nerve control by the brain or muscle strength. Hypotonia is a lack of resistance to passive movement, whereas muscle weakness results in impaired active movement. Central hypotonia originates from the central nervous system, while peripheral hypotonia is related to problems within the spinal cord, peripheral nerves and/or skeletal muscles. Severe hypotonia in infancy is commonly known as floppy baby syndrome. Recognizing hypotonia, even in early infancy, is usually relatively straightforward, but diagnosing the underlying cause can be difficult and often unsuccessful. The long-term effects of hypotonia on a child's development and later life depend primarily on the severity of the muscle weakness and the nature of the cause. Some disorders have a specific treatment but the principal treatment for most hypotonia of idiopathic or neurologic cause is physical therapy and/or occupational therapy for remediation.

<span class="mw-page-title-main">X-linked recessive inheritance</span> Mode of inheritance

X-linked recessive inheritance is a mode of inheritance in which a mutation in a gene on the X chromosome causes the phenotype to be always expressed in males and in females who are homozygous for the gene mutation, see zygosity. Females with one copy of the mutated gene are carriers.

<span class="mw-page-title-main">Androgen receptor</span> Mammalian protein found in humans

The androgen receptor (AR), also known as NR3C4, is a type of nuclear receptor that is activated by binding any of the androgenic hormones, including testosterone and dihydrotestosterone, in the cytoplasm and then translocating into the nucleus. The androgen receptor is most closely related to the progesterone receptor, and progestins in higher dosages can block the androgen receptor.

<span class="mw-page-title-main">Spinocerebellar ataxia</span> Medical condition

Spinocerebellar ataxia (SCA) is a progressive, degenerative, genetic disease with multiple types, each of which could be considered a neurological condition in its own right. An estimated 150,000 people in the United States have a diagnosis of spinocerebellar ataxia at any given time. SCA is hereditary, progressive, degenerative, and often fatal. There is no known effective treatment or cure. SCA can affect anyone of any age. The disease is caused by either a recessive or dominant gene. In many cases people are not aware that they carry a relevant gene until they have children who begin to show signs of having the disorder.

Trinucleotide repeat disorders, also known as microsatellite expansion diseases, are a set of over 50 genetic disorders caused by trinucleotide repeat expansion, a kind of mutation in which repeats of three nucleotides increase in copy numbers until they cross a threshold above which they become unstable. Depending on its location, the unstable trinucleotide repeat may cause defects in a protein encoded by a gene; change the regulation of gene expression; produce a toxic RNA, or lead to chromosome instability. In general, the larger the expansion the faster the onset of disease, and the more severe the disease becomes.

<span class="mw-page-title-main">Congenital muscular dystrophy</span> Medical condition

Congenital muscular dystrophies are autosomal recessively-inherited muscle diseases. They are a group of heterogeneous disorders characterized by muscle weakness which is present at birth and the different changes on muscle biopsy that ranges from myopathic to overtly dystrophic due to the age at which the biopsy takes place.

<span class="mw-page-title-main">Spinal muscular atrophy</span> Rare congenital neuromuscular disorder

Spinal muscular atrophy (SMA) is a rare neuromuscular disorder that results in the loss of motor neurons and progressive muscle wasting. It is usually diagnosed in infancy or early childhood and if left untreated it is the most common genetic cause of infant death. It may also appear later in life and then have a milder course of the disease. The common feature is progressive weakness of voluntary muscles, with arm, leg and respiratory muscles being affected first. Associated problems may include poor head control, difficulties swallowing, scoliosis, and joint contractures.

<span class="mw-page-title-main">Fazio–Londe disease</span> Medical condition

Fazio–Londe disease (FLD), also called progressive bulbar palsy of childhood, is a very rare inherited motor neuron disease of children and young adults and is characterized by progressive paralysis of muscles innervated by cranial nerves.

Progressive bulbar palsy (PBP) is a medical condition. It belongs to a group of disorders known as motor neuron diseases. PBP is a disease that attacks the nerves supplying the bulbar muscles. These disorders are characterized by the degeneration of motor neurons in the cerebral cortex, spinal cord, brain stem, and pyramidal tracts. This specifically involves the glossopharyngeal nerve (IX), vagus nerve (X), and hypoglossal nerve (XII).

<span class="mw-page-title-main">Dentatorubral–pallidoluysian atrophy</span> Congenital disorder of nervous system

Dentatorubral–pallidoluysian atrophy (DRPLA) is an autosomal dominant spinocerebellar degeneration caused by an expansion of a CAG repeat encoding a polyglutamine tract in the atrophin-1 protein. It is also known as Haw River Syndrome and Naito–Oyanagi disease. Although this condition was perhaps first described by Smith et al. in 1958, and several sporadic cases have been reported from Western countries, this disorder seems to be very rare except in Japan.

<span class="mw-page-title-main">ALS</span> Rare neurodegenerative disease

Amyotrophic lateral sclerosis (ALS), also known as motor neurone disease (MND) or Lou Gehrig's disease, is a rare neurodegenerative disease that results in the progressive loss of motor neurons that control voluntary muscles. ALS is the most common form of the motor neuron diseases. Early symptoms of ALS include stiff muscles, muscle twitches, gradual increasing weakness, and muscle wasting. Limb-onset ALS begins with weakness in the arms or legs, while bulbar-onset ALS begins with difficulty in speaking or swallowing. Around half of people with ALS develop at least mild difficulties with thinking and behavior, and about 15% develop frontotemporal dementia. Motor neuron loss continues until the abilities to eat, speak, move, or, lastly, breathe are lost.

<span class="mw-page-title-main">X-linked spinal muscular atrophy type 2</span> Medical condition

X-linked spinal muscular atrophy type 2, also known as arthrogryposis multiplex congenita X-linked type 1 (AMCX1), is a rare neurological disorder involving death of motor neurons in the anterior horn of spinal cord resulting in generalised muscle wasting (atrophy). The disease is caused by a mutation in UBA1 gene and is passed in an X-linked recessive manner by carrier mothers to affected sons.

<span class="mw-page-title-main">Distal spinal muscular atrophy type 1</span> Medical condition

Distal spinal muscular atrophy type 1 (DSMA1), also known as spinal muscular atrophy with respiratory distress type 1 (SMARD1), is a rare neuromuscular disorder involving death of motor neurons in the spinal cord which leads to a generalised progressive atrophy of body muscles.

<span class="mw-page-title-main">Mild androgen insensitivity syndrome</span> Medical condition

Mild androgen insensitivity syndrome (MAIS) is a condition that results in a mild impairment of the cell's ability to respond to androgens. The degree of impairment is sufficient to impair spermatogenesis and / or the development of secondary sexual characteristics at puberty in males, but does not affect genital differentiation or development. Female genital and sexual development is not significantly affected by the insensitivity to androgens; as such, MAIS is only diagnosed in males. The clinical phenotype associated with MAIS is a normal male habitus with mild spermatogenic defect and / or reduced secondary terminal hair.

<span class="mw-page-title-main">Congenital distal spinal muscular atrophy</span> Hereditary condition characterized by muscle wasting

Congenital distal spinal muscular atrophy is a hereditary condition characterized by muscle wasting (atrophy), particularly of distal muscles in legs and hands, and by early-onset contractures of the hip, knee, and ankle. Affected individuals often have shorter lower limbs relative to the trunk and upper limbs. The condition is a result of a loss of anterior horn cells localized to lumbar and cervical regions of the spinal cord early in infancy, which in turn is caused by a mutation of the TRPV4 gene. The disorder is inherited in an autosomal dominant manner. Arm muscle and function, as well as cardiac and respiratory functions are typically well preserved.

<span class="mw-page-title-main">Epigenetics of neurodegenerative diseases</span> Field of study

Neurodegenerative diseases are a heterogeneous group of complex disorders linked by the degeneration of neurons in either the peripheral nervous system or the central nervous system. Their underlying causes are extremely variable and complicated by various genetic and/or environmental factors. These diseases cause progressive deterioration of the neuron resulting in decreased signal transduction and in some cases even neuronal death. Peripheral nervous system diseases may be further categorized by the type of nerve cell affected by the disorder. Effective treatment of these diseases is often prevented by lack of understanding of the underlying molecular and genetic pathology. Epigenetic therapy is being investigated as a method of correcting the expression levels of misregulated genes in neurodegenerative diseases.

<span class="mw-page-title-main">Spinal muscular atrophy with lower extremity predominance 2A</span> Rare genetic disease

Spinal muscular atrophy with lower extremity predominance 2A (SMALED2A) is a rare neuromuscular disorder characterised by muscle weakness predominantly in legs. The disorder is usually diagnosed shortly after birth; affected children have a delayed motor development, waddling gait, difficulties walking, sometimes develop spasticity. Sensation, swallowing and cognitive development are not affected. The disorder is slowly progressive throughout the lifetime.

References

  1. Arvin, Shelley (2013-04-01). "Analysis of inconsistencies in terminology of spinal and bulbar muscular atrophy and its effect on retrieval of research". Journal of the Medical Library Association. 101 (2): 147–150. doi:10.3163/1536-5050.101.2.010. ISSN   1536-5050. PMC   3634378 . PMID   23646030.
  2. Chen CJ, Fischbeck KH (2006). "Ch. 13: Clinical aspects and the genetic and molecular biology of Kennedy's disease". In Tetsuo Ashizawa, Wells, Robert V. (eds.). Genetic Instabilities and Neurological Diseases (2nd ed.). Boston: Academic Press. pp. 211–222. ISBN   978-0-12-369462-1.
  3. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 Pradat, Pierre-François; Bernard, Emilien; Corcia, Philippe; Couratier, Philippe; Jublanc, Christel; Querin, Giorgia; Morélot Panzini, Capucine; Salachas, François; Vial, Christophe; Wahbi, Karim; Bede, Peter; Desnuelle, Claude (2020-04-10). "The French national protocol for Kennedy's disease (SBMA): consensus diagnostic and management recommendations". Orphanet Journal of Rare Diseases. 15 (1): 90. doi: 10.1186/s13023-020-01366-z . ISSN   1750-1172. PMC   7149864 . PMID   32276665.
  4. 1 2 3 Kennedy, W. R.; Alter, M.; Sung, J. H. (1968). "Progressive proximal spinal and bulbar muscular atrophy of late onset. A sex-linked recessive trait". Neurology. 18 (7): 671–680. doi:10.1212/WNL.18.7.671. PMID   4233749. S2CID   45735233.
  5. 1 2 3 4 5 A, La Spada (1993–2020). "Spinal and Bulbar Muscular Atrophy". GeneReviews [Internet]. PMID   20301508.
  6. Laskaratos, Achilleas; Breza, Marianthi; Karadima, Georgia; Koutsis, Georgios (2020-06-22). "Wide range of reduced penetrance alleles in spinal and bulbar muscular atrophy: a model-based approach". Journal of Medical Genetics. 58 (6): jmedgenet–2020–106963. doi:10.1136/jmedgenet-2020-106963. ISSN   0022-2593. PMID   32571900. S2CID   219991108.
  7. 1 2 Merry, D. E. (2005). "Animal Models of Kennedy Disease". NeuroRx. 2 (3): 471–479. doi:10.1602/neurorx.2.3.471. PMC   1144490 . PMID   16389310.
  8. 1 2 3 4 5 Breza, Marianthi; Koutsis, Georgios (March 2019). "Kennedy's disease (spinal and bulbar muscular atrophy): a clinically oriented review of a rare disease". Journal of Neurology. 266 (3): 565–573. doi:10.1007/s00415-018-8968-7. ISSN   1432-1459. PMID   30006721. S2CID   49722696.
  9. 1 2 3 4 5 6 Lieberman, Andrew P. (2018). "Spinal and bulbar muscular atrophy". Neurogenetics, Part II. Handbook of Clinical Neurology. Vol. 148. pp. 625–632. doi:10.1016/B978-0-444-64076-5.00040-5. ISBN   978-0-444-64076-5. ISSN   0072-9752. PMID   29478604.
  10. 1 2 3 4 5 6 7 Hashizume, Atsushi; Fischbeck, Kenneth H.; Pennuto, Maria; Fratta, Pietro; Katsuno, Masahisa (October 2020). "Disease mechanism, biomarker and therapeutics for spinal and bulbar muscular atrophy (SBMA)". Journal of Neurology, Neurosurgery, and Psychiatry. 91 (10): 1085–1091. doi:10.1136/jnnp-2020-322949. hdl: 11577/3398206 . ISSN   1468-330X. PMID   32934110. S2CID   221667439.
  11. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 Arnold, Frederick J.; Merry, Diane E. (October 2019). "Molecular Mechanisms and Therapeutics for SBMA/Kennedy's Disease". Neurotherapeutics. 16 (4): 928–947. doi:10.1007/s13311-019-00790-9. ISSN   1878-7479. PMC   6985201 . PMID   31686397.
  12. 1 2 Fernández-Rhodes, Lindsay E; Kokkinis, Angela D; White, Michelle J; Watts, Charlotte A; Auh, Sungyoung; Jeffries, Neal O; Shrader, Joseph A; Lehky, Tanya J; Li, Li; Ryder, Jennifer E; Levy, Ellen W; Solomon, Beth I; Harris-Love, Michael O; La Pean, Alison; Schindler, Alice B (2011-02-01). "Efficacy and safety of dutasteride in patients with spinal and bulbar muscular atrophy: a randomised placebo-controlled trial". The Lancet Neurology. 10 (2): 140–147. doi:10.1016/S1474-4422(10)70321-5. ISSN   1474-4422. PMC   3056353 . PMID   21216197.
  13. "Kennedy's Disease Information Page: National Institute of Neurological Disorders and Stroke (NINDS)". NIH. Retrieved 2016-03-23.
  14. Marcato, S.; Kleinbub, J. R.; Querin, G.; Pick, E.; Martinelli, I.; Bertolin, C.; Cipolletta, S.; Pegoraro, E.; Sorarù, G.; Palmieri, A. (2018-09-11). "Unimpaired Neuropsychological Performance and Enhanced Memory Recall in Patients with Sbma: A Large Sample Comparative Study". Scientific Reports. 8 (1): 13627. Bibcode:2018NatSR...813627M. doi:10.1038/s41598-018-32062-5. ISSN   2045-2322. PMC   6134140 . PMID   30206283.
  15. 1 2 Dejager, S. (2002-08-01). "A Comprehensive Endocrine Description of Kennedy's Disease Revealing Androgen Insensitivity Linked to CAG Repeat Length". Journal of Clinical Endocrinology & Metabolism. 87 (8): 3893–3901. doi: 10.1210/jcem.87.8.8780 . ISSN   0021-972X. PMID   12161529.
  16. Nakatsuji, Hideaki; Araki, Amane; Hashizume, Atsushi; Hijikata, Yasuhiro; Yamada, Shinichiro; Inagaki, Tomonori; Suzuki, Keisuke; Banno, Haruhiko; Suga, Noriaki; Okada, Yohei; Ohyama, Manabu; Nakagawa, Tohru; Kishida, Ken; Funahashi, Tohru; Shimomura, Iichiro (2017-05-01). "Correlation of insulin resistance and motor function in spinal and bulbar muscular atrophy". Journal of Neurology. 264 (5): 839–847. doi:10.1007/s00415-017-8405-3. ISSN   1432-1459. PMID   28229243. S2CID   3553270.
  17. Guber, Robert D.; Takyar, Varun; Kokkinis, Angela; Fox, Derrick A.; Alao, Hawwa; Kats, Ilona; Bakar, Dara; Remaley, Alan T.; Hewitt, Stephen M.; Kleiner, David E.; Liu, Chia-Ying; Hadigan, Colleen; Fischbeck, Kenneth H.; Rotman, Yaron; Grunseich, Christopher (2017-12-12). "Nonalcoholic fatty liver disease in spinal and bulbar muscular atrophy". Neurology. 89 (24): 2481–2490. doi:10.1212/WNL.0000000000004748. ISSN   0028-3878. PMC   5729799 . PMID   29142082.
  18. 1 2 3 4 5 6 7 Atsuta, Naoki (2006). "Natural history of spinal and bulbar muscular atrophy (SBMA): a study of 223 Japanese patients". Brain. 129 (6): 1446–1455. doi:10.1093/brain/awl096. PMID   16621916.
  19. Bunting, Emma L.; Hamilton, Joseph; Tabrizi, Sarah J. (2021-09-03). "Polyglutamine diseases". Current Opinion in Neurobiology. 72: 39–47. doi:10.1016/j.conb.2021.07.001. ISSN   1873-6882. PMID   34488036. S2CID   237407161.
  20. Adachi, H.; Waza, M.; Katsuno, M.; Tanaka, F.; Doyu, M.; Sobue, G. (2007-04-01). "Pathogenesis and molecular targeted therapy of spinal and bulbar muscular atrophy". Neuropathology and Applied Neurobiology. 33 (2): 135–151. doi:10.1111/j.1365-2990.2007.00830.x. ISSN   1365-2990. PMID   17359355. S2CID   73301743.
  21. 1 2 La Spada, Albert R.; Roling, Daniel B.; Harding, Anita E.; Warner, Carolyn L.; Spiegel, Roland; Hausmanowa-Petrusewicz, Irena; Yee, Woon-Chee; Fischbeck, Kenneth H. (December 1992). "Meiotic stability and genotype – phenotype correlation of the trinucleotide repeat in X–linked spinal and bulbar muscular atrophy". Nature Genetics. 2 (4): 301–304. doi:10.1038/ng1292-301. ISSN   1061-4036. PMID   1303283. S2CID   6603129.
  22. Fratta, Pietro; Nirmalananthan, Niranjanan; Masset, Luc; Skorupinska, Iwona; Collins, Toby; Cortese, Andrea; Pemble, Sally; Malaspina, Andrea; Fisher, Elizabeth M. C.; Greensmith, Linda; Hanna, Michael G. (2014-06-10). "Correlation of clinical and molecular features in spinal bulbar muscular atrophy". Neurology. 82 (23): 2077–2084. doi:10.1212/WNL.0000000000000507. ISSN   0028-3878. PMC   4075620 . PMID   24814851.
  23. Fan, Hueng-Chuen; Ho, Li-Ing; Chi, Ching-Shiang; Chen, Shyi-Jou; Peng, Giia-Sheun; Chan, Tzu-Min; Lin, Shinn-Zong; Harn, Horng-Jyh (May 2014). "Polyglutamine (PolyQ) Diseases: Genetics to Treatments". Cell Transplantation. 23 (4–5): 441–458. doi: 10.3727/096368914X678454 . ISSN   0963-6897. PMID   24816443. S2CID   27522175.
  24. 1 2 3 4 5 6 Giorgetti, Elisa; Lieberman, Andrew P. (November 2016). "Polyglutamine androgen receptor-mediated neuromuscular disease". Cellular and Molecular Life Sciences. 73 (21): 3991–3999. doi:10.1007/s00018-016-2275-1. ISSN   1420-9071. PMC   5045769 . PMID   27188284.
  25. Lund, A.; Udd, B.; Juvonen, V.; Andersen, P. M.; Cederquist, K.; Davis, M.; Gellera, C.; Kölmel, C.; Ronnevi, L. O.; Sperfeld, A. D.; Sörensen, S. A. (June 2001). "Multiple founder effects in spinal and bulbar muscular atrophy (SBMA, Kennedy disease) around the world". European Journal of Human Genetics. 9 (6): 431–436. doi: 10.1038/sj.ejhg.5200656 . ISSN   1018-4813. PMID   11436124. S2CID   24766290.
  26. Kuhlenbäumer, G.; Kress, W.; Ringelstein, E.B.; Stögbauer, F. (2001-01-12). "Thirty-seven CAG repeats in the androgen receptor gene in two healthy individuals". Journal of Neurology. 248 (1): 23–26. doi:10.1007/s004150170265. ISSN   0340-5354. PMID   11266016. S2CID   6736869.
  27. 1 2 3 Sujkowski, Alyson; Hong, Luke; Wessells, R.J.; Todi, Sokol V. (February 2022). "The protective role of exercise against age-related neurodegeneration". Ageing Research Reviews. 74: 101543. doi:10.1016/j.arr.2021.101543. ISSN   1568-1637. PMC   8761166 . PMID   34923167.
  28. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 Weydt, Patrick; Sagnelli, Anna; Rosenbohm, Angela; Fratta, Pietro; Pradat, Pierre-François; Ludolph, Albert C.; Pareyson, Davide (March 2016). "Clinical Trials in Spinal and Bulbar Muscular Atrophy-Past, Present, and Future". Journal of Molecular Neuroscience. 58 (3): 379–387. doi:10.1007/s12031-015-0682-7. ISSN   1559-1166. PMID   26572537. S2CID   17956032.
  29. 1 2 Dahlqvist, Julia Rebecka; Vissing, John (March 2016). "Exercise Therapy in Spinobulbar Muscular Atrophy and Other Neuromuscular Disorders". Journal of Molecular Neuroscience. 58 (3): 388–393. doi:10.1007/s12031-015-0686-3. ISSN   1559-1166. PMID   26585990. S2CID   17216531.
  30. Heje, Karen; Andersen, Grete; Buch, Astrid; Andersen, Henning; Vissing, John (2019-07-01). "High-intensity training in patients with spinal and bulbar muscular atrophy". Journal of Neurology. 266 (7): 1693–1697. doi:10.1007/s00415-019-09316-x. ISSN   1432-1459. PMID   31004213. S2CID   123973756.
  31. 1 2 Katsuno, Masahisa; Tanaka, Fumiaki; Adachi, Hiroaki; Banno, Haruhiko; Suzuki, Keisuke; Watanabe, Hirohisa; Sobue, Gen (December 2012). "Pathogenesis and therapy of spinal and bulbar muscular atrophy (SBMA)". Progress in Neurobiology. 99 (3): 246–256. doi:10.1016/j.pneurobio.2012.05.007. ISSN   1873-5118. PMID   22609045. S2CID   207406950.
  32. "Kennedy's Disease Association - National Organization for Rare Disorders". rarediseases.org. 2023-01-06. Retrieved 2023-01-24.
  33. "Kennedy's Disease | National Institute of Neurological Disorders and Stroke". www.ninds.nih.gov. Retrieved 2023-01-24.
  34. 1 2 "Who We Are". kennedysdisease.org. Retrieved 2023-01-24.
  35. 1 2 "Research Grants". kennedysdisease.org. Retrieved 2023-01-24.

Further reading