User:Kashmiri/sandbox/Spinal muscular atrophy

From Wikipedia, the free encyclopedia



The following list of suggested headings contains wikilinks; the actual headings should not.

  • Signs and symptoms or Characteristics
  • Causes: Includes Risk factors, triggers, Genetics or genome, Virology (e.g., structure/Morphology, replication).
  • Pathophysiology or Mechanism
  • Diagnosis: Includes characteristic biopsy findings and differential diagnosis.
  • Prevention or Screening (if the section only discusses secondary prevention it should follow the treatment section)
  • Treatment or Management: This might include any type of currently used treatment, such as diet, exercise, medication, palliative care, physical therapy, psychotherapy, self care, surgery, watchful waiting, and many other possibilities. Consider discussing treatments in a plausible order in which they might be tried, or discussing the most common treatments first. Avoid experimental/speculative treatments and preventive measures (e.g., prophylactic vaccines or infection-avoidance techniques). As per the policy of WP:NOTHOW, Wikipedia articles should not be written in a "how-to" style, but this does not not prevent adding official guidelines of treatments or managements if these can be presented in an objective manner and with medically reliable sources.
  • Prognosis
  • Epidemiology: factors such as incidence, prevalence, age distribution, and influence of gender.
  • History: Early discoveries, historical figures, and outdated treatments (not patient history)
  • Society and culture: This might include stigma, economics, religious aspects, awareness, legal issues, notable cases
  • Research directions: Include only if addressed by significant sources. See Trivia, and avoid useless statements like "More research is needed". Wikipedia is not a directory of clinical trials or researchers.
  • In special populations, such as In the elderly or In pregnancy or In children
  • In other animals


Spinal muscular atrophy[edit]

Kashmiri/sandbox/Spinal muscular atrophy

Spinal muscular atrophy (SMA) is an incurable autosomal recessive disease caused by a genetic defect in the SMN1 gene which codes SMN, a protein necessary for survival of motor neurons, and resulting in death of neuronal cells in the anterior horn of spinal cord and subsequent system-wide muscle wasting (atrophy).

Spinal muscular atrophy manifests in various degrees of severity which all have in common general muscle wasting and mobility impairment. Other body systems may be affected as well, particularly in early-onset forms. SMA is the most common genetic cause of infant death.

The term spinal muscular atrophy is sometimes also used with relation to certain very rare hereditary disorders characterised by similar symptoms – see spinal muscular atrophies for a comparison chart.

Classification[edit]

SMA manifests over a wide range of severity affecting infants through adults. The disease spectrum is variously divided into 3–5 types, in accordance either with the age of onset of symptoms or with the highest attained milestone of motor development.

The most commonly used classification is as follows:

Type Eponym Usual age of onset Characteristics OMIM
I: Infantile Werdnig–Hoffmann disease 0–6 months The severe form manifests in the first months of life, usually with a quick and unexpected onset ("floppy baby syndrome"). Rapid motor neuron death causes inefficiency of the major bodily organs - especially of the respiratory system - and pneumonia-induced respiratory failure is the most frequent cause of death. Babies diagnosed with SMA type I do not generally live past two years of age, with death occurring as early as within weeks in the most severe cases (sometimes termed SMA type 0). With proper respiratory support, those with milder SMA type I phenotypes, which account for around 10% of cases, are known to live into adolescence and adulthood. 253300
II: Intermediate Dubowitz disease 6–18 months The intermediate form affects children who are never able to stand and walk but who are able to maintain a sitting position at least some time in their life. The onset of weakness is usually noticed some time between 6 and 18 months. The progress is known to vary greatly, some patients gradually grow weaker over time while others through careful maintenance avoid any progression. Body muscles are weakened, and the respiratory system is a major concern. Life expectancy is somewhat reduced but most SMA II patients live well into adulthood. 253550
III: Juvenile Kugelberg–Welander disease >18 months The juvenile form usually manifests after 18 months of age and describes patients who are able to walk without support at some time, although many later lose this ability. Respiratory involvement is less noticeable, and life expectancy is normal or near normal. 253400
IV: Adult-onset Adulthood The adult-onset form (sometimes classified as a late-onset SMA type III) usually manifests after the third decade of life with gradual weakening of muscles – mainly affects proximal muscles of the extremities – frequently rendering the patient wheelchair-bound. Other complications are rare, and life expectancy is unaffected. 271150

The most severe form of SMA type I is sometimes termed SMA type 0 (or severe infantile SMA) and is diagnosed in babies that are born so weak that are able to survive only a few weeks even with intensive respiratory support. SMA type 0 should not be confused with SMARD1 which may have very similar symptoms and course but has a different genetic cause than SMA.

Development milestone attainment is commonly measured using a specially modified Hammersmith Functional Motor Scale.[1][2][3][4]

The term Werdnig-Hoffman disease is used after Johann Hoffmann and Guido Werdnig and the term Kugelberg-Welander disease after Erik Klas Hendrik Kugelberg and Lisa Welander, the medical professionals who were among the first to describe the condition.[5] Rarely used Dubowitz disease (not to be confused with Dubowitz syndrome) is named after Victor Dubowitz, an English neurologist who authored several studies on the intermediate SMA phenotype.

Causes[edit]

Pathophysiology[edit]

Spinal muscular atrophy has an autosomal recessive pattern of inheritance.

Spinal muscular atrophy is linked to a genetic mutation in the SMN1 gene.[6]

Human chromosome 5 contains two nearly identical genes at location 5q13: a telomeric copy SMN1 and a centromeric copy SMN2. In healthy individuals, the SMN1 gene codes the survival of motor neuron protein (SMN) which, as its name says, plays a crucial role in survival of motor neurons. The SMN2 gene, on the other hand - due to a variation in a single nucleotide (840.C→T) - undergoes alternative splicing at the junction of intron 6 to exon 8, with only 10-20% of SMN2 transcripts coding a fully functional survival of motor neuron protein (SMN-fl) and 80-90% of transcripts resulting in a truncated protein compound (SMNΔ7) which is rapidly degraded in the cell.

In SMA-affected individuals, the SMN1 gene is mutated in such a way that it is unable to correctly code the SMN protein - due to either a deletion occurring at exon 7 or to other point mutations (frequently resulting in the functional conversion of the SMN1 sequence into SMN2). All patients, however, retain at least one copy of the SMN2 gene (with most having 2-4 of them) which still code small amounts of SMN protein - around 10-20% of the normal level - allowing neurons to survive. In the long run, however, reduced availability of the SMN protein results in gradual death of motor neuron cells in the anterior horn of spinal cord and the brain. Consequently, motor muscles undergo progressive atrophy.

Muscles of lower extremities are usually affected first, followed by muscles of upper extremities, spine and neck and, in more severe cases, pulmonary and mastication muscles. Proximal muscles are always affected earlier and in a greater degree than distal.

The severity of SMA symptoms is broadly related to how well the remaining SMN2 genes can make up for the loss of SMN1. This is partly related to the number of SMN2 gene copies present on the chromosome. Whilst healthy individuals carry two SMN2 gene copies, SMA patients can have anything between 1 and 4 (or more) of them, with the greater the number of SMN2 copies the milder the disease severity. Thus, most SMA type I babies have one or two SMN2 copies; SMA II and III patients usually have at least three SMN2 copies; and SMA IV patients normally have at least four of them. However, the correlation between symptom severity and SMN2 copy number is not absolute and there seem to exist other factors impacting on the disease phenotype.[7]

Spinal muscular atrophy is inherited in an autosomal recessive pattern, which means that the defective gene is located on an autosome, and two copies of the defective gene - one from each parent - are required to inherit the disorder: the parents do not need to be themselves affected. SMA seems to appear de novo (i.e., without any hereditary causes) in around 2-4% of cases.

Spinal muscular atrophy affects individuals of all races, unlike other well known autosomal recessive disorders like sickle cell disease and cystic fibrosis which have significant differences in occurrence rate between races. The overall incidence of SMA, of all types and across all ethnic groups, is in the range of 1 per 10,000 individuals; the gene frequency is thus around 1:100, therefore, approximately one in 50 persons are carriers.[8][9] There are no known health consequences of being a carrier, and presently the only way one may know to consider the possibility is if a relative is affected.

Diagnosis[edit]

Prenatal screening is controversial, because of its cost on the one hand, and the severity of the disease on the other hand. Some researchers have concluded that population screening for SMA is not cost-effective, at a cost of $5 million per case averted in USA.[10] Others conclude that SMA meets the criteria for screening programs and relevant testing should be offered to all couples.[11]

Very severe SMA (type 0/I) can be sometimes evident before birth - reduction in fetal movement in the final months of pregnancy; else, it manifests within the first few weeks or months of life when abnormally low muscle tone is observed (the "floppy baby syndrome").

Further, for all SMA types,

Symptoms[edit]

The symptoms vary greatly depending on the SMA type involved, the stage of the disease and individual factors and commonly include:

  • Areflexia, particularly in extremities
  • Overall muscle weakness, poor muscle tone, limpness or a tendency to flop (the "floppy baby" syndrome)
  • Difficulty achieving developmental milestones, difficulty sitting/standing/walking
  • In infants: adopting of a frog-leg position when sitting (hips abducted and knees flexed)
  • Loss of strength of the pulmonary muscles: weak cough, weak cry (infants), accumulation of secretions in the lungs or throat, respiratory distress
  • Bell-shaped torso (caused by using only abdominal muscles for respiration)
  • Clenched fists with sweaty hands
  • Head often tilted to one side, even when lying down
  • Fasciculations (twitching) of the tongue
  • Difficulty sucking or swallowing, poor feeding
  • Arthrogryposis (multiple congenital contractures)
  • Weight lower than normal

Management[edit]

As with the majority of genetic diseases, there is no known cure for spinal muscular atrophy. Management focuses on the following areas of concern:

  • Orthopaedics — Weak spine muscles may lead to development of kyphosis, scoliosis and other orthopaedic problems. Spine fusion is sometimes performed in SMA I/II patients once they reach the age of 8-10 to relieve the pressure of a deformed spine on the lungs. SMA patients might also benefit greatly from various forms of physiotherapy and occupational therapy .
  • Respiratory care — Respiratory system requires utmost attention in SMA as once weakened it never fully recovers. Weakened pulmonary muscles in SMA type I/II patients can make breathing more difficult and pose a risk of hypoxiation, especially in sleep when muscles are more relaxed. Impaired cough reflex poses a constant risk of respiratory infection and pneumonia. Non-invasive ventilation (BiPAP) is frequently used and tracheostomy may be sometimes performed in more severe cases;[13] both methods of ventilation prolong survival in a comparable degree, although tracheostomy prevents speech development.[14]
  • Nutritional care — Difficulties in jaw opening, chewing and swallowing food might pose SMA patients at risk of malnutrition. A feeding tube can be necessary in SMA type I and more severe type II patients.[15][16][17] Additionally, metabolic abnormalities resulting from SMA impair β-oxidation of fatty acids in muscles and can lead to organic acidemia and consequent muscle damage, especially when fasting.[18][19] It is suggested that SMA patients, especially those with more severe forms of disease, reduce intake of fat and avoid prolonged fasting (i.e., eat more frequently than healthy people).[20]
  • MobilityAssistive technologies may help in managing movement and daily activity and greatly increase the quality of life.
  • Cardiology — Although heart is not a matter of routine concern, a link between SMA and certain heart conditions has been suggested.[21][22][23][24]
  • Mental health — SMA children do not differ from the general population in their behaviour; their cognitive development can be slightly faster, and certain aspects of their intelligence are above the average.[25][26][27] Despite their disability, SMA-affected people report high degree of satisfaction from life.[28]

Palliative care in SMA has been standardised in the Consensus Statement for Standard of Care in Spinal Muscular Atrophy which has been recommended for standard adoption worldwide.


Prognosis[edit]

Generally, patients tend to deteriorate over time, but prognosis varies with the SMA type and disease progress which shows a great degree of individual variability. The following description is only indicative.

  • Type 0: Babies of this type do not normally survive beyond the first few months.
  • Type I: The majority of children diagnosed with SMA type I do not reach the age of 10, recurrent respiratory problems being the primary cause of morbidity.[29] With proper care, milder SMA type I cases, which account for around 10% of cases, have lived into adolescence.[30]
  • Type II: The course of the disease is stable or slowly progressing and life expectancy is somewhat reduced compared to the healthy population, although patients usually live to become parents and grandparents.
  • Type III: Patients have normal or nearly normal life expectancy if standards of care are followed.
  • Type IV: This type usually means only mobility impairment and does not affect life expectancy.


Epidemiology[edit]

History[edit]

Research directions[edit]

Since the underlying genetic mechanism of SMA was described in 1990, several therapeutic approaches have been proposed. Since a vast number of in vitro and animal modelling studies suggest that restoration of SMN levels reverts SMA symptoms, the majority of emerging therapies focus on increasing the availability of SMN protein to motor neurons.

The main therapeutic pathways under research as of December 2011 include:[31][32][33][34][35][36][37][38][39]

  • Gene therapy — aims at correcting the SMN1 gene function through inserting specially crafted nucleotide sequences with the help of a viral vector.[40] In the context of SMA, it is currently being researched using the scAAV9 viral vector at the Ohio State University and Nationwide Children's Hospital, USA, and the University of Sheffield, United Kingdom, as well as by Genzyme Corporation, USA, and Généthon, France. In one study this method has resulted in the greatest survival increase achieved to-date in a SMNΔ7 mouse model (median survival of 400 days in treated mice as opposed to 15 days in untreated mice). [citation needed] Safety and pharmacokinetics of scAAV9 viral vector has been tested in non-human primates.[41]
  • Stem cell therapy — aims at offering protection to affected neurons through injection of specially prepared human stem cells in the spinal cord which subsequently develop into neuronal cells able to code full-length SMN protein, and is developed commercially in the context of SMA by California Stem Cell, USA. Experimental stem cell therapy is also offered to SMA patients - based on limited research and with unclear outcome - in private clinics in Brazil, China, Russia and Ukraine.
  • SMN2 activation — aims at increasing expression of the SMN2 gene and thus increasing the amount of full-length SMN available; compounds under investigation include:
  • Butyrates: sodium butyrate and sodium phenylbutyrate — promising in vitro and demonstrated effective in mouse models,[43][44][45] proved ineffective in symptomatic SMA patients (probably due to extremely short half-life),[46] still being trialled in pre-symptomatic type I/II infants[47]
  • Valproic acid — formerly used widely on experimental basis due to earlier research showing its effectiveness in vitro[48] and in mouse models,[49] in achievable concentrations demonstrated ineffective in SMA patients[50][51][52] and even shown to aggravate SMA symptoms[53]
  • M344 — shown very effective in mouse models,[54] so far not trialled in SMA patients
  • CBHA, SBHA — shown very promising in vitro
  • Entinostat (MS-275) — shown very promising in vitro
  • Panobinostat (LBH-589) — shown very effective in mouse models,[55] not trialled in SMA patients due to toxicity at required dosage
  • Trichostatin A — shown effective in mouse models,[56][57] so far not trialled in SMA patients
  • Vorinostat (SAHA) — shown effective in mouse models,[58] so far not trialled in SMA patients
  • Hydroxycarbamide (hydroxyurea) — shown effective in mouse models[59] and subsequently commercially researched by Novo Nordisk, Denmark, but demonstrated no effect on SMA patients in subsequent clinical trials[60]
  • Natural polyphenol compounds: resveratrol, curcumin — moderate effectiveness on muscle strength supported by anecdotal evidence from patients and limited research in vitro[61][62]
  • Prolactin — recently shown effective in mouse models,[63] so far not trialled in SMA patients
  • Salbutamol (albuterol) — demonstrated moderately effective in vitro[64] and in two clinical trials involving SMA II/III patients[65][66]
  • SMN2 alternative splicing modulation — targets the alternative splicing of the SMN2 gene so as to achieve a higher proportion of full-length SMN transcripts (sometimes called "gene conversion SMN2→SMN1"); compounds under investigation include:
  • ISIS-SMNx — a proprietary molecule under development by Isis Pharmaceuticals, USA, and as of July 2012, posed for a phase II clinical trial; has Fast Track Designation in the USA and Orphan Medicinal Product Recommendation in the European Union
  • PTK-SMA1 — a proprietary small molecule splicing modulator of the tetracyclines group under development by Paratek Pharmaceutical, USA
  • RG3039 (formerly, Quinazoline495) — a proprietary quinazoline derivative under development by Repligen Corporation, USA, and as of July 2012, scheduled for phase II clinical trial; has an Orphan Drug Designation and Fast Track Designation in the USA and Orphan Medicinal Product Recommendation in the European Union
  • SMN stabilisation — aims at stabilising the SMNΔ7 protein (the short-lived defective protein coded by the SMN2 gene) so that it is able to sustain neuronal cells;[73] investigated compounds include:
  • TC-007 — a proprietary aminoglycoside antibiotic under commercial development by Tikvah Therapeutics, USA
  • Neuroprotection — aims at prolonging survival of motor neurons even with low levels of SMN; investigated compounds include:
  • An unclear mechanism of action is found in the following compounds currently under research:

In vivo research is usually conducted using genetically engineered Drosophila,[85] zebrafish[86] and mouse[87] models; larger animal models are under development.[88] SMA patients can have a chance of participating in the research by enering their details into international SMA patient registries. A list of clinical trials targeting SMA can be consulted here [1].

It has to be noted, though, that SMA therapeutics seem to be most effective when given immediately after birth, then losing their efficacy with the patient's age. This might be related to the variation in time of the needs for SMN protein by neuronal cells. However, this also poses a major therapeutic problem as hardly ever is SMA diagnosed at birth.[89][90]

External links[edit]

Standards:

Patient registries:

SMA organisations:

SMA patient community support:

See also[edit]

References[edit]

  1. ^ Main, M.; Kairon, H.; Mercuri, E.; Muntoni, F. (2003). "The Hammersmith Functional Motor Scale for Children with Spinal Muscular Atrophy: A Scale to Test Ability and Monitor Progress in Children with Limited Ambulation". European Journal of Paediatric Neurology. 7 (4): 155–159. doi:10.1016/S1090-3798(03)00060-6. PMID 12865054.
  2. ^ Krosschell, K. J.; Maczulski, J. A.; Crawford, T. O.; Scott, C.; Swoboda, K. J. (2006). "A modified Hammersmith functional motor scale for use in multi-center research on spinal muscular atrophy". Neuromuscular Disorders. 16 (7): 417–426. doi:10.1016/j.nmd.2006.03.015. PMC 3260054. PMID 16750368.
  3. ^ O'Hagen, J. M.; Glanzman, A. M.; McDermott, M. P.; Ryan, P. A.; Flickinger, J.; Quigley, J.; Riley, S.; Sanborn, E.; Irvine, C.; Martens, W. B.; Annis, C.; Tawil, R.; Oskoui, M.; Darras, B. T.; Finkel, R. S.; De Vivo, D. C. (2007). "An expanded version of the Hammersmith Functional Motor Scale for SMA II and III patients". Neuromuscular Disorders. 17 (9–10): 693–697. doi:10.1016/j.nmd.2007.05.009. PMID 17658255.
  4. ^ Glanzman, A. M.; O'Hagen, J. M.; McDermott, M. P.; Martens, W. B.; Flickinger, J.; Riley, S.; Quigley, J.; Montes, J.; Dunaway, S.; Deng, L.; Chung, W. K.; Tawil, R.; Darras, B. T.; De Vivo, D. C.; Kaufmann, P.; Finkel, R. S.; Pediatric Neuromuscular Clinical Research Network for Spinal Muscular Atrophy (PNCR) (2011). "Validation of the Expanded Hammersmith Functional Motor Scale in Spinal Muscular Atrophy Type II and III". Journal of Child Neurology. 26 (12): 1499–1507. doi:10.1177/0883073811420294. PMID 21940700.
  5. ^ Dubowitz, V. (2009). "Ramblings in the history of spinal muscular atrophy". Neuromuscular Disorders. 19 (1): 69–73. doi:10.1016/j.nmd.2008.10.004. PMID 18951794.
  6. ^ Brzustowicz, L. M.; Lehner, T.; Castilla, L. H.; Penchaszadeh, G. K.; Wilhelmsen, K. C.; Daniels, R.; Davies, K. E.; Leppert, M.; Ziter, F.; Wood, D.; Dubowitz, V.; Zerres, K.; Hausmanowa-Petrusewicz, I.; Ott, J.; Munsat, T. L.; Gilliam, T. C. (1990). "Genetic mapping of chronic childhood-onset spinal muscular atrophy to chromosome 5q11.2–13.3". Nature. 344 (6266): 540–541. doi:10.1038/344540a0. PMID 2320125.
  7. ^ Jędrzejowska, M.; Milewski, M.; Zimowski, J.; Borkowska, J.; Kostera-Pruszczyk, A.; Sielska, D.; Jurek, M.; Hausmanowa-Petrusewicz, I. (2009). "Phenotype modifiers of spinal muscular atrophy: The number of SMN2 gene copies, deletion in the NAIP gene and probably gender influence the course of the disease". Acta Biochimica Polonica. 56 (1): 103–108. PMID 19287802.
  8. ^ Su, Y. N.; Hung, C. C.; Lin, S. Y.; Chen, F. Y.; Chern, J. P. S.; Tsai, C.; Chang, T. S.; Yang, C. C.; Li, H.; Ho, H. N.; Lee, C. N. (2011). Schrijver, Iris (ed.). "Carrier Screening for Spinal Muscular Atrophy (SMA) in 107,611 Pregnant Women during the Period 2005–2009: A Prospective Population-Based Cohort Study". PLoS ONE. 6 (2): e17067. doi:10.1371/journal.pone.0017067. PMC 3045421. PMID 21364876.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  9. ^ Sugarman, E. A.; Nagan, N.; Zhu, H.; Akmaev, V. R.; Zhou, Z.; Rohlfs, E. M.; Flynn, K.; Hendrickson, B. C.; Scholl, T.; Sirko-Osadsa, D. A.; Allitto, B. A. (2011). "Pan-ethnic carrier screening and prenatal diagnosis for spinal muscular atrophy: Clinical laboratory analysis of >72 400 specimens". European Journal of Human Genetics. 20 (1): 27–32. doi:10.1038/ejhg.2011.134. PMC 3234503. PMID 21811307.
  10. ^ Little, S. E.; Janakiraman, V.; Kaimal, A.; Musci, T.; Ecker, J.; Caughey, A. B. (2010). "The cost-effectiveness of prenatal screening for spinal muscular atrophy". American Journal of Obstetrics and Gynecology. 202 (3): 253.2e1. doi:10.1016/j.ajog.2010.01.032. PMID 20207244.
  11. ^ Prior, T. W.; Professional Practice Guidelines Committee (2008). "Carrier screening for spinal muscular atrophy". Genetics in Medicine. 10 (11): 840–842. doi:10.1097/GIM.0b013e318188d069. PMC 3110347. PMID 18941424.
  12. ^ Rutkove, S. B.; Shefner, J. M.; Gregas, M.; Butler, H.; Caracciolo, J.; Lin, C.; Fogerson, P. M.; Mongiovi, P.; Darras, B. T. (2010). "Characterizing spinal muscular atrophy with electrical impedance myography". Muscle & Nerve. 42 (6): 915. doi:10.1002/mus.21784.
  13. ^ Bach, J. R.; Niranjan, V.; Weaver, B. (2000). "Spinal Muscular Atrophy Type 1: A Noninvasive Respiratory Management Approach". Chest. 117 (4): 1100–1105. doi:10.1378/chest.117.4.1100. PMID 10767247.
  14. ^ Bach, J. R.; Saltstein, K.; Sinquee, D.; Weaver, B.; Komaroff, E. (2007). "Long-Term Survival in Werdnig–Hoffmann Disease". American Journal of Physical Medicine & Rehabilitation. 86 (5): 339. doi:10.1097/PHM.0b013e31804a8505. PMID 17449977.
  15. ^ Messina, S.; Pane, M.; De Rose, P.; Vasta, I.; Sorleti, D.; Aloysius, A.; Sciarra, F.; Mangiola, F.; Kinali, M.; Bertini, E.; Mercuri, E. (2008). "Feeding problems and malnutrition in spinal muscular atrophy type II". Neuromuscular Disorders. 18 (5): 389–393. doi:10.1016/j.nmd.2008.02.008. PMID 18420410.
  16. ^ Chen, Y. S.; Shih, H. H.; Chen, T. H.; Kuo, C. H.; Jong, Y. J. (2011). "Prevalence and Risk Factors for Feeding and Swallowing Difficulties in Spinal Muscular Atrophy Types II and III". The Journal of Pediatrics. doi:10.1016/j.jpeds.2011.08.016.
  17. ^ Tilton, A.; Miller, M.; Khoshoo, V. (1998). "Nutrition and swallowing in pediatric neuromuscular patients". Seminars in Pediatric Neurology. 5 (2): 106–115. doi:10.1016/S1071-9091(98)80026-0. PMID 9661244.
  18. ^ Tein, I.; Sloane, A. E.; Donner, E. J.; Lehotay, D. C.; Millington, D. S.; Kelley, R. I. (1995). "Fatty acid oxidation abnormalities in childhood-onset spinal muscular atrophy: Primary or secondary defect(s)?". Pediatric neurology. 12 (1): 21–30. doi:10.1016/0887-8994(94)00100-G. PMID 7748356.
  19. ^ Crawford, T. O.; Sladky, J. T.; Hurko, O.; Besner-Johnston, A.; Kelley, R. I. (1999). "Abnormal fatty acid metabolism in childhood spinal muscular atrophy". Annals of Neurology. 45 (3): 337–343. doi:10.1002/1531-8249(199903)45:3<337::AID-ANA9>3.0.CO;2-U. PMID 10072048.
  20. ^ Leighton, S. (2003). "Nutrition issues associated with spinal muscular atrophy". Nutrition & Dietetics. 60 (2): 92–96.
  21. ^ Rudnik-Schoneborn, S.; Heller, R.; Berg, C.; Betzler, C.; Grimm, T.; Eggermann, T.; Eggermann, K.; Wirth, R.; Wirth, B.; Zerres, K. (2008). "Congenital heart disease is a feature of severe infantile spinal muscular atrophy". Journal of Medical Genetics. 45 (10): 635–638. doi:10.1136/jmg.2008.057950. PMID 18662980.
  22. ^ Heier, C. R.; Satta, R.; Lutz, C.; Didonato, C. J. (2010). "Arrhythmia and cardiac defects are a feature of spinal muscular atrophy model mice". Human Molecular Genetics. 19 (20): 3906–3918. doi:10.1093/hmg/ddq330. PMC 2947406. PMID 20693262.
  23. ^ Shababi, M.; Habibi, J.; Yang, H. T.; Vale, S. M.; Sewell, W. A.; Lorson, C. L. (2010). "Cardiac defects contribute to the pathology of spinal muscular atrophy models". Human Molecular Genetics. 19 (20): 4059–4071. doi:10.1093/hmg/ddq329. PMID 20696672.
  24. ^ Bevan, A. K.; Hutchinson, K. R.; Foust, K. D.; Braun, L.; McGovern, V. L.; Schmelzer, L.; Ward, J. G.; Petruska, J. C.; Lucchesi, P. A.; Burghes, A. H. M.; Kaspar, B. K. (2010). "Early heart failure in the SMNΔ7 model of spinal muscular atrophy and correction by postnatal scAAV9-SMN delivery". Human Molecular Genetics. 19 (20): 3895–3905. doi:10.1093/hmg/ddq300. PMC 2947399. PMID 20639395.
  25. ^ Von Gontard, A.; Zerres, K.; Backes, M.; Laufersweiler-Plass, C.; Wendland, C.; Melchers, P.; Lehmkuhl, G.; Rudnik-Schöneborn, S. (2002). "Intelligence and cognitive function in children and adolescents with spinal muscular atrophy". Neuromuscular Disorders. 12 (2): 130–136. doi:10.1016/S0960-8966(01)00274-7. PMID 11738354.
  26. ^ Billard, C.; Gillet, P.; Signoret, J. L.; Uicaut, E.; Bertrand, P.; Fardeau, M.; Barthez-Carpentier, M. A.; Santini, J. J. (1992). "Cognitive functions in duchenne muscular dystrophy: A reappraisal and comparison with spinal muscular atrophy". Neuromuscular Disorders. 2 (5–6): 371–378. doi:10.1016/S0960-8966(06)80008-8. PMID 1300185.
  27. ^ Laufersweiler-Plass, C.; Rudnik-Schöneborn, S.; Zerres, K.; Backes, M.; Lehmkuhl, G.; Von Gontard, A. (2002). "Behavioural problems in children and adolescents with spinal muscular atrophy and their siblings". Developmental Medicine & Child Neurology. 45. doi:10.1017/S0012162203000082.
  28. ^ De Oliveira, C. M.; Araújo, A. P. D. Q. C. (2011). "Self-reported quality of life has no correlation with functional status in children and adolescents with spinal muscular atrophy". European Journal of Paediatric Neurology. 15 (1): 36–39. doi:10.1016/j.ejpn.2010.07.003. PMID 20800519.
  29. ^ Yuan, N.; Wang, C. H.; Trela, A.; Albanese, C. T. (2007). "Laparoscopic Nissen Fundoplication During Gastrostomy Tube Placement and Noninvasive Ventilation May Improve Survival in Type I and Severe Type II Spinal Muscular Atrophy". Journal of Child Neurology. 22 (6): 727–731. doi:10.1177/0883073807304009. PMID 17641258.
  30. ^ Bach, J. R. (2007). "Medical Considerations of Long-Term Survival of Werdnig–Hoffmann Disease". American Journal of Physical Medicine & Rehabilitation. 86 (5): 349–55. doi:10.1097/PHM.0b013e31804b1d66. PMID 17449979.
  31. ^ Pruss, R. M.; Giraudon-Paoli, M.; Morozova, S.; Berna, P.; Abitbol, J. L.; Bordet, T. (2010). "Drug discovery and development for spinal muscular atrophy: Lessons from screening approaches and future challenges for clinical development". Future Medicinal Chemistry. 2 (9): 1429–1440. doi:10.4155/FMC.10.228. PMID 21426138.
  32. ^ Sproule, D. M.; Kaufmann, P. (2010). "Therapeutic developments in spinal muscular atrophy". Therapeutic Advances in Neurological Disorders. 3 (3): 173–185. doi:10.1177/1756285610369026. PMC 3002649. PMID 21179609.
  33. ^ Fuller, H. R.; Barišić, M.; Šešo-Šimić, Đ. I.; Špeljko, T.; Morris, G. E.; Šimić, G. (2010). "Treatment strategies for spinal muscular atrophy". Translational Neuroscience. 1 (4): 308. doi:10.2478/v10134-010-0045-4.
  34. ^ Sendtner, M. (2010). "Therapy development in spinal muscular atrophy". Nature Neuroscience. 13 (7): 795–799. doi:10.1038/nn.2565. PMID 20581815.
  35. ^ a b Bosboom, W. M.; Vrancken, A. F. E.; Van Den Berg, L. H.; Wokke, J. H.; Iannaccone, S. T. (2009). Bosboom, Wendy MJ (ed.). "Drug treatment for spinal muscular atrophy type I". The Cochrane Library. doi:10.1002/14651858.CD006281.pub2.
  36. ^ Bosboom, W. M.; Vrancken, A. F. E.; Van Den Berg, L. H.; Wokke, J. H.; Iannaccone, S. T. (2009). Bosboom, Wendy MJ (ed.). "Drug treatment for spinal muscular atrophy types II and III". The Cochrane Library. doi:10.1002/14651858.CD006282.pub2.
  37. ^ Wadman, R. I.; Bosboom, W. M.; Van Den Berg, L. H.; Wokke, J. H.; Iannaccone, S. T.; Vrancken, A. F. E. (2011). Wadman, Renske I (ed.). "Drug treatment for spinal muscular atrophy type I". The Cochrane Library. doi:10.1002/14651858.CD006281.pub3.
  38. ^ Wadman, R. I.; Bosboom, W. M.; Van Den Berg, L. H.; Wokke, J. H.; Iannaccone, S. T.; Vrancken, A. F. E. (2011). Wadman, Renske I (ed.). "Drug treatment for spinal muscular atrophy types II and III". The Cochrane Library. doi:10.1002/14651858.CD006282.pub3.
  39. ^ Lewelt, A.; Newcomb, T. M.; Swoboda, K. J. (2011). "New Therapeutic Approaches to Spinal Muscular Atrophy". Current Neurology and Neuroscience Reports. 12 (1): 42–53. doi:10.1007/s11910-011-0240-9. PMC 3260050. PMID 22134788.
  40. ^ Passini, M. A.; Cheng, S. H. (2011). "Prospects for the gene therapy of spinal muscular atrophy". Trends in Molecular Medicine. 17 (5): 259–265. doi:10.1016/j.molmed.2011.01.002. PMID 21334976.
  41. ^ Bevan, A. K.; Duque, S.; Foust, K. D.; Morales, P. R.; Braun, L.; Schmelzer, L.; Chan, C. M.; McCrate, M.; Chicoine, L. G.; Coley, B. D.; Porensky, P. N.; Kolb, S. J.; Mendell, J. R.; Burghes, A. H.; Kaspar, B. K. (2011). "Systemic Gene Delivery in Large Species for Targeting Spinal Cord, Brain, and Peripheral Tissues for Pediatric Disorders". Molecular Therapy. 19 (11): 1971–1980. doi:10.1038/mt.2011.157. PMC 3222525. PMID 21811247.
  42. ^ Evans, M. C.; Cherry, J. J.; Androphy, E. J. (2011). "Differential regulation of the SMN2 gene by individual HDAC proteins". Biochemical and Biophysical Research Communications. 414 (1): 25–30. doi:10.1016/j.bbrc.2011.09.011. PMID 21925145.
  43. ^ Chang, J. -G.; Hsieh-Li, H. -M.; Jong, Y. -J.; Wang, N. M.; Tsai, C. -H.; Li, H. (2001). "Treatment of spinal muscular atrophy by sodium butyrate". Proceedings of the National Academy of Sciences. 98 (17): 9808. doi:10.1073/pnas.171105098.
  44. ^ Andreassi, C.; Angelozzi, C.; Tiziano, F. D.; Vitali, T.; De Vincenzi, E.; Boninsegna, A.; Villanova, M.; Bertini, E.; Pini, A.; Neri, G.; Brahe, C. (2003). "Phenylbutyrate increases SMN expression in vitro: Relevance for treatment of spinal muscular atrophy". European Journal of Human Genetics. 12 (1): 59–65. doi:10.1038/sj.ejhg.5201102. PMID 14560316.
  45. ^ Brahe, C.; Vitali, T.; Tiziano, F. D.; Angelozzi, C.; Pinto, A. M.; Borgo, F.; Moscato, U.; Bertini, E.; Mercuri, E.; Neri, G. (2004). "Phenylbutyrate increases SMN gene expression in spinal muscular atrophy patients". European Journal of Human Genetics. 13 (2): 256–259. doi:10.1038/sj.ejhg.5201320. PMID 15523494.
  46. ^ Mercuri, E.; Bertini, E.; Messina, S.; Solari, A.; d'Amico, A.; Angelozzi, C.; Battini, R.; Berardinelli, A.; Boffi, P.; Bruno, C.; Cini, C.; Colitto, F.; Kinali, M.; Minetti, C.; Mongini, T.; Morandi, L.; Neri, G.; Orcesi, S.; Pane, M.; Pelliccioni, M.; Pini, A.; Tiziano, F. D.; Villanova, M.; Vita, G.; Brahe, C. (2007). "Randomized, double-blind, placebo-controlled trial of phenylbutyrate in spinal muscular atrophy". Neurology. 68 (1): 51–55. doi:10.1212/01.wnl.0000249142.82285.d6. PMID 17082463.
  47. ^ "Study to Evaluate Sodium Phenylbutyrate in Pre-symptomatic Infants With Spinal Muscular Atrophy (STOPSMA)". Retrieved 28 December 2011.
  48. ^ Brichta, L.; Hofmann, Y.; Hahnen, E.; Siebzehnrubl, F. A.; Raschke, H.; Blumcke, I.; Eyupoglu, I. Y.; Wirth, B. (2003). "Valproic acid increases the SMN2 protein level: A well-known drug as a potential therapy for spinal muscular atrophy". Human Molecular Genetics. 12 (19): 2481–2489. doi:10.1093/hmg/ddg256. PMID 12915451.
  49. ^ Tsai, L. K.; Tsai, M. S.; Ting, C. H.; Li, H. (2008). "Multiple therapeutic effects of valproic acid in spinal muscular atrophy model mice". Journal of Molecular Medicine. 86 (11): 1243–1254. doi:10.1007/s00109-008-0388-1. PMID 18649067.
  50. ^ Swoboda, K. J.; Scott, C. B.; Crawford, T. O.; Simard, L. R.; Reyna, S. P.; Krosschell, K. J.; Acsadi, G.; Elsheik, B.; Schroth, M. K.; d'Anjou, G.; Lasalle, B.; Prior, T. W.; Sorenson, S. L.; MacZulski, J. A.; Bromberg, M. B.; Chan, G. M.; Kissel, J. T.; Project Cure Spinal Muscular Atrophy Investigators Network (2010). Boutron, Isabelle (ed.). "SMA CARNI-VAL Trial Part I: Double-Blind, Randomized, Placebo-Controlled Trial of L-Carnitine and Valproic Acid in Spinal Muscular Atrophy". PLoS ONE. 5 (8): e12140. doi:10.1371/journal.pone.0012140. PMC 2924376. PMID 20808854.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  51. ^ Kissel, J. T.; Scott, C. B.; Reyna, S. P.; Crawford, T. O.; Simard, L. R.; Krosschell, K. J.; Acsadi, G.; Elsheik, B.; Schroth, M. K.; d'Anjou, G.; Lasalle, B.; Prior, T. W.; Sorenson, S.; MacZulski, J. A.; Bromberg, M. B.; Chan, G. M.; Swoboda, K. J.; Project Cure Spinal Muscular Atrophy Investigators' Network (2011). Feany, Mel B. (ed.). "SMA CARNI-VAL TRIAL PART II: A Prospective, Single-Armed Trial of L-Carnitine and Valproic Acid in Ambulatory Children with Spinal Muscular Atrophy". PLoS ONE. 6 (7): e21296. doi:10.1371/journal.pone.0021296. PMC 3130730. PMID 21754985.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  52. ^ Darbar, I. A.; Plaggert, P. G.; Resende, M. B. D.; Zanoteli, E.; Reed, U. C. (2011). "Evaluation of muscle strength and motor abilities in children with type II and III spinal muscle atrophy treated with valproic acid". BMC Neurology. 11: 36. doi:10.1186/1471-2377-11-36. PMC 3078847. PMID 21435220.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  53. ^ Rak, K.; Lechner, B. D.; Schneider, C.; Drexl, H.; Sendtner, M.; Jablonka, S. (2009). "Valproic acid blocks excitability in SMA type I mouse motor neurons". Neurobiology of Disease. 36 (3): 477–487. doi:10.1016/j.nbd.2009.08.014. PMID 19733665.
  54. ^ Riessland, M.; Brichta, L.; Hahnen, E.; Wirth, B. (2006). "The benzamide M344, a novel histone deacetylase inhibitor, significantly increases SMN2 RNA/protein levels in spinal muscular atrophy cells". Human Genetics. 120 (1): 101–110. doi:10.1007/s00439-006-0186-1. PMID 16724231.
  55. ^ Garbes, L.; Riessland, M.; Hölker, I.; Heller, R.; Hauke, J.; Tränkle, C.; Coras, R.; Blümcke, I.; Hahnen, E.; Wirth, B. (2009). "LBH589 induces up to 10-fold SMN protein levels by several independent mechanisms and is effective even in cells from SMA patients non-responsive to valproate". Human Molecular Genetics. 18 (19): 3645–3658. doi:10.1093/hmg/ddp313. PMID 19584083.
  56. ^ Narver, H. L.; Kong, L.; Burnett, B. G.; Choe, D. W.; Bosch-Marcé, M.; Taye, A. A.; Eckhaus, M. A.; Sumner, C. J. (2008). "Sustained improvement of spinal muscular atrophy mice treated with trichostatin a plus nutrition". Annals of Neurology. 64 (4): 465–470. doi:10.1002/ana.21449. PMID 18661558.
  57. ^ Avila, A. M.; Burnett, B. G.; Taye, A. A.; Gabanella, F.; Knight, M. A.; Hartenstein, P.; Cizman, Z.; Di Prospero, N. A.; Pellizzoni, L.; Fischbeck, K. H.; Sumner, C. J. (2007). "Trichostatin a increases SMN expression and survival in a mouse model of spinal muscular atrophy". Journal of Clinical Investigation. 117 (3): 659–671. doi:10.1172/JCI29562. PMC 1797603. PMID 17318264.
  58. ^ Riessland, M.; Ackermann, B.; Förster, A.; Jakubik, M.; Hauke, J.; Garbes, L.; Fritzsche, I.; Mende, Y.; Blumcke, I.; Hahnen, E.; Wirth, B. (2010). "SAHA ameliorates the SMA phenotype in two mouse models for spinal muscular atrophy". Human Molecular Genetics. 19 (8): 1492–1506. doi:10.1093/hmg/ddq023. PMID 20097677.
  59. ^ Grzeschik, S. M.; Ganta, M.; Prior, T. W.; Heavlin, W. D.; Wang, C. H. (2010). "Hydroxyurea enhances SMN2 gene expression in spinal muscular atrophy cells". Annals of Neurology. 58 (2): 194–202. doi:10.1002/ana.20548. PMID 16049920.
  60. ^ Chen, T. - H.; Chang, J. - G.; Yang, Y. - H.; Mai, H. - H.; Liang, W. - C.; Wu, Y. - C.; Wang, H. - Y.; Huang, Y. - B.; Wu, S. - M.; Chen, Y. - C.; Yang, S. - N.; Jong, Y. - J. (2010). "Randomized, double-blind, placebo-controlled trial of hydroxyurea in spinal muscular atrophy". Neurology. 75 (24): 2190–2197. doi:10.1212/WNL.0b013e3182020332. PMID 21172842.
  61. ^ Sakla, M. S.; Lorson, C. L. (2007). "Induction of full-length survival motor neuron by polyphenol botanical compounds". Human Genetics. 122 (6): 635–643. doi:10.1007/s00439-007-0441-0. PMID 17962980.
  62. ^ Dayangaç-Erden, D.; Bora, G.; Ayhan, P.; Kocaefe, Ç.; Dalkara, S.; Yelekçi, K.; Demir, A. S.; Erdem-Yurter, H. (2009). "Histone Deacetylase Inhibition Activity and Molecular Docking of (E )-Resveratrol: Its Therapeutic Potential in Spinal Muscular Atrophy". Chemical Biology & Drug Design. 73 (3): 355. doi:10.1111/j.1747-0285.2009.00781.x. {{cite journal}}: hair space character in |title= at position 68 (help)
  63. ^ Farooq, F.; Molina, F. A. A.; Hadwen, J.; MacKenzie, D.; Witherspoon, L.; Osmond, M.; Holcik, M.; MacKenzie, A. (2011). "Prolactin increases SMN expression and survival in a mouse model of severe spinal muscular atrophy via the STAT5 pathway". Journal of Clinical Investigation. 121 (8): 3042–3050. doi:10.1172/JCI46276. PMC 3148738. PMID 21785216.
  64. ^ Angelozzi, C.; Borgo, F.; Tiziano, F. D.; Martella, A.; Neri, G.; Brahe, C. (2007). "Salbutamol increases SMN mRNA and protein levels in spinal muscular atrophy cells". Journal of Medical Genetics. 45 (1): 29–31. doi:10.1136/jmg.2007.051177. PMID 17932121.
  65. ^ Pane, M.; Staccioli, S.; Messina, S.; d'Amico, A.; Pelliccioni, M.; Mazzone, E. S.; Cuttini, M.; Alfieri, P.; Battini, R.; Main, M.; Muntoni, F.; Bertini, E.; Villanova, M.; Mercuri, E. (2008). "Daily salbutamol in young patients with SMA type II". Neuromuscular Disorders. 18 (7): 536–540. doi:10.1016/j.nmd.2008.05.004. PMID 18579379.
  66. ^ Tiziano, F. D.; Lomastro, R.; Pinto, A. M.; Messina, S.; d'Amico, A.; Fiori, S.; Angelozzi, C.; Pane, M.; Mercuri, E.; Bertini, E.; Neri, G.; Brahe, C. (2010). "Salbutamol increases survival motor neuron (SMN) transcript levels in leucocytes of spinal muscular atrophy (SMA) patients: Relevance for clinical trial design". Journal of Medical Genetics. 47 (12): 856–858. doi:10.1136/jmg.2010.080366. PMID 20837492.
  67. ^ Andreassi, C.; Jarecki, J.; Zhou, J.; Coovert, D. D.; Monani, U. R.; Chen, X.; Whitney, M.; Pollok, B.; Zhang, M.; Androphy, E.; Burghes, A. H. (2001). "Aclarubicin treatment restores SMN levels to cells derived from type I spinal muscular atrophy patients". Human Molecular Genetics. 10 (24): 2841–2849. doi:10.1093/hmg/10.24.2841. PMID 11734549.
  68. ^ Dimatteo, D.; Callahan, S.; Kmiec, E. B. (2008). "Genetic conversion of an SMN2 gene to SMN1: A novel approach to the treatment of spinal muscular atrophy". Experimental Cell Research. 314 (4): 878–886. doi:10.1016/j.yexcr.2007.10.012. PMID 18078930.
  69. ^ Burghes, A. H. M.; McGovern, V. L. (2010). "Antisense oligonucleotides and spinal muscular atrophy: Skipping along". Genes & Development. 24 (15): 1574. doi:10.1101/gad.1961710.
  70. ^ Passini, M. A.; Bu, J.; Richards, A. M.; Kinnecom, C.; Sardi, S. P.; Stanek, L. M.; Hua, Y.; Rigo, F.; Matson, J.; Hung, G.; Kaye, E. M.; Shihabuddin, L. S.; Krainer, A. R.; Bennett, C. F.; Cheng, S. H. (2011). "Antisense Oligonucleotides Delivered to the Mouse CNS Ameliorate Symptoms of Severe Spinal Muscular Atrophy". Science Translational Medicine. 3 (72): 72ra18. doi:10.1126/scitranslmed.3001777. PMC 3140425. PMID 21368223.
  71. ^ Butchbach, M. E. R.; Singh, J.; Thorsteinsdóttir, M.; Saieva, L.; Slominski, E.; Thurmond, J.; Andrésson, T.; Zhang, J.; Edwards, J. D.; Simard, L. R.; Pellizzoni, L.; Jarecki, J.; Burghes, A. H. M.; Gurney, M. E. (2009). "Effects of 2,4-diaminoquinazoline derivatives on SMN expression and phenotype in a mouse model for spinal muscular atrophy". Human Molecular Genetics. 19 (3): 454–467. doi:10.1093/hmg/ddp510. PMC 2798721. PMID 19897588.
  72. ^ Zhang, M. L.; Lorson, C. L.; Androphy, E. J.; Zhou, J. (2001). "An in vivo reporter system for measuring increased inclusion of exon 7 in SMN2 mRNA: Potential therapy of SMA". Gene Therapy. 8 (20): 1532–1538. doi:10.1038/sj.gt.3301550. PMID 11704813.
  73. ^ Burnett, B. G.; Munoz, E.; Tandon, A.; Kwon, D. Y.; Sumner, C. J.; Fischbeck, K. H. (2008). "Regulation of SMN Protein Stability". Molecular and Cellular Biology. 29 (5): 1107–1115. doi:10.1128/MCB.01262-08. PMC 2643817. PMID 19103745.
  74. ^ Mattis, V. B.; Rai, R.; Wang, J.; Chang, C. W. T.; Coady, T.; Lorson, C. L. (2006). "Novel aminoglycosides increase SMN levels in spinal muscular atrophy fibroblasts". Human Genetics. 120 (4): 589–601. doi:10.1007/s00439-006-0245-7. PMID 16951947.
  75. ^ Mattis, V. B.; Fosso, M. Y.; Chang, C. W.; Lorson, C. L. (2009). "Subcutaneous administration of TC007 reduces disease severity in an animal model of SMA". BMC Neuroscience. 10: 142. doi:10.1186/1471-2202-10-142. PMC 2789732. PMID 19948047.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  76. ^ Lunn, M. R.; Root, D. E.; Martino, A. M.; Flaherty, S. P.; Kelley, B. P.; Coovert, D. D.; Burghes, A. H.; Thi Man, N.; Morris, G. E.; Zhou, J.; Androphy, E. J.; Sumner, C. J.; Stockwell, B. R. (2004). "Indoprofen Upregulates the Survival Motor Neuron Protein through a Cyclooxygenase-Independent Mechanism". Chemistry & Biology. 11 (11): 1489–1493. doi:10.1016/j.chembiol.2004.08.024. PMC 3160629. PMID 15555999.
  77. ^ Nizzardo, M.; Nardini, M.; Ronchi, D.; Salani, S.; Donadoni, C.; Fortunato, F.; Colciago, G.; Falcone, M.; Simone, C.; Riboldi, G.; Govoni, A.; Bresolin, N.; Comi, G. P.; Corti, S. (2011). "Beta-lactam antibiotic offers neuroprotection in a spinal muscular atrophy model by multiple mechanisms". Experimental Neurology. 229 (2): 214–225. doi:10.1016/j.expneurol.2011.01.017. PMID 21295027.
  78. ^ Hedlund, E. (2011). "The protective effects of beta-lactam antibiotics in motor neuron disorders". Experimental Neurology. 231 (1): 14–18. doi:10.1016/j.expneurol.2011.06.002. PMID 21693120.
  79. ^ Rose, F. F.; Mattis, V. B.; Rindt, H.; Lorson, C. L. (2009). "Delivery of recombinant follistatin lessens disease severity in a mouse model of spinal muscular atrophy". Human Molecular Genetics. 18 (6): 997–1005. doi:10.1093/hmg/ddn426. PMC 2649020. PMID 19074460.
  80. ^ Takeuchi, Y.; Miyanomae, Y.; Komatsu, H.; Oomizono, Y.; Nishimura, A.; Okano, S.; Nishiki, T.; Sawada, T. (1994). "Efficacy of Thyrotropin-Releasing Hormone in the Treatment of Spinal Muscular Atrophy". Journal of Child Neurology. 9 (3): 287–289. doi:10.1177/088307389400900313. PMID 7930408.
  81. ^ Tzeng, A. C.; Cheng, J.; Fryczynski, H.; Niranjan, V.; Stitik, T.; Sial, A.; Takeuchi, Y.; Foye, P.; Deprince, M.; Bach, J. R. (2000). "A study of thyrotropin-releasing hormone for the treatment of spinal muscular atrophy: A preliminary report". American journal of physical medicine & rehabilitation / Association of Academic Physiatrists. 79 (5): 435–440. doi:10.1097/00002060-200009000-00005. PMID 10994885.
  82. ^ Kato, Z.; Okuda, M.; Okumura, Y.; Arai, T.; Teramoto, T.; Nishimura, M.; Kaneko, H.; Kondo, N. (2009). "Oral Administration of the Thyrotropin-Releasing Hormone (TRH) Analogue, Taltireline Hydrate, in Spinal Muscular Atrophy". Journal of Child Neurology. 24 (8): 1010–1012. doi:10.1177/0883073809333535. PMID 19666885.
  83. ^ "Roche signs agreement with PTC Therapeutics to advance treatment for Spinal Muscular Atrophy (SMA)". Retrieved 28 December 2011.
  84. ^ "Rethropin: drug pipeline". Retrieved 08 May 2012. {{cite web}}: Check date values in: |accessdate= (help)
  85. ^ Chang, H. C. H.; Dimlich, D. N.; Yokokura, T.; Mukherjee, A.; Kankel, M. W.; Sen, A.; Sridhar, V.; Fulga, T. A.; Hart, A. C.; Van Vactor, D.; Artavanis-Tsakonas, S. (2008). Lewin, Alfred (ed.). "Modeling Spinal Muscular Atrophy in Drosophila". PLoS ONE. 3 (9): e3209. doi:10.1371/journal.pone.0003209. PMC 2527655. PMID 18791638.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  86. ^ Beattie, C. E.; Carrel, T. L.; McWhorter, M. L. (2007). "Fishing for a Mechanism: Using Zebrafish to Understand Spinal Muscular Atrophy". Journal of Child Neurology. 22 (8): 995–1003. doi:10.1177/0883073807305671. PMID 17761655.
  87. ^ Sleigh, J. N.; Gillingwater, T. H.; Talbot, K. (2011). "The contribution of mouse models to understanding the pathogenesis of spinal muscular atrophy". Disease Models & Mechanisms. 4 (4): 457–467. doi:10.1242/dmm.007245. PMC 3124050. PMID 21708901.
  88. ^ "The GSF and FightSMA Announce 100K Research Award for the Development of a Large Animal Model of Spinal Muscular Atrophy". Retrieved 18 December 2011.
  89. ^ Le, T. T.; McGovern, V. L.; Alwine, I. E.; Wang, X.; Massoni-Laporte, A.; Rich, M. M.; Burghes, A. H. M. (2011). "Temporal requirement for high SMN expression in SMA mice". Human Molecular Genetics. 20 (18): 3578–3591. doi:10.1093/hmg/ddr275. PMC 3159555. PMID 21672919.
  90. ^ Porensky, P. N.; Mitrpant, C.; McGovern, V. L.; Bevan, A. K.; Foust, K. D.; Kaspar, B. K.; Wilton, S. D.; Burghes, A. H. M. (2011). "A single administration of morpholino antisense oligomer rescues spinal muscular atrophy in the mouse". Human Molecular Genetics. 21 (7): 1625–38. doi:10.1093/hmg/ddr600. PMID 22186025.