fbpx
Wikipedia

Rett syndrome

Rett syndrome (RTT) is a genetic disorder that typically becomes apparent after 6–18 months of age and almost exclusively in females.[3] Symptoms include impairments in language and coordination, and repetitive movements.[3] Those affected often have slower growth, difficulty walking, and a smaller head size.[3][4] Complications of Rett syndrome can include seizures, scoliosis, and sleeping problems.[3] The severity of the condition is variable.[4]

Rett syndrome
Other namesCerebroatrophic hyperammonemia (obsolete),[1] dementia, ataxia, and loss of purposeful hand use syndrome[2]
A girl with Rett Syndrome smiling at the camera
SpecialtyPsychiatry, pediatrics, neurology
SymptomsImpairments in language and coordination, and repetitive movements, slower growth, smaller head[3]
ComplicationsSeizures, scoliosis, sleeping problems[3]
Usual onsetAfter 6–18 months of age[3]
DurationLifelong[4]
CausesMutation in the MECP2 gene[3]
Diagnostic methodBased on symptoms, genetic testing[4]
Differential diagnosisAngelman syndrome, autism, cerebral palsy, Childhood disintegrative disorder, various neurodegenerative disorders[5]
TreatmentSpecial education, physiotherapy, braces[4]
MedicationAnticonvulsants[4]
PrognosisLife expectancy for many is middle age.[4]
Frequency1 in 8,500 females[3]
Lethal in males, with rare exceptions.

Rett syndrome is due to a genetic mutation in the MECP2 gene,[3] on the X chromosome.[4] It almost always occurs as a new mutation, with less than one percent of cases being inherited.[3][4] It occurs almost exclusively in girls;[3] boys who have a similar mutation typically die shortly after birth.[4] Diagnosis is based on the symptoms and can be confirmed with genetic testing.[4]

There is no known cure for Rett syndrome.[4] Treatment is directed at improving symptoms.[4] Anticonvulsants may be used to help with seizures.[4] Special education, physiotherapy, and leg braces may also be useful depending on the needs of the child.[4] Many of those with the condition live into middle age.[4]

The condition affects about 1 in 8,500 females.[3] In 1999, Lebanese-American physician Huda Zoghbi discovered the mutation that causes the condition.[6][7]

Signs and symptoms edit

Stage I edit

Stage I, called early-onset, typically begins between 6 and 18 months of age.[4] This stage is often overlooked because symptoms of the disorder may be somewhat vague, and parents and doctors may not notice the subtle slowing of development at first.[4] The infant may begin to show less eye contact and have reduced interest in toys. There may be delays in gross motor skills such as sitting or crawling.[4] Hand-wringing and decreasing head growth may occur, but not enough to draw attention. This stage usually lasts for a few months but can continue for more than a year.[4]

Stage II edit

Stage II, or the rapid destructive stage, usually begins between ages 1 and 4 and may last for weeks or months.[4] Its onset may be rapid or gradual as the child loses purposeful hand skills and spoken language.[4] Characteristic hand movements such as wringing, washing, clapping, or tapping, as well as repeatedly moving the hands to the mouth often begin during this stage which is called mouthing.[4] The child may hold the hands clasped behind the back or held at the sides, with random touching, grasping, and releasing.[4] The movements continue while the child is awake but disappear during sleep.[4] Breathing irregularities such as episodes of apnea and hyperventilation may occur, although breathing usually improves during sleep.[4] Some girls also display autistic-like symptoms such as loss of social interaction and communication.[4] Walking may be unsteady and initiating motor movements can be difficult. Slowed head growth is usually noticed during this stage.[4]

Stage III edit

Stage III, or the plateau or pseudo-stationary stage, usually begins between ages 2 and 10 and can last for years.[4] Apraxia, motor problems, and seizures are prominent during this stage.[4] However, there may be improvement in behavior, with less irritability, crying, and autistic-like features.[4] In stage III there may be more interest in the surroundings and alertness, attention span, and communication skills may improve.[4] Many girls remain in this stage for most of their lives.[4]

Stage IV edit

Stage IV, or the late motor deterioration stage, can last for years or decades.[4] Prominent features include reduced mobility, curvature of the spine, and muscle weakness, rigidity, spasticity, and increased muscle tone with abnormal posturing of an arm or leg.[4] Girls who were previously able to walk may stop walking.[4] Cognition, communication, or hand skills generally do not decline in stage IV.[4] Repetitive hand movements may decrease and eye gaze usually improves.[4]

Variants edit

The signs and symptoms of the typical form of the Rett syndrome are well described. In addition to the classical form of Rett syndrome, several atypical forms have been described over the years;[8] the main groups are:

  • Congenital variant (Rolando variant): in this severe subtype of Rett syndrome, the development of the patients and their head circumference are abnormal from birth.[9] The typical gaze of Rett syndrome patients is usually absent;
  • Zappella variant of Rett Syndrome or preserved speech variant: in this subtype of Rett syndrome the patients acquire some manual skills and language is partially recovered around the age of 5 years (that is after the regression phase). Height, weight and head circumference are often in the normal range, and a good gross motor function can be observed.[10][11][12][13][14][15] The Zappella variant is a milder form of Rett syndrome;
  • Hanefeld variant or early epilepsy variant. In this form of Rett syndrome, the patients have epilepsy before 5 months of age.[16]

The definition itself of the Rett syndrome has been refined over the years: as the atypical forms subsist near to the classical form (Hagberg & Gillberg, 1993), the "Rett Complex" terminology has been introduced.[17][18]

Cause edit

Genetically, Rett syndrome (RTT) is caused by mutations in the gene MECP2 located on the X chromosome (which is involved in transcriptional silencing and epigenetic regulation of methylated DNA), and can arise sporadically or from germline mutations. In less than 10% of RTT cases, mutations in the genes CDKL5 or FOXG1 have also been found to resemble it.[medical citation needed] Rett syndrome is initially diagnosed by clinical observation, but the diagnosis is definitive when there is a genetic defect in the MECP2 gene.

It has been argued that Rett syndrome is in fact a neurodevelopmental condition as opposed to a neurodegenerative condition. One piece of evidence for this is that mice with induced Rett Syndrome show no neuronal death, and some studies have suggested that their phenotypes can be partially rescued by adding functional MECP2 gene back when they are adults. This information has also helped lead to further studies aiming to treat the disorder.[19]

Sporadic mutations edit

In at least 95% of Rett syndrome cases, the cause is a de novo mutation in the child. That is, it is not inherited from either parent. Parents are generally genotypically normal, without a MECP2 mutation.[citation needed]

In cases of the sporadic form of RTT, the mutated MECP2 is thought to derive almost exclusively from a de novo mutation on the male copy of the X chromosome.[20] It is not yet known what causes the sperm to mutate, and such mutations are rare.

Germline mutations edit

It can also be inherited from phenotypically normal mothers who have a germline mutation in the gene encoding methyl-CpG-binding protein-2, MeCP2.[21] In these cases, inheritance follows an X-linked dominant pattern and is seen almost exclusively in females, as most males die in utero or shortly after birth.[22] MECP2 is found near the end of the long arm of the X chromosome at Xq28. An atypical form of RTT, characterized by infantile spasms or early onset epilepsy, can also be caused by a mutation to the gene encoding cyclin-dependent kinase-like 5 (CDKL5). As stated by Aine Merwick, Margaret O'Brien, and Norman Delanty in an article on gene disorders titled Complex single gene disorders and epilepsy, "Rett syndrome affects one in every 12,500 female live births by age 12 years."[23]

Mechanism edit

 
The location of the gene responsible for Rett syndrome

Pontine noradrenergic deficits edit

Brain levels of norepinephrine are lower in people with Rett syndrome[24] (reviewed in[25]). The genetic loss of MECP2 changes the properties of cells in the locus coeruleus, the exclusive source of noradrenergic innervation to the cerebral cortex and hippocampus.[26][27] These changes include hyperexcitability and decreased functioning of its noradrenergic innervation.[28] Moreover, a reduction of the tyrosine hydroxylase (Th) mRNA level, the rate-limiting enzyme in catecholamine synthesis, was detected in the whole pons of MECP2-null male as well as in adult heterozygous (MECP2+/-) female mice.[29] Using immunoquantitative techniques, a decrease of Th protein staining level, number of locus coeruleus Th-expressing neurons and density of dendritic arborization surrounding the structure was shown in symptomatic MeCP2-deficient mice.[29] However, locus coeruleus cells are not dying, but are more likely losing their fully mature phenotype, since no apoptotic neurons in the pons were detected.[29]

Researchers have concluded that "Because these neurons are a pivotal source of norepinephrine throughout the brainstem and forebrain and are involved in the regulation of diverse functions disrupted in Rett syndrome, such as respiration and cognition, we hypothesize that the locus coeruleus is a critical site at which loss of MECP2 results in CNS dysfunction." The restoration of normal locus coeruleus function may therefore be of potential therapeutic value in the treatment of Rett syndrome.[28][29]

Midbrain dopaminergic disturbances edit

The majority of dopamine in the mammalian brain is synthesized by nuclei located in the mesencephalon. The substantia nigra pars compacta (SNpc), the ventral tegmental area (VTA) and the retrorubral field (RRF) contain dopaminergic neurons expressing tyrosine hydroxylase (Th, i.e. the rate-limiting enzyme in catecholamine synthesis).[30][31][32]

The nigro-striatal pathway originates from the SNpc; its principal rostral target is the caudate-putamen (CPu), which it irradiates through the median forebrain bundle (MFB). This connection is involved in the tight modulation of motor strategies computed by a cortico-basal ganglia-thalamo-cortical loop.[33]

Indeed, based on the canonical anatomofunctional model of basal ganglia, nigrostriatal dopamine is able to modulate the motor loop by acting on dopaminergic receptors located on striatal GABAergic medium spiny neurons.[34]

Dysregulation of the nigrostriatal pathway is causative from Parkinson disease (PD) in humans.[35] Toxic and/or genetic ablation of SNpc neurons produces experimental parkinsonism in mice and primates.[36] The common features of PD and PD animal models are motor impairments[37] (hypotonia, bradykinesia, hypokinesia).

RTT pathology, in some aspects, overlaps the motor phenotype observed in PD patients.[38][39][40] Several neuropathological studies on postmortem brain samples argued for an SNpc alteration, evidenced by neuromelanin hypopigmentation, reduction in the structure area, and even, controversially, signs of apoptosis. In parallel, a hypometabolism was underlined by a reduction of several catecholamines (dopamine, noradrenaline, adrenaline) and their principal metabolic by-products.[25] Mouse models of RTT are available; the most studied are constitutively deleted Mecp2 mice developed by Adrian Bird or Katelyn McCormick laboratories.[41][42][43][44]

In accordance with the motor spectrum of the RTT phenotype, Mecp2-null mice show motor abnormalities from postnatal day 30 that worsen until death. These models offer a crucial substrate to elucidate the molecular and neuroanatomical correlates of MeCP2-deficiency.[45] Recently (2008), it was shown that the conditional deletion of Mecp2 in catecholaminergic neurons (by crossing of Th-Cre mice with loxP-flanked Mecp2 ones) recapitulates a motor symptomatology; it was further documented that brain levels of Th in mice lacking MeCP2 in catecholaminergic neurons only are reduced, participating to the motor phenotype.[46]

However, the most studied model for the evaluation of therapeutics is the Mecp2-null mouse (totally devoid of MeCP2). In this context, a reduction in the number and soma size of Th-expressing neurons is present from 5 weeks of age and is accompanied by a decrease of Th immunoreactivity in the caudate-putamen, the principal target of dopaminergic neurons arising from the SNpc.[47] Moreover, a neurochemical analysis of dopaminergic contents in microdissected midbrain and striatal areas revealed a reduction of dopamine at five and nine weeks of age. It is noteworthy that later on (at nine weeks), the morphological parameters remain altered but not worsened, whereas the phenotype progresses and behavioral deficits are more severe. The amount of fully activated Th (Serine40-phosphorylated isoform) in neurons that remain in the SNpc is mildly affected at 5 weeks but severely impaired by 9 weeks.[47] Finally, using a chronic and oral L-Dopa treatment on MeCP2-deficient mice, authors reported an amelioration of some of the motor deficits previously identified.[47] Altogether, these results argue for an alteration of the nigrostriatal dopaminergic pathway in MeCP2-deficient animals as a contributor of the neuromotor deficits.[47]

There is an association of Rett syndrome with brain-derived neurotrophic factor (BDNF).[48]

Molecular functions of MECP2 in Rett syndrome pathology edit

As reviewed by Sharifi and Yasui,[49] MECP2 protein, encoded by the MECP2 gene binds to DNA with a high affinity for CpG methylated DNA sites and affects transcription. MECP2 can bind to 5mc (5-methylcytosine) and 5hmc (5-hydroxymethylcytosine) with similar affinity, and these dinucleotides account for the majority of MECP2 binding sites in the mammalian genome. MECP2 is involved in higher order chromatin organization and appears necessary for compacting chromosomes. MECP2 binding to DNA influences mRNA splicing events. MECP2 also appears to function in DNA repair processes. MECP2-/+ deficient female mice have elevated rates of cell death when exposed to DNA damaging agents and are prone to early senescence.[49]

Interactive pathway map edit

An interactive pathway map of Rett syndrome has been published.[50]

Diagnosis edit

 
A girl with Rett Syndrome mouthing her hands, a common behavior with Rett Syndrome

Prior to the discovery of a genetic cause, Rett syndrome had been designated as a pervasive developmental disorder by the Diagnostic and Statistical Manual of Mental Disorders (DSM), together with the autism spectrum disorders. Some argued against this conclusive assignment because RTT resembles non-autistic disorders such as fragile X syndrome, tuberous sclerosis, or Down syndrome that also exhibit autistic features.[51] After research proved the molecular mechanism, in 2013 the DSM-5 removed the syndrome altogether from classification as a mental disorder.[52]

Rett syndrome diagnosis involves close observation of the child's growth and development to observe any abnormalities in regards to developmental milestones.[53] A diagnosis is considered when decreased head growth is observed. Conditions with similar symptoms must first be ruled out.[53]

There are certain criteria that must be met for the diagnosis. A blood test can rule in or rule out the presence of the MECP2 mutation, however, this mutation is present in other conditions as well.[54]

For a classic diagnosis, all four criteria for ruling in a diagnosis must be met, as well as the two criteria for ruling out a diagnosis. Supportive criteria may also be present, but are not required for diagnosis. For an atypical or variant diagnosis, at least two of the four criteria for ruling in the diagnosis must be met, as well as five of the eleven supportive criteria. A period of symptom regression followed by recovery or symptom stabilization must also occur.[54] Children are often misdiagnosed as having autism, cerebral palsy, or another form of developmental delay. A positive test for the MECP2 mutation is not enough to make a diagnosis.[54]

Ruling in[54]

  • Decreased or loss of use of fine motor skills
  • Decreased or loss of verbal speech
  • Abnormalities during gait
  • Repetitive hand movements such as wringing/squeezing or clapping/tapping

Ruling out[54]

  • Traumatic or anoxic/hypoxic brain injury, neurometabolic disease, or severe infection that may better explain symptoms
  • Abnormal psychomotor development during the first six months of life

Supportive criteria[54]

  • Breathing disturbances when awake
  • Bruxism while awake
  • Impaired sleep pattern
  • Abnormal muscle tone
  • Peripheral vasomotor disturbances
  • Scoliosis/kyphosis
  • Growth retardation
  • Small cold hands and feet
  • Inappropriate laughing/screaming spells
  • Diminished response to pain
  • Intense eye communication (eye pointing)

Differential diagnosis edit

Signs of Rett syndrome that are similar to autism:

  • screaming fits
  • inconsolable crying
  • avoidance of eye contact
  • lack of social/emotional reciprocity
  • markedly impaired use of nonverbal behaviors to regulate social interaction
  • loss of speech
  • sensory problems
  • sleep regression

Signs of Rett syndrome that are also present in cerebral palsy (regression of the type seen in Rett syndrome would be unusual in cerebral palsy; this confusion could rarely be made):

Treatment edit

Currently there is no cure for Rett syndrome.[4] Treatment is directed towards improving function and addressing symptoms.[4] A multi-disciplinary team approach is typically used to treat the person throughout life. This team may include a primary care physician, physical therapist, occupational therapist, speech-language pathologist, nutritionist, and support services in academic and occupational settings. Some children may require special equipment and aids such as braces to arrest scoliosis, splints to modify hand movements, and nutritional programs to help them maintain adequate weight.[4]

Because of the increased risk of sudden cardiac death, when long QT syndrome is found on an annual screening EKG it is treated with an anti-arrhythmic such as a beta-blocker. There is some evidence that phenytoin may be more effective than a beta-blocker.[55]

While medicinal interventions to mitigate breathing challenges in children with Rett Syndrome (RTT) are still being developed,[56] children with RTT may be prescribed rebreathing techniques (e.g., rebreathing masks), oxygen delivery, or non-invasive ventilation as preventative or rescue breathing treatments.[57] High oxidative stress levels in individuals with RTT have exacerbated effects on their cardiorespiratory health and functionality,[56] dramatically increasing the risk for sudden cardiac death—an anomaly that has an associated 300x increased occurrence risk in children with Rett Syndrome.[58] Due to this, it is vital to closely monitor atypical breathing behaviors in children with RTT, making sure to effectively use lifesaving respiratory improvement devices and strategies as prescribed.[59]  

Prescribed treatment methods may vary depending on the breathing characteristic phenotype expressed by the child. Physicians have identified three major RTT breathing phenotypes; forceful breathers, feeble breathers, and apneustic breathers.[60] For forceful breathers, for example, rebreathing masks may be used while the child is awake.[60]

In Dec 2021, Australian company Neuren Pharmaceuticals reported positive results in a phase 3 trial of trofinetide for the treatment of Rett syndrome.[61] In Sep 2022, the FDA accepted a new drug application for trofinetide and granted it priority review.[62] In March 2023 the drug received FDA approval for treatment of Rett Syndrome.[63]

Prognosis edit

 
Girl with Rett syndrome with stereotyped hand movements

Males with pathogenic MECP2 mutations usually die within the first 2 years from severe encephalopathy, unless they have one or more extra X chromosomes, or have somatic mosaicism.

Male fetuses with the disorder rarely survive to term. Because the disease-causing gene is located on the X chromosome, a female born with an MECP2 mutation on her X chromosome has another X chromosome with an ostensibly normal copy of the same gene, while a male with the mutation on his X chromosome has no other X chromosome, only a Y chromosome; thus, he has no normal gene. Without a normal gene to provide normal proteins in addition to the abnormal proteins caused by a MECP2 mutation, the XY karyotype male fetus is unable to slow the development of the disease, hence the failure of many male fetuses with a MECP2 mutation to survive to term.

Females with a MECP2 mutation, however, have a non-mutant chromosome that provides them enough normal protein to survive longer. Research shows that males with Rett syndrome may result from Klinefelter's syndrome, in which the male has an XXY karyotype.[64] Thus, a non-mutant MECP2 gene is necessary for a Rett's-affected embryo to survive in most cases, and the embryo, male or female, must have another X chromosome.

There have, however, been several cases of 46,XY karyotype males with a MECP2 mutation (associated with classical Rett syndrome in females) carried to term, who were affected by neonatal encephalopathy and died before 2 years of age.[65] The incidence of Rett syndrome in males is unknown, partly owing to the low survival of male fetuses with the Rett syndrome-associated MECP2 mutations, and partly to differences between signs caused by MECP2 mutations and those caused by Rett's.[65]

Females can live up to 40 years or more. Laboratory studies on Rett syndrome may show abnormalities such as:

A high proportion of deaths are abrupt, but most have no identifiable cause; in some instances death is the result most likely of:

History edit

Andreas Rett, a pediatrician in Vienna Austria, first described the condition in 1966.[4][67] As his writings were in German, they did not become widely known in the English-speaking world.[6] Bengt Hagberg, a Swedish pediatrician, published an English article in 1983 and named the condition after Rett.[6] In 1999, Lebanese-American physician Huda Zoghbi discovered the mutation that causes the condition.[6][7]

Research edit

Gene therapy is under study in animal models to achieve regulated expression of a normal MECP2 gene.[4] In March 2022, Taysha Gene Therapies announced that they had received Clinical Trial Application (CTA) approval from Health Canada for a clinical trial of their investigational gene therapy for adult females with Rett Syndrome.[68]

In fiction edit

In August 2021, a novel by British author Victoria Scott, Patience, was published by Head of Zeus.[69] The novel featured a character with Rett syndrome and explored recent developments in gene therapy.

See also edit

References edit

  1. ^ Davis, Andrew S. (25 October 2010). [Rett syndrome Handbook of Pediatric Neuropsychology]. Springer Publishing Company. ISBN 978-0826157362. from the original on 5 November 2017. Rett initially called this syndrome cerebroaatrophic hyperammonemia, but the elevated ammonia levels in the bloodstream were later found to be only rarely associated with this condition (can Acker, Loncola, & Can Acker, 2005). {{cite book}}: Check |url= value (help)
  2. ^ "MeSH Browser". meshb.nlm.nih.gov. from the original on 4 December 2020. Retrieved 22 October 2019.
  3. ^ a b c d e f g h i j k l m "Rett syndrome". Genetics Home Reference. December 2013. from the original on 14 October 2017. Retrieved 14 October 2017.
  4. ^ a b c d e f g h i j k l m n o p q r s t u v w x y z aa ab ac ad ae af ag ah ai aj ak al am an ao ap aq "Rett Syndrome Fact Sheet". National Institute of Neurological Disorders and Stroke. from the original on 14 October 2017. Retrieved 14 October 2017.
  5. ^ "Rett Syndrome". NORD (National Organization for Rare Disorders). 2015. from the original on 19 February 2017. Retrieved 14 October 2017.
  6. ^ a b c d Percy, Alan (January 2014). "The American History of Rett Syndrome". Pediatric Neurology. 50 (1): 1–3. doi:10.1016/j.pediatrneurol.2013.08.018. PMC 3874243. PMID 24200039.
  7. ^ a b Amir, Ruthie; Van den Veyver, Ignatia; Wan, Mimi; Tran, Charles; Francke, Uta; Zoghbi, Huda (1999). "Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2". Nature Genetics. 23 (2): 185–8. doi:10.1038/13810. PMID 10508514. S2CID 3350350.
  8. ^ Neul, Jeffrey l.; Kaufmann, Walter E.; Glaze, Daniel G.; Christodoulou, John; Clarke, Angus J.; Bahi-Buisson, Nadia; Leonard, Helen; Bailey, Mark E. S.; Schanen, N. Carolyn; Zappella, Michele; Renieri, Alessandra; Huppke, Peter; Percy, Alan K.; et al. (Rettsearch Consortium) (2010). "Rett syndrome: Revised diagnostic criteria and nomenclature". Annals of Neurology. 68 (6): 944–50. doi:10.1002/ana.22124. PMC 3058521. PMID 21154482.
  9. ^ Ariani, Francesca; Hayek, Giuseppe; Rondinella, Dalila; Artuso, Rosangela; Mencarelli, Maria Antonietta; Spanhol-Rosseto, Ariele; Pollazzon, Marzia; Buoni, Sabrina; Spiga, Ottavia; Ricciardi, Sara; Meloni, Ilaria; Longo, Ilaria; Mari, Francesca; Broccoli, Vania; Zappella, Michele; Renieri, Alessandra (11 July 2008). "FOXG1 is Responsible for the Congenital Variant of Rett Syndrome". The American Journal of Human Genetics. 83 (1): 89–93. doi:10.1016/j.ajhg.2008.05.015. PMC 2443837. PMID 18571142.
  10. ^ Zappella, Michele (1992). "The rett girls with preserved speech". Brain and Development. 14 (2): 98–101. doi:10.1016/S0387-7604(12)80094-5. PMID 1621933. S2CID 4782923.
  11. ^ Skjeldal, O. H.; Von Tetzchner, S.; Jacobsen, K.; Smith, L.; Heiberg, A. (2007). "Rett Syndrome - Distribution of Phenotypes with Special Attention to the Preserved Speech Variant". Neuropediatrics. 26 (2): 87. doi:10.1055/s-2007-979732. PMID 7566462. S2CID 260243402.
  12. ^ Sørensen, E.; Viken, B. (20 February 1995). "[Rett syndrome a developmental disorder. Presentation of a variant with preserved speech]". Tidsskrift for den Norske Laegeforening (in Norwegian). 115 (5): 588–590. ISSN 0029-2001. PMID 7900110.
  13. ^ Zappella, M (1997). "The preserved speech variant of the Rett complex: A report of 8 cases". European Child & Adolescent Psychiatry. 6 (Suppl 1): 23–5. PMID 9452915.
  14. ^ Renieri, A.; Mari, F.; Mencarelli, M.A.; Scala, E.; Ariani, F.; Longo, I.; Meloni, I.; Cevenini, G.; Pini, G.; Hayek, G.; Zappella, M. (March 2009). "Diagnostic criteria for the Zappella variant of Rett syndrome (the preserved speech variant)". Brain and Development. 31 (3): 208–16. doi:10.1016/j.braindev.2008.04.007. PMID 18562141. S2CID 6223422.
  15. ^ Buoni, Sabrina; Zannolli, Raffaella; De Felice, Claudio; De Nicola, Anna; Guerri, Vanessa; Guerra, Beatrice; Casali, Stefania; Pucci, Barbara; Corbini, Letizia; Mari, Francesca; Renieri, Alessandra; Zappella, Michele; Hayek, Joseph (May 2010). "EEG features and epilepsy in MECP2-mutated patients with the Zappella variant of Rett syndrome". Clinical Neurophysiology. 121 (5): 652–7. doi:10.1016/j.clinph.2010.01.003. PMID 20153689. S2CID 12976926.
  16. ^ Huppke, Peter; Held, Melanie; Laccone, Franco; Hanefeld, Folker (2003). "The spectrum of phenotypes in females with Rett Syndrome". Brain and Development. 25 (5): 346–51. doi:10.1016/S0387-7604(03)00018-4. PMID 12850514. S2CID 9566219.
  17. ^ Gillberg, d. (1997). "Communication in Rett syndrome complex". European Child & Adolescent Psychiatry. 6 (Suppl 1): 21–2. PMID 9452914.
  18. ^ Zappella, Michele; Gillberg, Christopher; Ehlers, Stephan (1998). "The preserved speech variant: A subgroup of the Rett complex: A clinical report of 30 cases". Journal of Autism and Developmental Disorders. 28 (6): 519–26. doi:10.1023/A:1026052128305. PMID 9932238. S2CID 22152062.
  19. ^ Guy, J.; Gan, J.; Selfridge, J.; Cobb, S.; Bird, A. (2007). "Reversal of Neurological Defects in a Mouse Model of Rett Syndrome". Science. 315 (5815): 1143–7. Bibcode:2007Sci...315.1143G. doi:10.1126/science.1138389. PMC 7610836. PMID 17289941. S2CID 25172134.
  20. ^ Trappe, R.; Laccone, F.; Cobilanschi, J.; Meins, M.; Huppke, P.; Hanefeld, F.; Engel, W. (2001). "MECP2 Mutations in Sporadic Cases of Rett Syndrome Are Almost Exclusively of Paternal Origin". The American Journal of Human Genetics. 68 (5): 1093–101. doi:10.1086/320109. PMC 1226090. PMID 11309679.
  21. ^ Zoghbi, Huda Y.; Van Den Veyver, Ruthie E.; Wan, Ignatia B.; Tran, Mimi; Francke, Charles Q.; Zoghbi, Uta (1999). "Rett syndrome is caused by mutations in X-linked MECP2, encoding methyl-CpG-binding protein 2". Nature Genetics. 23 (2): 185–8. doi:10.1038/13810. PMID 10508514. S2CID 3350350.
  22. ^ "Rett syndrome". Genetics Home Reference. from the original on 27 July 2016. Retrieved 29 May 2016.
  23. ^ Merwick, Aine; O'Brien, Margaret; Delanty, Norman (2012). "Complex single gene disorders and epilepsy". Epilepsia. 53 (s4): 81–91. doi:10.1111/j.1528-1167.2012.03617.x. ISSN 1528-1167. PMID 22946725. S2CID 37226510.
  24. ^ Zoghbi, Huda Y.; Milstien, Sheldon; Butler, Ian J.; Smith, E. O'Brian; Kaufman, Seymour; Glaze, Daniel G.; Percy, Alan K. (1989). "Cerebrospinal fluid biogenic amines and biopterin in Rett syndrome". Annals of Neurology. 25 (1): 56–60. doi:10.1002/ana.410250109. PMID 2913929. S2CID 351243.
  25. ^ a b Roux, Jean-Christophe; Villard, Laurent (2009). "Biogenic Amines in Rett Syndrome: The Usual Suspects". Behavior Genetics. 40 (1): 59–75. doi:10.1007/s10519-009-9303-y. PMID 19851857. S2CID 20352177.
  26. ^ Hokfelt, T.; Martensson, R.; Bjorklund, A.; Kleinau, S.; Goldstein, M (1984). "Distribution maps of tyrosine-hydroxylase-immunoreactive neurons in the rat brain". In Bjorklund, A.; Hokfelt, T. (eds.). Handbook of Chemical Neuroanatomy. Classical Transmitters in the CNS, Part I. Vol. 2. New York: Elsevier. pp. 277–379.
  27. ^ Berridge, Craig W; Waterhouse, Barry D (2003). "The locus coeruleus–noradrenergic system: Modulation of behavioral state and state-dependent cognitive processes". Brain Research Reviews. 42 (1): 33–84. doi:10.1016/S0165-0173(03)00143-7. PMID 12668290. S2CID 477754.
  28. ^ a b Taneja, P.; Ogier, M.; Brooks-Harris, G.; Schmid, D. A.; Katz, D. M.; Nelson, S. B. (2009). "Pathophysiology of Locus Ceruleus Neurons in a Mouse Model of Rett Syndrome". Journal of Neuroscience. 29 (39): 12187–95. doi:10.1523/JNEUROSCI.3156-09.2009. PMC 2846656. PMID 19793977.
  29. ^ a b c d Roux, Jean-Christophe; Panayotis, Nicolas; Dura, Emmanuelle; Villard, Laurent (2009). "Progressive noradrenergic deficits in the locus coeruleus of Mecp2 deficient mice". Journal of Neuroscience Research. 88 (7): 1500–9. doi:10.1002/jnr.22312. PMID 19998492. S2CID 3404695.
  30. ^ Björklund, A.; Lindvall, O (1984). "Dopamine-containing systems in the CNS". In Björklund, A.; Hökfelt, T. (eds.). Handbook of Chemical Neuroanatomy. Classical Transmitters in the CNS, Part l. Vol. 2. New York: Elsevier. pp. 55–122.
  31. ^ Hokfelt, T.; Martensson, R.; Björklund, A.; Kleinau, S.; Goldstein, M. (1984). "Distribution maps of tyrosine-hydroxylase-immunoreactive neurons in the rat brain". In Björklund, A.; Hökfelt, T. (eds.). Handbook of Chemical Neuroanatomy. Classical Transmitters in the CNS, Part I. Vol. 2. New York: Elsevier. pp. 277–379.
  32. ^ Björklund, Anders; Dunnett, Stephen B. (2007). "Dopamine neuron systems in the brain: An update". Trends in Neurosciences. 30 (5): 194–202. doi:10.1016/j.tins.2007.03.006. PMID 17408759. S2CID 14239716.
  33. ^ Parent, André; Hazrati, Lili-Naz (1995). "Functional anatomy of the basal ganglia. I. The cortico-basal ganglia-thalamo-cortical loop". Brain Research Reviews. 20 (1): 91–127. doi:10.1016/0165-0173(94)00007-C. PMID 7711769. S2CID 28252990.
  34. ^ Gerfen, Charles R. (2000). "Molecular effects of dopamine on striatal-projection pathways". Trends in Neurosciences. 23 (10 Suppl): S64–70. doi:10.1016/S1471-1931(00)00019-7. PMID 11052222. S2CID 3965480.
  35. ^ Lees, Andrew J; Hardy, John; Revesz, Tamas (2009). "Parkinson's disease". The Lancet. 373 (9680): 2055–66. doi:10.1016/S0140-6736(09)60492-X. PMID 19524782. S2CID 42608600.
  36. ^ Dauer, William; Przedborski, Serge (2003). "Parkinson's Disease". Neuron. 39 (6): 889–909. doi:10.1016/S0896-6273(03)00568-3. PMID 12971891. S2CID 10400095.
  37. ^ Jenner, Peter (2009). "Functional models of Parkinson's disease: A valuable tool in the development of novel therapies". Annals of Neurology. 64: S16–29. doi:10.1002/ana.21489. PMID 19127585. S2CID 26065287.
  38. ^ Fitzgerald, Patricia M.; Jankovic, Joseph; Percy, Alan K. (1990). "Rett syndrome and associated movement disorders". Movement Disorders. 5 (3): 195–202. doi:10.1002/mds.870050303. PMID 2388636. S2CID 43376602.
  39. ^ Neul, Jeffrey L.; Zoghbi, Huda Y. (2004). "Rett Syndrome: A Prototypical Neurodevelopmental Disorder". The Neuroscientist. 10 (2): 118–28. doi:10.1177/1073858403260995. PMID 15070486. S2CID 9617631.
  40. ^ Segawa, Masaya (2005). "Early motor disturbances in Rett syndrome and its pathophysiological importance". Brain and Development. 27: S54–S58. doi:10.1016/j.braindev.2004.11.010. PMID 16182486. S2CID 30218744.
  41. ^ Guy, Jacky; Hendrich, Brian; Holmes, Megan; Martin, Joanne E.; Bird, Adrian (2001). "A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome". Nature Genetics. 27 (3): 322–6. doi:10.1038/85899. hdl:1842/727. PMID 11242117. S2CID 8698208.
  42. ^ Chen, Richard Z.; Akbarian, Schahram; Tudor, Matthew; Jaenisch, Rudolf (2001). "Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-like phenotype in mice". Nature Genetics. 27 (3): 327–31. doi:10.1038/85906. PMID 11242118. S2CID 24979562.
  43. ^ Nan, X; Ng, H. H.; Johnson, C. A.; Laherty, C. D.; Turner, B. M.; Eisenman, R. N.; Bird, A (1998). "Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex". Nature. 393 (6683): 386–9. Bibcode:1998Natur.393..386N. doi:10.1038/30764. PMID 9620804. S2CID 4427745.
  44. ^ Cheval, H; Guy, J; Merusi, C; De Sousa, D; Selfridge, J; Bird, A (2012). "Postnatal inactivation reveals enhanced requirement for MeCP2 at distinct age windows". Human Molecular Genetics. 21 (17): 3806–14. doi:10.1093/hmg/dds208. PMC 3412380. PMID 22653753.  
  45. ^ Ricceri, Laura; De Filippis, Bianca; Laviola, Giovanni (2008). "Mouse models of Rett syndrome: From behavioural phenotyping to preclinical evaluation of new therapeutic approaches". Behavioural Pharmacology. 19 (5–6): 501–17. doi:10.1097/FBP.0b013e32830c3645. PMID 18690105. S2CID 33364486.
  46. ^ Samaco, R. C.; Mandel-Brehm, C.; Chao, H.-T.; Ward, C. S.; Fyffe-Maricich, S. L.; Ren, J.; Hyland, K.; Thaller, C.; Maricich, S. M.; Humphreys, P.; Greer, J. J.; Percy, A.; Glaze, D. G.; Zoghbi, H. Y.; Neul, J. L. (2009). "Loss of MeCP2 in aminergic neurons causes cell-autonomous defects in neurotransmitter synthesis and specific behavioral abnormalities". Proceedings of the National Academy of Sciences. 106 (51): 21966–71. Bibcode:2009PNAS..10621966S. doi:10.1073/pnas.0912257106. JSTOR 40536204. PMC 2799790. PMID 20007372.
  47. ^ a b c d Panayotis, Nicolas; Pratte, Michel; Borges-Correia, Ana; Ghata, Adeline; Villard, Laurent; Roux, Jean-Christophe (2011). "Morphological and functional alterations in the substantia nigra pars compacta of the Mecp2-null mouse". Neurobiology of Disease. 41 (2): 385–97. doi:10.1016/j.nbd.2010.10.006. PMID 20951208. S2CID 25414717.
  48. ^ Sun, Yi E.; Wu, Hao (2006). "The Ups and Downs of BDNF in Rett Syndrome". Neuron. 49 (3): 321–3. doi:10.1016/j.neuron.2006.01.014. PMID 16446133.
  49. ^ a b Sharifi, Osman; Yasui, Dag H. (2021). "The Molecular Functions of MeCP2 in Rett Syndrome Pathology". Frontiers in Genetics. 12: 624290. doi:10.3389/fgene.2021.624290. PMC 8102816. PMID 33968128.
  50. ^ Ehrhart, Friederike; Coort, Susan L. M.; Cirillo, Elisa; Smeets, Eric; Evelo, Chris T.; Curfs, Leopold M. G. (25 November 2016). "Rett syndrome – biological pathways leading from MECP2 to disorder phenotypes". Orphanet Journal of Rare Diseases. 11 (1): 158. doi:10.1186/s13023-016-0545-5. PMC 5123333. PMID 27884167.
  51. ^ Tsai, Luke Y. (1992). "Is Rett syndrome a subtype of pervasive developmental disorders?" (PDF). Journal of Autism and Developmental Disorders. 22 (4): 551–61. doi:10.1007/BF01046327. hdl:2027.42/44607. PMID 1483976. S2CID 17817425. from the original on 29 August 2021. Retrieved 20 April 2018.
  52. ^ Abbeduto, Leonard; Ozonoff, Susan; Thurman, Angela John; McDuffie, Angela; Schweitzer, Julie (18 March 2014). Hales, Robert; Yudofsky, Stuart; Robert, Laura Weiss (eds.). Chapter 8. Neurodevelopmental Disorders, The American Psychiatric Publishing Textbook of Psychiatry (6 ed.). Arlington, VA: American Psychiatric Publishing. doi:10.1176/appi.books.9781585625031.rh08. ISBN 978-1-58562-444-7. S2CID 241966275.
  53. ^ a b "Rett syndrome Tests and diagnosis". Mayo Clinic. from the original on 30 October 2017.
  54. ^ a b c d e f "About Rett syndrome - Rett Syndrome Diagnosis". rettsyndrome.org. International Rett Syndrome Foundation. from the original on 29 October 2017. Retrieved 10 May 2020.
  55. ^ McCauley, Mark D.; Wang, Tiannan; Mike, Elise; Herrera, Jose; Beavers, David L.; Huang, Teng-Wei; Ward, Christopher S.; Skinner, Steven; Percy, Alan K. (14 December 2011). "Pathogenesis of Lethal Cardiac Arrhythmias in Mecp2 Mutant Mice: Implication for Therapy in Rett Syndrome". Science Translational Medicine. 3 (113): 113ra125. doi:10.1126/scitranslmed.3002982. ISSN 1946-6234. PMC 3633081. PMID 22174313.
  56. ^ a b Mackay, Jessica; Downs, Jenny; Wong, Kingsley; Heyworth, Jane; Epstein, Amy; Leonard, Helen (2017). "Autonomic breathing abnormalities in Rett syndrome: caregiver perspectives in an international database study". Journal of Neurodevelopmental Disorders. 9: 15. doi:10.1186/s11689-017-9196-7. ISSN 1866-1947. PMC 5410057. PMID 28465761.
  57. ^ Harish Kumar, S. (19 January 2017). "Cardio respiratory physiotherapy management in Rett's syndrome - Literature review". International Journal of Pharma and Bio Sciences: 5 – via ResearchGate.
  58. ^ Kyle, Stephanie M.; Vashi, Neeti; Justice, Monica J. (February 2018). "Rett syndrome: a neurological disorder with metabolic components". Open Biology. 8 (2): 170216. doi:10.1098/rsob.170216. ISSN 2046-2441. PMC 5830535. PMID 29445033.
  59. ^ De Felice, Claudio; Maffei, Silvia; Signorini, Cinzia; Leoncini, Silvia; Lunghetti, Stefano; Valacchi, Giuseppe; D'Esposito, Maurizio; Filosa, Stefania; Della Ragione, Floriana; Butera, Gianfranco; Favilli, Roberto (April 2012). "Subclinical myocardial dysfunction in Rett syndrome". European Heart Journal: Cardiovascular Imaging. 13 (4): 339–345. doi:10.1093/ejechocard/jer256. ISSN 2047-2412. PMID 22113206. from the original on 29 November 2021. Retrieved 29 November 2021.
  60. ^ a b Smeets, Eric E. J.; Julu, Peter O. O.; Waardenburg, Dick van; Engerström, Ingegerd Witt; Hansen, Stig; Apartopoulos, Flora; Curfs, Leopold M. G.; Schrander-Stumpel, Connie T. R. M. (1 November 2006). "Management of a severe forceful breather with Rett Syndrome using carbogen". Brain and Development. 28 (10): 625–632. doi:10.1016/j.braindev.2006.04.010. ISSN 0387-7604. PMID 16765005. S2CID 15545729. from the original on 1 October 2022. Retrieved 29 November 2021.
  61. ^ Positive top-line results from pivotal Phase 3 trial in Rett syndrome (PDF) (Report). Australian Stock Exchange. 7 December 2021.
  62. ^ Rett Syndrome New Drug Application accepted for Priority Review by FDA (PDF) (Report). Australian Stock Exchange. 13 September 2022.
  63. ^ "Breakthrough drug from Dame Margaret Brimble's lab". University of Auckland. 11 March 2023. Retrieved 6 May 2023.
  64. ^ Schwartzman, J. S.; Bernardino, Andrea; Nishimura, Agnes; Gomes, Raquel R.; Zatz, Mayana (2001). "Rett Syndrome in a Boy with a 47,XXY Karyotype Confirmed by a Rare Mutation in the MECP2 Gene". Neuropediatrics. 32 (3): 162–4. doi:10.1055/s-2001-16620. PMID 11521215. S2CID 260240039.
  65. ^ a b Hardwick, Simon A; Reuter, Kirsten; Williamson, Sarah L; Vasudevan, Vidya; Donald, Jennifer; Slater, Katrina; Bennetts, Bruce; Bebbington, Ami; Leonard, Helen; Williams, Simon R; Smith, Robert L; Cloosterman, Desiree; Christodoulou, John (2007). "Delineation of large deletions of the MECP2 gene in Rett syndrome patients, including a familial case with a male proband". European Journal of Human Genetics. 15 (12): 1218–29. doi:10.1038/sj.ejhg.5201911. PMID 17712354.
  66. ^ Acampa, M.; Guideri, F. (May 2006). "Cardiac disease and Rett syndrome". Archives of Disease in Childhood. 91 (5): 440–443. doi:10.1136/adc.2005.090290. ISSN 1468-2044. PMC 2082747. PMID 16632674.
  67. ^ Rett, A. (10 September 1966). "[On an unusual brain atrophy syndrome in hyperammonemia in childhood]". Wiener Medizinische Wochenschrift (in German). 116 (37): 723–726. ISSN 0043-5341. PMID 5300597.
  68. ^ "Taysha Gene Therapies Announces Initiation of Clinical Development of TSHA-102 in Rett Syndrome". Taysha Gene Therapies. March 2022. from the original on 30 March 2022. Retrieved 6 May 2022.
  69. ^ "title". Head of Zeus. from the original on 1 October 2022. Retrieved 7 May 2022.

rett, syndrome, confused, with, tourette, syndrome, genetic, disorder, that, typically, becomes, apparent, after, months, almost, exclusively, females, symptoms, include, impairments, language, coordination, repetitive, movements, those, affected, often, have,. Not to be confused with Tourette syndrome Rett syndrome RTT is a genetic disorder that typically becomes apparent after 6 18 months of age and almost exclusively in females 3 Symptoms include impairments in language and coordination and repetitive movements 3 Those affected often have slower growth difficulty walking and a smaller head size 3 4 Complications of Rett syndrome can include seizures scoliosis and sleeping problems 3 The severity of the condition is variable 4 Rett syndromeOther namesCerebroatrophic hyperammonemia obsolete 1 dementia ataxia and loss of purposeful hand use syndrome 2 A girl with Rett Syndrome smiling at the cameraSpecialtyPsychiatry pediatrics neurologySymptomsImpairments in language and coordination and repetitive movements slower growth smaller head 3 ComplicationsSeizures scoliosis sleeping problems 3 Usual onsetAfter 6 18 months of age 3 DurationLifelong 4 CausesMutation in the MECP2 gene 3 Diagnostic methodBased on symptoms genetic testing 4 Differential diagnosisAngelman syndrome autism cerebral palsy Childhood disintegrative disorder various neurodegenerative disorders 5 TreatmentSpecial education physiotherapy braces 4 MedicationAnticonvulsants 4 PrognosisLife expectancy for many is middle age 4 Frequency1 in 8 500 females 3 Lethal in males with rare exceptions Rett syndrome is due to a genetic mutation in the MECP2 gene 3 on the X chromosome 4 It almost always occurs as a new mutation with less than one percent of cases being inherited 3 4 It occurs almost exclusively in girls 3 boys who have a similar mutation typically die shortly after birth 4 Diagnosis is based on the symptoms and can be confirmed with genetic testing 4 There is no known cure for Rett syndrome 4 Treatment is directed at improving symptoms 4 Anticonvulsants may be used to help with seizures 4 Special education physiotherapy and leg braces may also be useful depending on the needs of the child 4 Many of those with the condition live into middle age 4 The condition affects about 1 in 8 500 females 3 In 1999 Lebanese American physician Huda Zoghbi discovered the mutation that causes the condition 6 7 Contents 1 Signs and symptoms 1 1 Stage I 1 2 Stage II 1 3 Stage III 1 4 Stage IV 1 5 Variants 2 Cause 2 1 Sporadic mutations 2 2 Germline mutations 3 Mechanism 3 1 Pontine noradrenergic deficits 3 2 Midbrain dopaminergic disturbances 3 3 Molecular functions of MECP2 in Rett syndrome pathology 3 4 Interactive pathway map 4 Diagnosis 4 1 Differential diagnosis 5 Treatment 6 Prognosis 7 History 8 Research 9 In fiction 10 See also 11 ReferencesSigns and symptoms editStage I edit Stage I called early onset typically begins between 6 and 18 months of age 4 This stage is often overlooked because symptoms of the disorder may be somewhat vague and parents and doctors may not notice the subtle slowing of development at first 4 The infant may begin to show less eye contact and have reduced interest in toys There may be delays in gross motor skills such as sitting or crawling 4 Hand wringing and decreasing head growth may occur but not enough to draw attention This stage usually lasts for a few months but can continue for more than a year 4 Stage II edit Stage II or the rapid destructive stage usually begins between ages 1 and 4 and may last for weeks or months 4 Its onset may be rapid or gradual as the child loses purposeful hand skills and spoken language 4 Characteristic hand movements such as wringing washing clapping or tapping as well as repeatedly moving the hands to the mouth often begin during this stage which is called mouthing 4 The child may hold the hands clasped behind the back or held at the sides with random touching grasping and releasing 4 The movements continue while the child is awake but disappear during sleep 4 Breathing irregularities such as episodes of apnea and hyperventilation may occur although breathing usually improves during sleep 4 Some girls also display autistic like symptoms such as loss of social interaction and communication 4 Walking may be unsteady and initiating motor movements can be difficult Slowed head growth is usually noticed during this stage 4 Stage III edit Stage III or the plateau or pseudo stationary stage usually begins between ages 2 and 10 and can last for years 4 Apraxia motor problems and seizures are prominent during this stage 4 However there may be improvement in behavior with less irritability crying and autistic like features 4 In stage III there may be more interest in the surroundings and alertness attention span and communication skills may improve 4 Many girls remain in this stage for most of their lives 4 Stage IV edit Stage IV or the late motor deterioration stage can last for years or decades 4 Prominent features include reduced mobility curvature of the spine and muscle weakness rigidity spasticity and increased muscle tone with abnormal posturing of an arm or leg 4 Girls who were previously able to walk may stop walking 4 Cognition communication or hand skills generally do not decline in stage IV 4 Repetitive hand movements may decrease and eye gaze usually improves 4 Variants edit The signs and symptoms of the typical form of the Rett syndrome are well described In addition to the classical form of Rett syndrome several atypical forms have been described over the years 8 the main groups are Congenital variant Rolando variant in this severe subtype of Rett syndrome the development of the patients and their head circumference are abnormal from birth 9 The typical gaze of Rett syndrome patients is usually absent Zappella variant of Rett Syndrome or preserved speech variant in this subtype of Rett syndrome the patients acquire some manual skills and language is partially recovered around the age of 5 years that is after the regression phase Height weight and head circumference are often in the normal range and a good gross motor function can be observed 10 11 12 13 14 15 The Zappella variant is a milder form of Rett syndrome Hanefeld variant or early epilepsy variant In this form of Rett syndrome the patients have epilepsy before 5 months of age 16 The definition itself of the Rett syndrome has been refined over the years as the atypical forms subsist near to the classical form Hagberg amp Gillberg 1993 the Rett Complex terminology has been introduced 17 18 Cause editGenetically Rett syndrome RTT is caused by mutations in the gene MECP2 located on the X chromosome which is involved in transcriptional silencing and epigenetic regulation of methylated DNA and can arise sporadically or from germline mutations In less than 10 of RTT cases mutations in the genes CDKL5 or FOXG1 have also been found to resemble it medical citation needed Rett syndrome is initially diagnosed by clinical observation but the diagnosis is definitive when there is a genetic defect in the MECP2 gene It has been argued that Rett syndrome is in fact a neurodevelopmental condition as opposed to a neurodegenerative condition One piece of evidence for this is that mice with induced Rett Syndrome show no neuronal death and some studies have suggested that their phenotypes can be partially rescued by adding functional MECP2 gene back when they are adults This information has also helped lead to further studies aiming to treat the disorder 19 Sporadic mutations edit In at least 95 of Rett syndrome cases the cause is a de novo mutation in the child That is it is not inherited from either parent Parents are generally genotypically normal without a MECP2 mutation citation needed In cases of the sporadic form of RTT the mutated MECP2 is thought to derive almost exclusively from a de novo mutation on the male copy of the X chromosome 20 It is not yet known what causes the sperm to mutate and such mutations are rare Germline mutations edit It can also be inherited from phenotypically normal mothers who have a germline mutation in the gene encoding methyl CpG binding protein 2 MeCP2 21 In these cases inheritance follows an X linked dominant pattern and is seen almost exclusively in females as most males die in utero or shortly after birth 22 MECP2 is found near the end of the long arm of the X chromosome at Xq28 An atypical form of RTT characterized by infantile spasms or early onset epilepsy can also be caused by a mutation to the gene encoding cyclin dependent kinase like 5 CDKL5 As stated by Aine Merwick Margaret O Brien and Norman Delanty in an article on gene disorders titled Complex single gene disorders and epilepsy Rett syndrome affects one in every 12 500 female live births by age 12 years 23 Mechanism edit nbsp The location of the gene responsible for Rett syndromePontine noradrenergic deficits edit Brain levels of norepinephrine are lower in people with Rett syndrome 24 reviewed in 25 The genetic loss of MECP2 changes the properties of cells in the locus coeruleus the exclusive source of noradrenergic innervation to the cerebral cortex and hippocampus 26 27 These changes include hyperexcitability and decreased functioning of its noradrenergic innervation 28 Moreover a reduction of the tyrosine hydroxylase Th mRNA level the rate limiting enzyme in catecholamine synthesis was detected in the whole pons of MECP2 null male as well as in adult heterozygous MECP2 female mice 29 Using immunoquantitative techniques a decrease of Th protein staining level number of locus coeruleus Th expressing neurons and density of dendritic arborization surrounding the structure was shown in symptomatic MeCP2 deficient mice 29 However locus coeruleus cells are not dying but are more likely losing their fully mature phenotype since no apoptotic neurons in the pons were detected 29 Researchers have concluded that Because these neurons are a pivotal source of norepinephrine throughout the brainstem and forebrain and are involved in the regulation of diverse functions disrupted in Rett syndrome such as respiration and cognition we hypothesize that the locus coeruleus is a critical site at which loss of MECP2 results in CNS dysfunction The restoration of normal locus coeruleus function may therefore be of potential therapeutic value in the treatment of Rett syndrome 28 29 Midbrain dopaminergic disturbances edit The majority of dopamine in the mammalian brain is synthesized by nuclei located in the mesencephalon The substantia nigra pars compacta SNpc the ventral tegmental area VTA and the retrorubral field RRF contain dopaminergic neurons expressing tyrosine hydroxylase Th i e the rate limiting enzyme in catecholamine synthesis 30 31 32 The nigro striatal pathway originates from the SNpc its principal rostral target is the caudate putamen CPu which it irradiates through the median forebrain bundle MFB This connection is involved in the tight modulation of motor strategies computed by a cortico basal ganglia thalamo cortical loop 33 Indeed based on the canonical anatomofunctional model of basal ganglia nigrostriatal dopamine is able to modulate the motor loop by acting on dopaminergic receptors located on striatal GABAergic medium spiny neurons 34 Dysregulation of the nigrostriatal pathway is causative from Parkinson disease PD in humans 35 Toxic and or genetic ablation of SNpc neurons produces experimental parkinsonism in mice and primates 36 The common features of PD and PD animal models are motor impairments 37 hypotonia bradykinesia hypokinesia RTT pathology in some aspects overlaps the motor phenotype observed in PD patients 38 39 40 Several neuropathological studies on postmortem brain samples argued for an SNpc alteration evidenced by neuromelanin hypopigmentation reduction in the structure area and even controversially signs of apoptosis In parallel a hypometabolism was underlined by a reduction of several catecholamines dopamine noradrenaline adrenaline and their principal metabolic by products 25 Mouse models of RTT are available the most studied are constitutively deleted Mecp2 mice developed by Adrian Bird or Katelyn McCormick laboratories 41 42 43 44 In accordance with the motor spectrum of the RTT phenotype Mecp2 null mice show motor abnormalities from postnatal day 30 that worsen until death These models offer a crucial substrate to elucidate the molecular and neuroanatomical correlates of MeCP2 deficiency 45 Recently 2008 it was shown that the conditional deletion of Mecp2 in catecholaminergic neurons by crossing of Th Cre mice with loxP flanked Mecp2 ones recapitulates a motor symptomatology it was further documented that brain levels of Th in mice lacking MeCP2 in catecholaminergic neurons only are reduced participating to the motor phenotype 46 However the most studied model for the evaluation of therapeutics is the Mecp2 null mouse totally devoid of MeCP2 In this context a reduction in the number and soma size of Th expressing neurons is present from 5 weeks of age and is accompanied by a decrease of Th immunoreactivity in the caudate putamen the principal target of dopaminergic neurons arising from the SNpc 47 Moreover a neurochemical analysis of dopaminergic contents in microdissected midbrain and striatal areas revealed a reduction of dopamine at five and nine weeks of age It is noteworthy that later on at nine weeks the morphological parameters remain altered but not worsened whereas the phenotype progresses and behavioral deficits are more severe The amount of fully activated Th Serine40 phosphorylated isoform in neurons that remain in the SNpc is mildly affected at 5 weeks but severely impaired by 9 weeks 47 Finally using a chronic and oral L Dopa treatment on MeCP2 deficient mice authors reported an amelioration of some of the motor deficits previously identified 47 Altogether these results argue for an alteration of the nigrostriatal dopaminergic pathway in MeCP2 deficient animals as a contributor of the neuromotor deficits 47 There is an association of Rett syndrome with brain derived neurotrophic factor BDNF 48 Molecular functions of MECP2 in Rett syndrome pathology edit As reviewed by Sharifi and Yasui 49 MECP2 protein encoded by the MECP2 gene binds to DNA with a high affinity for CpG methylated DNA sites and affects transcription MECP2 can bind to 5mc 5 methylcytosine and 5hmc 5 hydroxymethylcytosine with similar affinity and these dinucleotides account for the majority of MECP2 binding sites in the mammalian genome MECP2 is involved in higher order chromatin organization and appears necessary for compacting chromosomes MECP2 binding to DNA influences mRNA splicing events MECP2 also appears to function in DNA repair processes MECP2 deficient female mice have elevated rates of cell death when exposed to DNA damaging agents and are prone to early senescence 49 Interactive pathway map edit An interactive pathway map of Rett syndrome has been published 50 Diagnosis edit nbsp A girl with Rett Syndrome mouthing her hands a common behavior with Rett SyndromePrior to the discovery of a genetic cause Rett syndrome had been designated as a pervasive developmental disorder by the Diagnostic and Statistical Manual of Mental Disorders DSM together with the autism spectrum disorders Some argued against this conclusive assignment because RTT resembles non autistic disorders such as fragile X syndrome tuberous sclerosis or Down syndrome that also exhibit autistic features 51 After research proved the molecular mechanism in 2013 the DSM 5 removed the syndrome altogether from classification as a mental disorder 52 Rett syndrome diagnosis involves close observation of the child s growth and development to observe any abnormalities in regards to developmental milestones 53 A diagnosis is considered when decreased head growth is observed Conditions with similar symptoms must first be ruled out 53 There are certain criteria that must be met for the diagnosis A blood test can rule in or rule out the presence of the MECP2 mutation however this mutation is present in other conditions as well 54 For a classic diagnosis all four criteria for ruling in a diagnosis must be met as well as the two criteria for ruling out a diagnosis Supportive criteria may also be present but are not required for diagnosis For an atypical or variant diagnosis at least two of the four criteria for ruling in the diagnosis must be met as well as five of the eleven supportive criteria A period of symptom regression followed by recovery or symptom stabilization must also occur 54 Children are often misdiagnosed as having autism cerebral palsy or another form of developmental delay A positive test for the MECP2 mutation is not enough to make a diagnosis 54 Ruling in 54 Decreased or loss of use of fine motor skills Decreased or loss of verbal speech Abnormalities during gait Repetitive hand movements such as wringing squeezing or clapping tappingRuling out 54 Traumatic or anoxic hypoxic brain injury neurometabolic disease or severe infection that may better explain symptoms Abnormal psychomotor development during the first six months of lifeSupportive criteria 54 Breathing disturbances when awake Bruxism while awake Impaired sleep pattern Abnormal muscle tone Peripheral vasomotor disturbances Scoliosis kyphosis Growth retardation Small cold hands and feet Inappropriate laughing screaming spells Diminished response to pain Intense eye communication eye pointing Differential diagnosis edit This section does not cite any sources Please help improve this section by adding citations to reliable sources Unsourced material may be challenged and removed January 2020 Learn how and when to remove this template message Signs of Rett syndrome that are similar to autism screaming fits inconsolable crying avoidance of eye contact lack of social emotional reciprocity markedly impaired use of nonverbal behaviors to regulate social interaction loss of speech sensory problems sleep regression Signs of Rett syndrome that are also present in cerebral palsy regression of the type seen in Rett syndrome would be unusual in cerebral palsy this confusion could rarely be made possible short stature sometimes with unusual body proportions because of difficulty walking or malnutrition caused by difficulty swallowing hypotonia delayed or absent ability to walk gait movement difficulties ataxia microcephaly in some abnormally small head poor head growth gastrointestinal problems some forms of spasticity chorea spasmodic movements of hand or facial muscles dystonia bruxism grinding of teethTreatment editMain article Treatment of Rett syndrome Currently there is no cure for Rett syndrome 4 Treatment is directed towards improving function and addressing symptoms 4 A multi disciplinary team approach is typically used to treat the person throughout life This team may include a primary care physician physical therapist occupational therapist speech language pathologist nutritionist and support services in academic and occupational settings Some children may require special equipment and aids such as braces to arrest scoliosis splints to modify hand movements and nutritional programs to help them maintain adequate weight 4 Because of the increased risk of sudden cardiac death when long QT syndrome is found on an annual screening EKG it is treated with an anti arrhythmic such as a beta blocker There is some evidence that phenytoin may be more effective than a beta blocker 55 While medicinal interventions to mitigate breathing challenges in children with Rett Syndrome RTT are still being developed 56 children with RTT may be prescribed rebreathing techniques e g rebreathing masks oxygen delivery or non invasive ventilation as preventative or rescue breathing treatments 57 High oxidative stress levels in individuals with RTT have exacerbated effects on their cardiorespiratory health and functionality 56 dramatically increasing the risk for sudden cardiac death an anomaly that has an associated 300x increased occurrence risk in children with Rett Syndrome 58 Due to this it is vital to closely monitor atypical breathing behaviors in children with RTT making sure to effectively use lifesaving respiratory improvement devices and strategies as prescribed 59 Prescribed treatment methods may vary depending on the breathing characteristic phenotype expressed by the child Physicians have identified three major RTT breathing phenotypes forceful breathers feeble breathers and apneustic breathers 60 For forceful breathers for example rebreathing masks may be used while the child is awake 60 In Dec 2021 Australian company Neuren Pharmaceuticals reported positive results in a phase 3 trial of trofinetide for the treatment of Rett syndrome 61 In Sep 2022 the FDA accepted a new drug application for trofinetide and granted it priority review 62 In March 2023 the drug received FDA approval for treatment of Rett Syndrome 63 Prognosis edit nbsp Girl with Rett syndrome with stereotyped hand movementsMales with pathogenic MECP2 mutations usually die within the first 2 years from severe encephalopathy unless they have one or more extra X chromosomes or have somatic mosaicism Male fetuses with the disorder rarely survive to term Because the disease causing gene is located on the X chromosome a female born with an MECP2 mutation on her X chromosome has another X chromosome with an ostensibly normal copy of the same gene while a male with the mutation on his X chromosome has no other X chromosome only a Y chromosome thus he has no normal gene Without a normal gene to provide normal proteins in addition to the abnormal proteins caused by a MECP2 mutation the XY karyotype male fetus is unable to slow the development of the disease hence the failure of many male fetuses with a MECP2 mutation to survive to term Females with a MECP2 mutation however have a non mutant chromosome that provides them enough normal protein to survive longer Research shows that males with Rett syndrome may result from Klinefelter s syndrome in which the male has an XXY karyotype 64 Thus a non mutant MECP2 gene is necessary for a Rett s affected embryo to survive in most cases and the embryo male or female must have another X chromosome There have however been several cases of 46 XY karyotype males with a MECP2 mutation associated with classical Rett syndrome in females carried to term who were affected by neonatal encephalopathy and died before 2 years of age 65 The incidence of Rett syndrome in males is unknown partly owing to the low survival of male fetuses with the Rett syndrome associated MECP2 mutations and partly to differences between signs caused by MECP2 mutations and those caused by Rett s 65 Females can live up to 40 years or more Laboratory studies on Rett syndrome may show abnormalities such as EEG abnormalities from 2 years of age atypical brain glycolipids elevated CSF levels of beta endorphin and glutamate reduction of substance P decreased levels of CSF nerve growth factorsA high proportion of deaths are abrupt but most have no identifiable cause in some instances death is the result most likely of spontaneous brainstem dysfunction cardiac arrest likely due to long QT syndrome ventricular tachycardia or other arrhythmias 66 seizures gastric perforationHistory editAndreas Rett a pediatrician in Vienna Austria first described the condition in 1966 4 67 As his writings were in German they did not become widely known in the English speaking world 6 Bengt Hagberg a Swedish pediatrician published an English article in 1983 and named the condition after Rett 6 In 1999 Lebanese American physician Huda Zoghbi discovered the mutation that causes the condition 6 7 Research editGene therapy is under study in animal models to achieve regulated expression of a normal MECP2 gene 4 In March 2022 Taysha Gene Therapies announced that they had received Clinical Trial Application CTA approval from Health Canada for a clinical trial of their investigational gene therapy for adult females with Rett Syndrome 68 In fiction editIn August 2021 a novel by British author Victoria Scott Patience was published by Head of Zeus 69 The novel featured a character with Rett syndrome and explored recent developments in gene therapy See also editList of syndromes Toxidrome Symptom Sequence medicine Characteristics of syndromic ASD conditionsReferences edit Davis Andrew S 25 October 2010 Rett syndrome Handbook of Pediatric Neuropsychology Springer Publishing Company ISBN 978 0826157362 Archived from the original on 5 November 2017 Rett initially called this syndrome cerebroaatrophic hyperammonemia but the elevated ammonia levels in the bloodstream were later found to be only rarely associated with this condition can Acker Loncola amp Can Acker 2005 a href Template Cite book html title Template Cite book cite book a Check url value help MeSH Browser meshb nlm nih gov Archived from the original on 4 December 2020 Retrieved 22 October 2019 a b c d e f g h i j k l m Rett syndrome Genetics Home Reference December 2013 Archived from the original on 14 October 2017 Retrieved 14 October 2017 a b c d e f g h i j k l m n o p q r s t u v w x y z aa ab ac ad ae af ag ah ai aj ak al am an ao ap aq Rett Syndrome Fact Sheet National Institute of Neurological Disorders and Stroke Archived from the original on 14 October 2017 Retrieved 14 October 2017 Rett Syndrome NORD National Organization for Rare Disorders 2015 Archived from the original on 19 February 2017 Retrieved 14 October 2017 a b c d Percy Alan January 2014 The American History of Rett Syndrome Pediatric Neurology 50 1 1 3 doi 10 1016 j pediatrneurol 2013 08 018 PMC 3874243 PMID 24200039 a b Amir Ruthie Van den Veyver Ignatia Wan Mimi Tran Charles Francke Uta Zoghbi Huda 1999 Rett syndrome is caused by mutations in X linked MECP2 encoding methyl CpG binding protein 2 Nature Genetics 23 2 185 8 doi 10 1038 13810 PMID 10508514 S2CID 3350350 Neul Jeffrey l Kaufmann Walter E Glaze Daniel G Christodoulou John Clarke Angus J Bahi Buisson Nadia Leonard Helen Bailey Mark E S Schanen N Carolyn Zappella Michele Renieri Alessandra Huppke Peter Percy Alan K et al Rettsearch Consortium 2010 Rett syndrome Revised diagnostic criteria and nomenclature Annals of Neurology 68 6 944 50 doi 10 1002 ana 22124 PMC 3058521 PMID 21154482 Ariani Francesca Hayek Giuseppe Rondinella Dalila Artuso Rosangela Mencarelli Maria Antonietta Spanhol Rosseto Ariele Pollazzon Marzia Buoni Sabrina Spiga Ottavia Ricciardi Sara Meloni Ilaria Longo Ilaria Mari Francesca Broccoli Vania Zappella Michele Renieri Alessandra 11 July 2008 FOXG1 is Responsible for the Congenital Variant of Rett Syndrome The American Journal of Human Genetics 83 1 89 93 doi 10 1016 j ajhg 2008 05 015 PMC 2443837 PMID 18571142 Zappella Michele 1992 The rett girls with preserved speech Brain and Development 14 2 98 101 doi 10 1016 S0387 7604 12 80094 5 PMID 1621933 S2CID 4782923 Skjeldal O H Von Tetzchner S Jacobsen K Smith L Heiberg A 2007 Rett Syndrome Distribution of Phenotypes with Special Attention to the Preserved Speech Variant Neuropediatrics 26 2 87 doi 10 1055 s 2007 979732 PMID 7566462 S2CID 260243402 Sorensen E Viken B 20 February 1995 Rett syndrome a developmental disorder Presentation of a variant with preserved speech Tidsskrift for den Norske Laegeforening in Norwegian 115 5 588 590 ISSN 0029 2001 PMID 7900110 Zappella M 1997 The preserved speech variant of the Rett complex A report of 8 cases European Child amp Adolescent Psychiatry 6 Suppl 1 23 5 PMID 9452915 Renieri A Mari F Mencarelli M A Scala E Ariani F Longo I Meloni I Cevenini G Pini G Hayek G Zappella M March 2009 Diagnostic criteria for the Zappella variant of Rett syndrome the preserved speech variant Brain and Development 31 3 208 16 doi 10 1016 j braindev 2008 04 007 PMID 18562141 S2CID 6223422 Buoni Sabrina Zannolli Raffaella De Felice Claudio De Nicola Anna Guerri Vanessa Guerra Beatrice Casali Stefania Pucci Barbara Corbini Letizia Mari Francesca Renieri Alessandra Zappella Michele Hayek Joseph May 2010 EEG features and epilepsy in MECP2 mutated patients with the Zappella variant of Rett syndrome Clinical Neurophysiology 121 5 652 7 doi 10 1016 j clinph 2010 01 003 PMID 20153689 S2CID 12976926 Huppke Peter Held Melanie Laccone Franco Hanefeld Folker 2003 The spectrum of phenotypes in females with Rett Syndrome Brain and Development 25 5 346 51 doi 10 1016 S0387 7604 03 00018 4 PMID 12850514 S2CID 9566219 Gillberg d 1997 Communication in Rett syndrome complex European Child amp Adolescent Psychiatry 6 Suppl 1 21 2 PMID 9452914 Zappella Michele Gillberg Christopher Ehlers Stephan 1998 The preserved speech variant A subgroup of the Rett complex A clinical report of 30 cases Journal of Autism and Developmental Disorders 28 6 519 26 doi 10 1023 A 1026052128305 PMID 9932238 S2CID 22152062 Guy J Gan J Selfridge J Cobb S Bird A 2007 Reversal of Neurological Defects in a Mouse Model of Rett Syndrome Science 315 5815 1143 7 Bibcode 2007Sci 315 1143G doi 10 1126 science 1138389 PMC 7610836 PMID 17289941 S2CID 25172134 Trappe R Laccone F Cobilanschi J Meins M Huppke P Hanefeld F Engel W 2001 MECP2 Mutations in Sporadic Cases of Rett Syndrome Are Almost Exclusively of Paternal Origin The American Journal of Human Genetics 68 5 1093 101 doi 10 1086 320109 PMC 1226090 PMID 11309679 Zoghbi Huda Y Van Den Veyver Ruthie E Wan Ignatia B Tran Mimi Francke Charles Q Zoghbi Uta 1999 Rett syndrome is caused by mutations in X linked MECP2 encoding methyl CpG binding protein 2 Nature Genetics 23 2 185 8 doi 10 1038 13810 PMID 10508514 S2CID 3350350 Rett syndrome Genetics Home Reference Archived from the original on 27 July 2016 Retrieved 29 May 2016 Merwick Aine O Brien Margaret Delanty Norman 2012 Complex single gene disorders and epilepsy Epilepsia 53 s4 81 91 doi 10 1111 j 1528 1167 2012 03617 x ISSN 1528 1167 PMID 22946725 S2CID 37226510 Zoghbi Huda Y Milstien Sheldon Butler Ian J Smith E O Brian Kaufman Seymour Glaze Daniel G Percy Alan K 1989 Cerebrospinal fluid biogenic amines and biopterin in Rett syndrome Annals of Neurology 25 1 56 60 doi 10 1002 ana 410250109 PMID 2913929 S2CID 351243 a b Roux Jean Christophe Villard Laurent 2009 Biogenic Amines in Rett Syndrome The Usual Suspects Behavior Genetics 40 1 59 75 doi 10 1007 s10519 009 9303 y PMID 19851857 S2CID 20352177 Hokfelt T Martensson R Bjorklund A Kleinau S Goldstein M 1984 Distribution maps of tyrosine hydroxylase immunoreactive neurons in the rat brain In Bjorklund A Hokfelt T eds Handbook of Chemical Neuroanatomy Classical Transmitters in the CNS Part I Vol 2 New York Elsevier pp 277 379 Berridge Craig W Waterhouse Barry D 2003 The locus coeruleus noradrenergic system Modulation of behavioral state and state dependent cognitive processes Brain Research Reviews 42 1 33 84 doi 10 1016 S0165 0173 03 00143 7 PMID 12668290 S2CID 477754 a b Taneja P Ogier M Brooks Harris G Schmid D A Katz D M Nelson S B 2009 Pathophysiology of Locus Ceruleus Neurons in a Mouse Model of Rett Syndrome Journal of Neuroscience 29 39 12187 95 doi 10 1523 JNEUROSCI 3156 09 2009 PMC 2846656 PMID 19793977 a b c d Roux Jean Christophe Panayotis Nicolas Dura Emmanuelle Villard Laurent 2009 Progressive noradrenergic deficits in the locus coeruleus of Mecp2 deficient mice Journal of Neuroscience Research 88 7 1500 9 doi 10 1002 jnr 22312 PMID 19998492 S2CID 3404695 Bjorklund A Lindvall O 1984 Dopamine containing systems in the CNS In Bjorklund A Hokfelt T eds Handbook of Chemical Neuroanatomy Classical Transmitters in the CNS Part l Vol 2 New York Elsevier pp 55 122 Hokfelt T Martensson R Bjorklund A Kleinau S Goldstein M 1984 Distribution maps of tyrosine hydroxylase immunoreactive neurons in the rat brain In Bjorklund A Hokfelt T eds Handbook of Chemical Neuroanatomy Classical Transmitters in the CNS Part I Vol 2 New York Elsevier pp 277 379 Bjorklund Anders Dunnett Stephen B 2007 Dopamine neuron systems in the brain An update Trends in Neurosciences 30 5 194 202 doi 10 1016 j tins 2007 03 006 PMID 17408759 S2CID 14239716 Parent Andre Hazrati Lili Naz 1995 Functional anatomy of the basal ganglia I The cortico basal ganglia thalamo cortical loop Brain Research Reviews 20 1 91 127 doi 10 1016 0165 0173 94 00007 C PMID 7711769 S2CID 28252990 Gerfen Charles R 2000 Molecular effects of dopamine on striatal projection pathways Trends in Neurosciences 23 10 Suppl S64 70 doi 10 1016 S1471 1931 00 00019 7 PMID 11052222 S2CID 3965480 Lees Andrew J Hardy John Revesz Tamas 2009 Parkinson s disease The Lancet 373 9680 2055 66 doi 10 1016 S0140 6736 09 60492 X PMID 19524782 S2CID 42608600 Dauer William Przedborski Serge 2003 Parkinson s Disease Neuron 39 6 889 909 doi 10 1016 S0896 6273 03 00568 3 PMID 12971891 S2CID 10400095 Jenner Peter 2009 Functional models of Parkinson s disease A valuable tool in the development of novel therapies Annals of Neurology 64 S16 29 doi 10 1002 ana 21489 PMID 19127585 S2CID 26065287 Fitzgerald Patricia M Jankovic Joseph Percy Alan K 1990 Rett syndrome and associated movement disorders Movement Disorders 5 3 195 202 doi 10 1002 mds 870050303 PMID 2388636 S2CID 43376602 Neul Jeffrey L Zoghbi Huda Y 2004 Rett Syndrome A Prototypical Neurodevelopmental Disorder The Neuroscientist 10 2 118 28 doi 10 1177 1073858403260995 PMID 15070486 S2CID 9617631 Segawa Masaya 2005 Early motor disturbances in Rett syndrome and its pathophysiological importance Brain and Development 27 S54 S58 doi 10 1016 j braindev 2004 11 010 PMID 16182486 S2CID 30218744 Guy Jacky Hendrich Brian Holmes Megan Martin Joanne E Bird Adrian 2001 A mouse Mecp2 null mutation causes neurological symptoms that mimic Rett syndrome Nature Genetics 27 3 322 6 doi 10 1038 85899 hdl 1842 727 PMID 11242117 S2CID 8698208 Chen Richard Z Akbarian Schahram Tudor Matthew Jaenisch Rudolf 2001 Deficiency of methyl CpG binding protein 2 in CNS neurons results in a Rett like phenotype in mice Nature Genetics 27 3 327 31 doi 10 1038 85906 PMID 11242118 S2CID 24979562 Nan X Ng H H Johnson C A Laherty C D Turner B M Eisenman R N Bird A 1998 Transcriptional repression by the methyl CpG binding protein MeCP2 involves a histone deacetylase complex Nature 393 6683 386 9 Bibcode 1998Natur 393 386N doi 10 1038 30764 PMID 9620804 S2CID 4427745 Cheval H Guy J Merusi C De Sousa D Selfridge J Bird A 2012 Postnatal inactivation reveals enhanced requirement for MeCP2 at distinct age windows Human Molecular Genetics 21 17 3806 14 doi 10 1093 hmg dds208 PMC 3412380 PMID 22653753 nbsp Ricceri Laura De Filippis Bianca Laviola Giovanni 2008 Mouse models of Rett syndrome From behavioural phenotyping to preclinical evaluation of new therapeutic approaches Behavioural Pharmacology 19 5 6 501 17 doi 10 1097 FBP 0b013e32830c3645 PMID 18690105 S2CID 33364486 Samaco R C Mandel Brehm C Chao H T Ward C S Fyffe Maricich S L Ren J Hyland K Thaller C Maricich S M Humphreys P Greer J J Percy A Glaze D G Zoghbi H Y Neul J L 2009 Loss of MeCP2 in aminergic neurons causes cell autonomous defects in neurotransmitter synthesis and specific behavioral abnormalities Proceedings of the National Academy of Sciences 106 51 21966 71 Bibcode 2009PNAS 10621966S doi 10 1073 pnas 0912257106 JSTOR 40536204 PMC 2799790 PMID 20007372 a b c d Panayotis Nicolas Pratte Michel Borges Correia Ana Ghata Adeline Villard Laurent Roux Jean Christophe 2011 Morphological and functional alterations in the substantia nigra pars compacta of the Mecp2 null mouse Neurobiology of Disease 41 2 385 97 doi 10 1016 j nbd 2010 10 006 PMID 20951208 S2CID 25414717 Sun Yi E Wu Hao 2006 The Ups and Downs of BDNF in Rett Syndrome Neuron 49 3 321 3 doi 10 1016 j neuron 2006 01 014 PMID 16446133 a b Sharifi Osman Yasui Dag H 2021 The Molecular Functions of MeCP2 in Rett Syndrome Pathology Frontiers in Genetics 12 624290 doi 10 3389 fgene 2021 624290 PMC 8102816 PMID 33968128 Ehrhart Friederike Coort Susan L M Cirillo Elisa Smeets Eric Evelo Chris T Curfs Leopold M G 25 November 2016 Rett syndrome biological pathways leading from MECP2 to disorder phenotypes Orphanet Journal of Rare Diseases 11 1 158 doi 10 1186 s13023 016 0545 5 PMC 5123333 PMID 27884167 Tsai Luke Y 1992 Is Rett syndrome a subtype of pervasive developmental disorders PDF Journal of Autism and Developmental Disorders 22 4 551 61 doi 10 1007 BF01046327 hdl 2027 42 44607 PMID 1483976 S2CID 17817425 Archived from the original on 29 August 2021 Retrieved 20 April 2018 Abbeduto Leonard Ozonoff Susan Thurman Angela John McDuffie Angela Schweitzer Julie 18 March 2014 Hales Robert Yudofsky Stuart Robert Laura Weiss eds Chapter 8 Neurodevelopmental Disorders The American Psychiatric Publishing Textbook of Psychiatry 6 ed Arlington VA American Psychiatric Publishing doi 10 1176 appi books 9781585625031 rh08 ISBN 978 1 58562 444 7 S2CID 241966275 a b Rett syndrome Tests and diagnosis Mayo Clinic Archived from the original on 30 October 2017 a b c d e f About Rett syndrome Rett Syndrome Diagnosis rettsyndrome org International Rett Syndrome Foundation Archived from the original on 29 October 2017 Retrieved 10 May 2020 McCauley Mark D Wang Tiannan Mike Elise Herrera Jose Beavers David L Huang Teng Wei Ward Christopher S Skinner Steven Percy Alan K 14 December 2011 Pathogenesis of Lethal Cardiac Arrhythmias in Mecp2 Mutant Mice Implication for Therapy in Rett Syndrome Science Translational Medicine 3 113 113ra125 doi 10 1126 scitranslmed 3002982 ISSN 1946 6234 PMC 3633081 PMID 22174313 a b Mackay Jessica Downs Jenny Wong Kingsley Heyworth Jane Epstein Amy Leonard Helen 2017 Autonomic breathing abnormalities in Rett syndrome caregiver perspectives in an international database study Journal of Neurodevelopmental Disorders 9 15 doi 10 1186 s11689 017 9196 7 ISSN 1866 1947 PMC 5410057 PMID 28465761 Harish Kumar S 19 January 2017 Cardio respiratory physiotherapy management in Rett s syndrome Literature review International Journal of Pharma and Bio Sciences 5 via ResearchGate Kyle Stephanie M Vashi Neeti Justice Monica J February 2018 Rett syndrome a neurological disorder with metabolic components Open Biology 8 2 170216 doi 10 1098 rsob 170216 ISSN 2046 2441 PMC 5830535 PMID 29445033 De Felice Claudio Maffei Silvia Signorini Cinzia Leoncini Silvia Lunghetti Stefano Valacchi Giuseppe D Esposito Maurizio Filosa Stefania Della Ragione Floriana Butera Gianfranco Favilli Roberto April 2012 Subclinical myocardial dysfunction in Rett syndrome European Heart Journal Cardiovascular Imaging 13 4 339 345 doi 10 1093 ejechocard jer256 ISSN 2047 2412 PMID 22113206 Archived from the original on 29 November 2021 Retrieved 29 November 2021 a b Smeets Eric E J Julu Peter O O Waardenburg Dick van Engerstrom Ingegerd Witt Hansen Stig Apartopoulos Flora Curfs Leopold M G Schrander Stumpel Connie T R M 1 November 2006 Management of a severe forceful breather with Rett Syndrome using carbogen Brain and Development 28 10 625 632 doi 10 1016 j braindev 2006 04 010 ISSN 0387 7604 PMID 16765005 S2CID 15545729 Archived from the original on 1 October 2022 Retrieved 29 November 2021 Positive top line results from pivotal Phase 3 trial in Rett syndrome PDF Report Australian Stock Exchange 7 December 2021 Rett Syndrome New Drug Application accepted for Priority Review by FDA PDF Report Australian Stock Exchange 13 September 2022 Breakthrough drug from Dame Margaret Brimble s lab University of Auckland 11 March 2023 Retrieved 6 May 2023 Schwartzman J S Bernardino Andrea Nishimura Agnes Gomes Raquel R Zatz Mayana 2001 Rett Syndrome in a Boy with a 47 XXY Karyotype Confirmed by a Rare Mutation in the MECP2 Gene Neuropediatrics 32 3 162 4 doi 10 1055 s 2001 16620 PMID 11521215 S2CID 260240039 a b Hardwick Simon A Reuter Kirsten Williamson Sarah L Vasudevan Vidya Donald Jennifer Slater Katrina Bennetts Bruce Bebbington Ami Leonard Helen Williams Simon R Smith Robert L Cloosterman Desiree Christodoulou John 2007 Delineation of large deletions of the MECP2 gene in Rett syndrome patients including a familial case with a male proband European Journal of Human Genetics 15 12 1218 29 doi 10 1038 sj ejhg 5201911 PMID 17712354 Acampa M Guideri F May 2006 Cardiac disease and Rett syndrome Archives of Disease in Childhood 91 5 440 443 doi 10 1136 adc 2005 090290 ISSN 1468 2044 PMC 2082747 PMID 16632674 Rett A 10 September 1966 On an unusual brain atrophy syndrome in hyperammonemia in childhood Wiener Medizinische Wochenschrift in German 116 37 723 726 ISSN 0043 5341 PMID 5300597 Taysha Gene Therapies Announces Initiation of Clinical Development of TSHA 102 in Rett Syndrome Taysha Gene Therapies March 2022 Archived from the original on 30 March 2022 Retrieved 6 May 2022 title Head of Zeus Archived from the original on 1 October 2022 Retrieved 7 May 2022 Retrieved from https en wikipedia org w index php title Rett syndrome amp oldid 1188870890, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.