22q13 deletion syndrome: Difference between revisions

From Wikipedia, the free encyclopedia
Content deleted Content added
m WP:CHECKWIKI error fixes using AWB (12061)
The content of the page as it was not accurate with the current scientific data and medical consensus of the disorder.
Tags: nowiki added Visual edit
Line 16: Line 16:
|GeneReviewsNBK=NBK1198
|GeneReviewsNBK=NBK1198
}}
}}
Phelan-McDermid syndrome (PMS), also called 22q13 deletion syndrome, is a rare genetic condition characterized by global developmental delay, intellectual disability (ID), absent or delayed speech, autism spectrum disorder (ASD), hypotonia and mild dysmorphic features<ref>{{Cite journal|last=Soorya|first=Latha|last2=Kolevzon|first2=Alexander|last3=Zweifach|first3=Jessica|last4=Lim|first4=Teresa|last5=Dobry|first5=Yuriy|last6=Schwartz|first6=Lily|last7=Frank|first7=Yitzchak|last8=Wang|first8=A Ting|last9=Cai|first9=Guiqing|date=2013-06-11|title=Prospective investigation of autism and genotype-phenotype correlations in 22q13 deletion syndrome and SHANK3 deficiency|url=http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3707861/|journal=Molecular Autism|volume=4|pages=18|doi=10.1186/2040-2392-4-18|issn=2040-2392|pmc=3707861|pmid=23758760}}</ref><ref>{{Cite journal|last=Phelan|first=K.|last2=McDermid|first2=H.E.|title=The 22q13.3 Deletion Syndrome (Phelan-McDermid Syndrome)|url=http://www.karger.com/doi/10.1159/000334260|journal=Molecular Syndromology|doi=10.1159/000334260|pmc=3366702|pmid=22670140}}</ref><ref>{{Cite journal|last=Kolevzon|first=Alexander|last2=Angarita|first2=Benjamin|last3=Bush|first3=Lauren|last4=Wang|first4=A. Ting|last5=Frank|first5=Yitzchak|last6=Yang|first6=Amy|last7=Rapaport|first7=Robert|last8=Saland|first8=Jeffrey|last9=Srivastava|first9=Shubhika|date=2014-01-01|title=Phelan-McDermid syndrome: a review of the literature and practice parameters for medical assessment and monitoring|url=https://www.ncbi.nlm.nih.gov/pubmed/25784960|journal=Journal of Neurodevelopmental Disorders|volume=6|issue=1|pages=39|doi=10.1186/1866-1955-6-39|issn=1866-1947|pmc=4362650|pmid=25784960}}</ref>.   Affected individuals present in addition with an array of medical, neurological and behavioral symptoms (tables 1 and 2)<ref>{{Cite journal|last=Phelan|first=M. C.|last2=Rogers|first2=R. C.|last3=Saul|first3=R. A.|last4=Stapleton|first4=G. A.|last5=Sweet|first5=K.|last6=McDermid|first6=H.|last7=Shaw|first7=S. R.|last8=Claytor|first8=J.|last9=Willis|first9=J.|date=2001-06-15|title=22q13 deletion syndrome|url=https://www.ncbi.nlm.nih.gov/pubmed/11391650|journal=American Journal of Medical Genetics|volume=101|issue=2|pages=91–99|issn=0148-7299|pmid=11391650}}</ref><sup>,</sup><ref>{{Cite journal|last=Sarasua|first=Sara M.|last2=Boccuto|first2=Luigi|last3=Sharp|first3=Julia L.|last4=Dwivedi|first4=Alka|last5=Chen|first5=Chin-Fu|last6=Rollins|first6=Jonathan D.|last7=Rogers|first7=R. Curtis|last8=Phelan|first8=Katy|last9=DuPont|first9=Barbara R.|date=2014-07-01|title=Clinical and genomic evaluation of 201 patients with Phelan-McDermid syndrome|url=http://www.ncbi.nlm.nih.gov/pubmed/24481935|journal=Human Genetics|volume=133|issue=7|pages=847–859|doi=10.1007/s00439-014-1423-7|issn=1432-1203|pmid=24481935}}</ref>.  The core clinical and neurobehavioral symptoms in PMS syndrome are caused by the loss of one functional copy of the ''SHANK3'' gene (haploinsufficiency)<sup>1</sup>, which is located in the distal segment of the long arm of chromosome 22<ref>{{Cite journal|last=Costales|first=Jesse|last2=Kolevzon|first2=Alexander|date=2016-04-01|title=The therapeutic potential of insulin-like growth factor-1 in central nervous system disorders|url=http://www.sciencedirect.com/science/article/pii/S0149763415300452|journal=Neuroscience & Biobehavioral Reviews|volume=63|pages=207–222|doi=10.1016/j.neubiorev.2016.01.001|pmc=4790729|pmid=26780584}}</ref> (figure 1).   The ''SHANK3'' gene encodes a protein that is critical for neuronal communication and synapse formation<ref>{{Cite journal|last=Harony-Nicolas|first=Hala|last2=Rubeis|first2=Silvia De|last3=Kolevzon|first3=Alexander|last4=Buxbaum|first4=Joseph D.|date=2015-12-01|title=Phelan McDermid Syndrome From Genetic Discoveries to Animal Models and Treatment|url=http://jcn.sagepub.com/content/30/14/1861|journal=Journal of Child Neurology|language=en|volume=30|issue=14|pages=1861–1870|doi=10.1177/0883073815600872|issn=0883-0738|pmid=26350728}}</ref><ref>{{Cite journal|last=Carbonetto|first=Salvatore|date=2014-02-01|title=A blueprint for research on Shankopathies: a view from research on autism spectrum disorder|url=http://www.ncbi.nlm.nih.gov/pubmed/24218108|journal=Developmental Neurobiology|volume=74|issue=2|pages=85–112|doi=10.1002/dneu.22150|issn=1932-846X|pmid=24218108}}</ref>.    About 80% of cases result from a spontaneous, or ''de novo'' mutation, and is not inherited from parents.
'''22q13 deletion syndrome''' (spoken as ''twenty-two q one three''<ref>Technically, it should be spoken as ''twenty-two q one three''</ref>) is a [[genetic disorder]] caused by deletions or rearrangements on the q terminal end (long arm) of [[chromosome 22]]. Any abnormal genetic variation in the q13 region that presents with significant manifestations ([[phenotype]]) typical of a terminal deletion should be diagnosed as 22q13 deletion syndrome. 22q13 deletion syndrome is often placed in the more general category of '''Phelan-McDermid Syndrome''' (abbreviated '''PMS'''), which includes some mutations and microdeletions. The PMS name is less precise, since there is disagreement among researchers as to which variants belong in the PMS category.<ref>{{cite journal |vauthors=Phelan K, Boccuto L, Rogers RC, Sarasua SM, McDermid HE |title=Letter to the editor regarding Disciglio et al.: interstitial 22q13 deletions not involving SHANK3 gene: a new contiguous gene syndrome. |journal=Am J Med Genet A |volume=167 |issue=7 |pages=1679–80 |year=2015 |pmid= 26295085 |doi= 10.1002/ajmg.a.36788 }}</ref> The Developmental Synaptopathies Consortium defines PMS as being caused by [[SHANK3]] mutations, a definition that excludes terminal deletions.<ref>https://www.rarediseasesnetwork.org/cms/dsc/About-Us (downloaded 21-September-2015)</ref> This latter definition of PMS is incompatible with the definition of 22q13 deletion syndrome by those who first described 22q13 deletion syndrome.<ref name="Phelan MC, McDermid HE 2011 186–201">{{cite journal |vauthors=Phelan MC, McDermid HE |title=The 22q13.3 Deletion Syndrome (Phelan-McDermid Syndrome). |journal=Mol Syndromol. |volume=2 |issue=1 |pages=186–201 |year=2011 |pmid= 22670140|doi= 10.1159/000334260 |pmc=3366702}}</ref>


'''History'''
A prototypical terminal deletion of 22q13 can be uncovered by [[karyotype]] analysis, but many terminal and interstitial deletions are too small. The availability of [[DNA microarray]] technology for revealing multiple genetic problems simultaneously has been the diagnostic tool of choice. The falling cost for whole [[exome sequencing]] and, eventually, [[whole genome sequencing]], may replace [[DNA microarray]] technology for candidate evaluation. However, [[fluorescence in situ hybridization]] (FISH) tests remain valuable for diagnosing cases of mosaicism ([[mosaic]] genetics) and chromosomal rearrangements (e.g., [[ring chromosome]], unbalanced [[chromosomal translocation]]). Although early researchers sought a monogenic (single gene [[genetic disorder]]) explanation, recent studies have not supported that hypothesis (see Etiology, below).


The first case of PMS was described in 1985 by Watt et al., who described a 14 year old boy with severe intellectual disability, mild dysmorphic features and absent speech, which was associated with terminal loss the distal arm of chromosome 22(9).  In 1988, Phelan et al. described a similar clinical presentation associated with a ''de novo'' deletion in 22q13.3(4).  Subsequent cases where described in the following years with a similar clinical presentation.  Phelan et al (2001)(4), compared 37 subjects with 22q13 deletions with features of 24 cases described in the literature finding that the most common features were global developmental delay, absent or delayed speech and hypotonia.  In 2001, Bonaglia et al.(10), described a case that associated the 22q.13 deletion syndrome with a disruption of the ''SHANK3'' gene (also called ''ProSAP2'').  The following year, Anderlid et al. (2002)(11), refined the area in 22q13 presumably responsible for the common phenotypic presentation of the syndrome to a 100kb  in 22q13.3 (image).  Out of the three genes affected, ''SHANK3'' was identified as the critical gene due to its expression pattern and function.  Wilson et al. (2003)(12) evaluated 56 patients with the  clinical presentation of PMS, all of whom had a functional loss of one copy of the ''SHANK3'' gene, further confirming the role of ''SHANK3'' in the major neuropsychiatric manifestations of the syndrome.  Since then multiple other studies have reported similar findings with smaller deletions of the region or mutations of the gene(13).
22q13 deletion syndrome is characterized by global developmental delay, absent or severely delayed speech, and neonatal [[hypotonia]].<ref name="Phelan MC, McDermid HE 2011 186–201"/> There are approximately 1300 diagnosed cases of 22q13 deletion syndrome worldwide.


'''Epidemiology'''
==Characteristics==
The core characteristics of 22q13 Deletion syndrome (listed above) have a major impact on the individual. However, in addition to these characteristics, there are other manifestations than may range from mild to severe:


The true prevalence of PMS has not been determined. More than 1200 people have been identified worldwide according the Phelan-McDermid Syndrome Foundation. However, it is believed to be underdiagnosed due to inadequate genetic testing and lack of specific clinical features. It is known to occur with equal frequency in males and females(7).   Studies using chromosomal microarray for diagnosis indicate that at least 0.5% of cases of ASD can be explained by mutations or deletions in the ''SHANK3'' gene(14). In addition when ASD is associated with ID, ''SHANK3'' mutations or deletions have been found in up to 2% of individuals(15,16).  The rate of ''SHANK3'' insufficiency in ID and ASD is still being determined; however, these findings imply that it is one of the most common single gene causes.
'''Physical'''
* Absent to severely delayed speech: 99%
* Normal to accelerated growth: 95%
* High tolerance to pain: 77%
* [[Hypotonia]] (poor muscle tone): 75%
* Dysplastic toenails: 73%
* Long eyelashes: 73%
* Poor [[thermoregulation]]: 68%
* Prominent, poorly formed ears: 65%
* Large or fleshy hands: 63%
* Pointed chin: 62%
* [[Dolichocephaly]] (elongated head): 57%
* [[Ptosis (eyelid)]] (droopy eyelids): 57%
* [[Gastroesophageal reflux]]: 42%
* [[Epileptic seizure]]s: 27%
* Kidney problems: 26%
* Delayed ability to walk: 18%


'''Behavioral'''
'''Etiology'''
* Chewing on non food items: 85%
* Delayed or unreliable toileting: 76%
* Impulsive behaviors: 47%
* Biting (self or others): 46%
* Problems sleeping: 46%
* Hair pulling: 41%
* Autistic behaviors: 31%
* Episodes of non-stop crying before age 5: 30%
* Teeth grinding: (unknown) %


The loss of one copy of ''SHANK3'' can result from chromosomal abnormalities in the terminal end of chromosome 22 or from a mutation in ''SHANK3'' leading to a loss of function in the protein.  The ''SHANK3'' gene codes for a protein (SHANK3) that is vital for synaptic formation and plasticity(17,18) (figure 2). It is expressed widely in the brain early in the postnatal period and its expression peaks during synaptic formation and maturation(13).   The protein is found in the hippocampus, the striatum, cerebellar granular cells, olfactory bulb and the thalamic nuclei(13,19).  SHANK proteins are critical for the architectural make-up of the dendritic spines and their expression is essential for cognition and learning(17).  SHANK3 is a scaffolding protein located in the postsynaptic density of excitatory synapses (figure 2)(7,13).  Genetic manipulation of ''Shank3'' in animal model systems leads to changes in the key components of glutamatergic synapse and dendritic spine density.
==Etiology==
Various deletions affect the terminal region of the long arm of chromosome 22 (the [[paternal]] chromosome in 75% of cases), from 22q13.3 to 22qter. Although the deletion is most typically a result of a de novo mutation, there is an inherited form resulting from familial [[chromosomal translocation]]s involving the 22 chromosome. In the de novo form, the size of the terminal deletion is variable and can go from 130 Kb (130,000 [[base pairs]]) to 9 Mb. Deletions smaller than 1 Mb are very rare (about 3%). The remaining 97% of terminal deletions impact about 30 to 190 genes (see list, below). At one time it was thought that deletion size was not related to the core clinical features.<ref name="Wilson HL, Wong AC, Shaw SR, et al. 2003 575–84">{{cite journal |vauthors=Wilson HL, Wong AC, Shaw SR |title=Molecular characterisation of the 22q13 deletion syndrome supports the role of haploinsufficiency of SHANK3/PROSAP2 in the major neurological symptoms |journal=J. Med. Genet. |volume=40 |issue=8 |pages=575–84 |year=2003 |pmid=12920066 |pmc=1735560 |doi= 10.1136/jmg.40.8.575 | url=http://jmg.bmj.com/cgi/pmidlookup?view=long&pmid=12920066|display-authors=etal}}</ref> That observation lead to an emphasis on the [[SHANK3]] gene, which resides close to the terminal end of chromosome 22. Interest in [[SHANK3]] grew as it became associated with [[Autism Spectrum Disorder]] (ASD) and [[Schizophrenia]].<ref>{{cite journal |author = Gauthier|title=De novo mutations in the gene encoding the synaptic scaffolding protein SHANK3 in patients ascertained for schizophrenia.| journal= Proc. Natl. Acad. Sci. USA | volume=107| issue=17 | pages=7863–8 | year=2010 | pmid= 20385823| doi= 10.1073/pnas.0906232107 | pmc=2867875|display-authors=etal}}</ref> Since then, twelve other genes on 22q13 (MAPK8IP2,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=MAPK8IP2&keywords=mapk8ip2</ref> CHKB,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=CHKB&keywords=CHKB</ref> SCO2,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=SCO2&keywords=sco2</ref> SBF1,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=SBF1&keywords=SBF1</ref> PLXNB2,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=PLXNB2&keywords=plxnb2</ref> MAPK12,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=MAPK12&keywords=mapk12</ref> PANX2,<ref>http://www.genecards.org/Search/Keyword?queryString=panx2</ref> BRD1,<ref>http://www.genecards.org/Search/Keyword?queryString=brd1</ref> CELSR1,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=CELSR1&keywords=CELSR1</ref> WNT7B,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=WNT7B&keywords=wnt7b</ref> TCF20<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=TCF20&keywords=tcf20</ref>) have been associated with [[Autism Spectrum Disorder]] and/or [[Schizophrenia]] (see references below). Some mutations of [[SHANK3]] mimic 22q13 deletion syndrome, but SHANK3 mutations and microdeletions have quite variable impact. Most mutations do not reflect loss of the entire gene.<ref>{{ cite journal | author= Wang X | title= Transcriptional and functional complexity of Shank3 provides a molecular framework to understand the phenotypic heterogeneity of SHANK3 causing autism and Shank3 mutant mice. | journal= [[Mol Autism]] | volume= 5| issue= 30| year= 2014 | pmid= 25071925| doi= 10.1186/2040-2392-5-30 |display-authors=etal}}</ref><ref>{{ cite journal | author= Zhou Y | title= Mice with Shank3 Mutations Associated with ASD and Schizophrenia Display Both Shared and Distinct Defects. | journal= [[Neuron (journal)|Neuron]] | volume= 89| issue= 1| year= 2016 | pmid= 26687841| doi= 10.1016/j.neuron.2015.11.023 |display-authors=etal | pages=147–62}}</ref><ref>{{ cite journal | author= Jaramillo TC | title= Altered Striatal Synaptic Function and Abnormal Behaviour in Shank3 Exon4-9 Deletion Mouse Model of Autism. | journal= [[Autism Res.]] | year= 2015 | pmid= 26626443 | doi= 10.1002/jcph.679 |display-authors=etal | volume=56 | pages=885–93}}</ref><ref>{{cite journal |author = Halbedl S|title=Shank3 is localized in axons and presynaptic specializations of developing hippocampal neurons and involved in the modulation of NMDA receptor levels at axon terminals.| journal= J Neurochem | issue= [Epub ahead of print]| year=2016 | pmid= 26725465| doi= 10.1111/jnc.13523 | pmc=|display-authors=etal | volume=137 | pages=26–32}}</ref> This has been demonstrated in mice, where deletion of the entire SHANK3 gene produces a weaker phenotype than many mutations and complete absence of SHANK3 (complete null) has less impact than some heterozygous mutations.<ref>{{cite journal |author = Wang X|title=Altered mGluR5-Homer scaffolds and corticostriatal connectivity in a Shank3 complete knockout model of autism.| journal= Nat Commun | volume= 7 | year=2016 | pmid= 27161151 | doi= 10.1038/ncomms11459 | display-authors=etal | pmc=4866051 | pages=11459}}</ref>


Chromosomal abnormalities leading to the loss of the terminal arm of chromosome 22 include unbalanced chromosomal translocations, terminal or interstitial deletions involving the 22q13.3 region, a ring chromosome, or other structural changes (figure 3). Terminal deletions account for about 75% of the cases of PMS(7).  Some studies suggest a relationship between deletion size and the extent or severity of certain clinical features, but this has not been well established and remains the focus of clinical research(1,20–24).    
Some of the core features of 22q13 deletion syndrome are dependent upon deletion size, and do not depend on the loss of [[SHANK3]].<ref>{{cite journal |author = Sarasua SM|title=Association between deletion size and important phenotypes expands the genomic region of interest in Phelan-McDermid syndrome (22q13 deletion syndrome). | journal=J Med Genet. | volume=48 | issue=11 | pages=761–6 | year=2011 | pmid=21984749 | doi= 10.1136/jmedgenet-2011-100225 |display-authors=etal}}</ref><ref>{{cite journal |author = Simenson K|title= A patient with the classic features of Phelan-McDermid syndrome and a high immunoglobulin E level caused by a cryptic interstitial 0.72-Mb deletion in the 22q13.2 region. |journal=Am J Med Genet A |year=2013 |pmid=24375995|doi= 10.1002/ajmg.a.36358 |volume=164A |issue=3 |pages=806–9|display-authors=etal}}</ref><ref name="Disciglio V et al. 2014 1666–76">{{cite journal |author = Disciglio V|title= Interstitial 22q13 Deletions Not Involving SHANK3 Gene: A New Contiguous Gene Syndrome. |journal=Am J Med Genet A |year=2014 | pmid=24700646 | doi= 10.1002/ajmg.a.36513 |volume=164 |issue=7 |pages=1666–76|display-authors=etal}}</ref> As noted above, the distal 1 Mb of 22q is a gene rich region. There are too few clinical cases to statistically measure the relationship between deletion size and phenotype in this region. A landmark study of [[induced pluripotent stem cell]] neurons cultured from patients with 22q13 deletion syndrome shows that restoration of the [[SHANK3]] protein levels can rescue fewer than half the glutamate neurons of neocortex, another indication of the strong impact of other genes in the distal 1 Mb of chromosome 22.<ref>{{ cite journal | author= Shcheglovitov A | title= SHANK3 and IGF1 restore synaptic deficits in neurons from 22q13 deletion syndrome patients. | journal= [[Nature (journal)|Nature]] | volume= 503 | issue= 7475| pages= 267–71 | year= 2013 | pmid= 24132240| doi= 10.1038/nature12618 |display-authors=etal}}</ref>


'''Clinical Description'''
There is an interest in the impact of [[MAPK8IP2]] (also called IB2) in 22q13 deletion syndrome.<ref>{{ cite journal | author= Aldinger KA | title= Cerebellar and posterior fossa malformations in patients with autism-associated chromosome 22q13 terminal deletion. | journal= [[Am J Med Genet A]] | volume= 161 | issue= 1| pages= 131–6 | year= 2013 | pmid= 23225497 | doi= 10.1002/ajmg.a.35700 |display-authors=etal}}</ref> MAPK8IP2 is especially interesting because it regulates the balance between [[NMDA receptor]]s and [[AMPA receptor]]s.<ref>{{ cite journal | author= Giza J | title= Behavioral and cerebellar transmission deficits in mice lacking the autism-linked gene islet brain-2. | journal= J Neurosci. | volume= 30| issue= 44| pages= 14805–16 | year= 2010 | pmid= 21048139 | doi= 10.1523/JNEUROSCI.1161-10.2010 | pmc=3200367|display-authors=etal}}</ref> The genes SULT4A1<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=SULT4A1&search=a909593f05863155b816a8fb7654c03b</ref> and PARVB<ref name="genecards.org">http://www.genecards.org/cgi-bin/carddisp.pl?gene=PARVB&search=6f331a34c3511163f07d03211274ad96</ref> may cause 22q13 deletion syndrome in cases of more proximal interstitial and large terminal deletions.<ref name="Disciglio V et al. 2014 1666–76"/> There are about 187 protein coding genes in the 22q13 region.<ref>http://genome.ucsc.edu/</ref> A group of genes (MPPED1,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=MPPED1&search=af0348b2e8f8bbe07815c7c4c35e1f8e</ref> CYB5R3,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=CYB5R3&search=f30516afb414af5d738f38bfdee0a8b4</ref> FBLN1,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=FBLN1&search=31c50040405215fff62221f468762f78</ref> NUP50,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=NUP50&search=ebf7ec6b4ee48d75243c7b448aa489a8</ref> C22orf9,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=KIAA0930&search=7fe64f97e1b1ff3046fce6978ce05ceb</ref> KIAA1644,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=KIAA1644&search=7699c1245c9f84709a4902cb2643f900</ref> PARVB,<ref name="genecards.org"/> TRMU,<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=TRMU&search=dfaeaec9ab390a77b7713cddf9e0d842</ref> WNT7B<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=WNT7B&search=b9a837acec2f26b76076ecd2d3887361</ref> and ATXN10<ref>http://www.genecards.org/cgi-bin/carddisp.pl?gene=ATXN10&search=8085643553fd43eaabcf7fac1618ef13</ref>), as well as [[microRNA]]s may all contribute to loss of language, a feature that varies notably with deletion size.<ref>{{ cite journal | author= Sarasua SM | title= Clinical and genomic evaluation of 201 patients with Phelan–McDermid syndrome. |journal= [[Human Genetics (journal)|Human Genetics]] | year= 2014 | pmid= 24481935 | doi=10.1007/s00439-014-1423-7 | volume=133 | issue=7 | pages=847–59|display-authors=etal}}</ref> The same study found that [[macrocephaly]] seen in 22q13 deletion syndrome patients may be associated with WNT7B.


Affected individuals present with a broad array of medical and behavioral manifestations (tables 1 and 2).  Patients are consistently characterized by global developmental delay, intellectual disability, speech abnormalities, ASD, hypotonia and mild dysmorphic features.    Table 1 summarizes the dysmorphic and medical conditions that have been reported in individuals with PMS.  Table 2 summarize the psychiatric and neurological associated with SHANK3 deficiency and PMS (1,4,21,23–33).  Most of the studies include small samples or relied on parental report or medical record review to collect information, which can account in part for the variability in the presentation of some of the presenting features.  Larger prospective studies are needed to further characterize the disorder genetic. It is likely that as the awareness, of PMS and the role of SHANK3 deficiency in ASD and ID, increases among the medical community more individuals will be identified which will provide a better understanding of the disorder.
Table of protein coding genes involved in 22q13 deletion syndrome (based on Human Genome Browser – hg38 assembly <ref>https://genome.ucsc.edu/index.html</ref>). Underline identifies 13 genes that are associated with autism.<ref>https://gene.sfari.org/autdb/HG_Home.do</ref><ref>{{ cite journal | author= Napoli E | title= Mitochondrial Dysfunction in Pten Haplo-Insufficient Mice with Social Deficits and Repetitive Behavior: Interplay between Pten and p53. |journal= [[PLoS One (journal)|PLoS One]] | year= 2012 | pmid= 22900024 | doi=10.1371/journal.pone.0042504 | volume=7 | issue=8 | pages=1–13|display-authors=etal | pmc=3416855}}</ref><ref>{{ cite journal | author= Iossifov I | title= Low load for disruptive mutations in autism genes and their biased transmission. |journal= [[Proc Natl Acad Sci U S A (journal)|Proc Natl Acad Sci U S A]] | year= 2015 | pmid= 26401017 | doi=10.1073/pnas.1516376112 | volume=112| issue=41 | pages=E5600-7|display-authors=etal | pmc=4611648}}</ref><ref>{{ cite journal | author= Davis LK | title= Loci nominally associated with autism from genome-wide analysis show enrichment of brain expression quantitative trait loci but not lymphoblastoid cell line expression quantitative trait loci. |journal= [[Mol Autism (journal)|Mol Autism]] | year= 2012 | pmid= 22591576 | doi=10.1186/2040-2392-3-3 | volume=3 | issue=1 | display-authors=etal | pmc=3484025 | pages=3}}</ref> Bold identifies genes associated with hypotonia (based on Human Phenotype Browser <ref>http://www.human-phenotype-ontology.org/</ref> search for 'hypotonia' and the OMIM database <ref>http://www.omim.org/</ref>).


{| class="wikitable"
{| class="wikitable"
|Dysmorphic Feature
|Percentage (%)
|Medical Comorbidities
|Percentage (%)
|-
|-
|Macrocephaly
|| RABL2B
|7-31
|| ACR
|Hypothyroidism
|| '''<u>SHANK3</u>'''
|3-6
|| '''ARSA'''
|-
|| <u>MAPK8IP2</u>
|Microcephaly
|| '''<u>CHKB</u>'''
|11-14
|| CPT1B
|Sleep disturbance
|| SYCE3
|41-46
|| KLHDC7B
|-
|| ODF3B
|Dolichocephaly
|| TYMP
|23-86
|| '''<u>SCO2</u>'''
|Gastroesophageal reflux
|-
|42-44
|| NCAPH2
|-
|| LMF2
|Long eyelashes
|| MIOX
|43-93
|| ADM2
|Increased pain threshold
|| <u>SBF1</u>
|10-88
|| PPP6R2
|-
|| DENND6B
|Bulbous nose
|| <u>PLXNB2</u>
|47-80
|| MAPK11
|Constipation/diarrhea
|| <u>MAPK12</u>
|38-41
|| HDAC10
|-
|| TUBGCP6
|High arched palate
|-
|25-47
|| SELO
|Brain imaging abnormalities
|| TRABD
|7-75
|| <u>PANX2</u>
|-
|| MOV10L1
|Malocclusion/widely spaced teeth
|| MLC1
|19
|| IL17REL
|Recurrent upper respiratory infections
|| PIM3
|8-53
|| CRELD2
|-
|| '''ALG12'''
|Full cheeks
|| ZBED4
|25
|| <u>BRD1</u>
|Renal abnormalities
|| FAM19A5
|-
|17-26
|-
|| FLJ32756
|Pointed chin
|| TBC1D22A
|22-62
|| CERK
|Lymphedema
|| GRAMD4
|8-53
|| <u>CELSR1</u>
|-
|| '''TRMU'''
|Large fleshy hands
|| BC069212
|33-68
|| GTSE1
|Seizures
|| TTC38
|14-41
|| PKDREJ
|-
|| CDPF1
|Hypoplastic/dysplastic nails
|| PPARA
|-
|3-78
|Strabismus
|| <u>WNT7B</u>
|6-26
|| ATXN10
|-
|| FBLN1
|Hyper-extensibility
|| RIBC2
|25-61
|| SMC1B
|Short stature
|| FAM118A
|11-13
|| UPK3A
|-
|| KIAA0930
|Abnormal spine curvature
|| NUP50
|22
|| PHF21B
|Tall stature/accelerated growth
|| PRR5-ARHGAP8
|3-18
|| LDOC1L
|-
|-
|Sacral dimple
|| KIAA1644
|13-37
|| PARVG
|Cardiac defects
|| TRNA_SeC
|3-25
|| PARVB
|-
|| SAMM50
|Syndactyly of toes 2 and 3
|| PNPLA3
|9-48
|| PNPLA5
|Precocious or delayed puberty
|| '''<u>SULT4A1</u>'''
|12
|| EFCAB6
|}
|| MPPED1
Table 1: Dysmorphic features and medical comorbid conditions that have been reported in individuals with Phelan McDermid Syndrome.
|| SCUBE1

|| TTLL12
{| class="wikitable"
|-
|'''Psychiatric and Neurologic Manifestations'''
|| TSPO
|'''Percentage (%)'''
|| MCAT
|-
|| BIK
|'''Autism Spectrum disorder'''
|| TTLL1
|>50
|| PACSIN2
|-
|| ARFGAP3
|'''Intellectual Disability'''
|| A4GALT
|>75
|| ATP5L2
|-
|| DL490307
|'''Global Developmental Delay'''
|| CYB5R3
|>75
|| RNU12
|-
|| POLDIP3
|'''Absent or severely affected speech'''
|-
|>75
|| SERHL2
|-
|| RRP7A
|'''Sensory seeking behaviors (mouthing of objects)'''
|| NFAM1
|>25
|| <u>TCF20</u>
|-
|| CYP2D6
|'''Teeth grinding'''
|| NDUFA6
|>25
|| SMDT1
|-
|| FAM109B
|'''Hyperactivity and inattention'''
|| '''NAGA'''
|>50
|| WBP2NL
|-
|| CENPM
|'''Stereotypical movements'''
|| TNFRSF13C
|>50
|-
|-
|| SHISA8
|'''Hypotonia'''
|| SREBF2
|>50
|| CCDC134
|-
|| MEI1
|'''Fine and gross motor abnormalities'''
|| C22orf46
|>90
|| NHP2L1
|-
|| XRCC6
|'''Poor fine motor coordination'''
|| DESI1
|>90
|| PMM1
|-
|| CSDC2
|'''Gait Abnormalities'''
|| POLR3H
|>90
|| '''ACO2'''
|-
|-
| '''Visual tracking abnormalities'''
|| PHF5A
|>85
|| TOB2
|-
|| TEF
|'''Seizure disorder'''
|| ZC3H7B
|17-41
|| RANGAP1
|-
|| CHADL
|'''Brain structural abnormalities'''
|| L3MBTL2
|44-100
|| EP300
|-
|| RBX1
|'''Sleep problems'''
|| DNAJB7
|>40
|| XPNPEP3
|}
|| ST13
Table 2: Psychiatric and Neurologic Manifestations associated with Phelan McDermid Syndrome
|-

|| SLC25A17
'''1.     Diagnosis and Management'''
|| MCHR1

|| MKL1
a.       Clinical Genetics and Genetic Testing
|| SGSM3

|| '''ADSL'''
Genetic testing is necessary to confirm ''SHANK3'' deficiency and the diagnosis of PMS.  A prototypical terminal deletion of 22q13 can be uncovered by [[karyotype]] analysis, but many terminal and interstitial deletions are too small to detect with this method(14,33). Chromosomal microarray should be ordered in children with suspected developmental delays or ASD(34,35).  Most cases will be identified by microarray; however, small variations in the ''SHANK3'' gene might be missed.  If ''SHANK3'' is highly suspected, Sanger or next generation sequencing or autism specific panels that include ''SHANK3'' are warranted.  The falling cost for whole [[exome sequencing]] may replace [[DNA microarray]] technology for candidate gene evaluation.  Biological parents should be tested with [[Fluorescence in situ hybridization|fluorescence ''in situ'' hybridization]] (FISH) to rule out balanced translocations or inversions.  Balanced translocation in a parent increases the risk for recurrence and heritability within families (figure 3)(36).
|| TNRC6B

|| FAM83F
Clinical genetic evaluations and dysmorphology exams should be done to evaluate growth, pubertal development, dysmorphic features (table 1) and screen for organ defects (table 2).
|| GRAP2

|| ENTHD1
b.      Cognitive and Behavioral Assessment
|| CACNA1I

|| RPS19BP1
All patients should undergo comprehensive developmental, cognitive and behavioral assessments by clinicians with experience in developmental disorders(1). Special emphasis should be placed on evaluating autism spectrum disorder (ASD) symptoms.  Gold standard assessment tools such as the Autism Standard Observation Schedule (ADOS), and the Autism Diagnostic Interview (ADI) are recommended; however, given the limitations of these tests in patients with profound intellectual disability results should be carefully integrated with expert clinical evaluation and results of cognitive testing.  Cognitive evaluation should be tailored for individuals with significant language and developmental delays(33). All patients should be referred for specialized speech/language, occupation and physical therapy evaluations.
|| ATF4

|-
c.       Neurological Management
|| SMCR7L

|| MGAT3
Individuals with PMS should be followed by a pediatric neurologist regularly to monitor motor development, coordination and gait, as well as conditions that might be associated with hypotonia, like neuromuscular scoliosis and feeding problems(1,4,37).   Head circumference should be performed routinely up until 36 months.  Given the high rate of seizure disorders (up to 41% of patients) reported in the literature in patients with PMS and its overall negative impact on development, an overnight video EEG should be considered early to rule out seizure activity.  In addition, a baseline structural brain MRI should be considered to rule out the presence of structural abnormalities(22).
|| TAB1

|| SNORD43
d.      Endocrinology
|| RPL3

|| PDGFB
Growth should be monitored routinely due to reports of short stature and accelerated growth.  In addition, all patients should have baseline assessment of thyroid function(33).  If clinically indicated, nutritional assessments should be contemplated(5,31).
|| CBX7

|| APOBEC3H
e.       Nephrology
|| APOBEC3F

|| APOBEC3D
All patients should have a baseline renal and bladder ultrasonography and a voiding cystourethrogram should be considered to rule out structural and functional abnormalities.  Renal abnormalities are reported in up to 38% of patients with PMS(1,38,39). Vesicouretral reflux, hydronephrosis, renal agenesis, dysplasic kidney, polycystic kidney and recurrent urinary tract infections have all been reported in patients with PMS.
|| APOBEC3C

|| APOBEC3B
f.       Cardiology
|-

|| CBX6
Congenital heart defects (CHD) are reported in samples of children with PMS with varying frequency (up to 25%)(29,36).  The most common CHD include tricuspid valve regurgitation, atrial septal defects and patent ductus arteriousus.  Cardiac evaluation, including echocardiography and electrocardiogram, should be performed to evaluate for CHD and cardiac function(33). 
|| NPTXR

|| DNAL4
g.      Gastroenterology
|| SUN2

|| GTPBP1
Gastrointestinal symptoms are common in individuals with PMS. Gastroesophageal reflux, constipation, diarrhea and cyclic vomiting are frequently described(1,5,40).
|| JOSD1
{| class="wikitable"
|| TOMM22
|'''Medical Specialty'''
|| CBY1
|'''Assessment Recommended'''
|| FAM227A
|-
|| DMC1
| rowspan="7" |'''Primary Care/Development Pediatrics'''
|| DDX17
|Careful and routine monitoring
|| KDELR3
|-
|-
|Hearing Assessment
|| KCNJ4
|-
|| CSNK1E
|Visual Assessment
|| TMEM184B
|-
|| MAFF
|Monitoring of height, weight and BMI
|| MAFF
|-
|| '''PLA2G6'''
|Otolaryngology (ENT)
|| BAIAP2L2
|-
|| SLC16A8
|Pediatric dentistry
|| PICK1
|-
|| '''SOX10'''
|Physiatrist/physical therapy
|| POLR2F
|-
|| C22orf23
| rowspan="7" |'''Psychiatric and Psychology'''
|-
|Psychiatric evaluation with focus on autism spectrum disorder
|| MICALL1
|-
|| EIF3L
|Autism Diagnostic Observation Schedule (ADOS)
|| ANKRD54
|-
|| GALR3
|Cognitive or Developmental Assessment
|| GCAT
|-
|| H1F0
|Speech and Language Evaluation/Therapy
|| TRIOBP
|-
|| NOL12
|Adaptive Function Testing
|| LGALS1
|-
|| SH3BP1
|Educational Assessment
|| GGA1
|-
|| LGALS2
|Occupational Therapy
|-
|-
|| CDC42EP1
| rowspan="4" |'''Neurology'''
|| CARD10
|Motor development, coordination and gait monitoring, as well as conditions that might be associated with hypotonia, like neuromuscular scoliosis and feeding problems
|| MFNG
|-
|| ELFN2
|Overnight video EEG
|| CYTH4
|}
|-
|Structural brain MRI
|-
|Head circumference up to 36 months
|-
|'''Nephrology'''
|Renal and bladder ultrasonography
|-
| rowspan="2" |'''Cardiology'''
|Echocardiogram
|-
|Electrocardiogram
|-
| rowspan="2" |'''Endocrinology'''
|Thyroid function
|-
|Nutritional assessment
|}
Table 3: Clinical Assessment Recommendations in Phelan McDermid Syndrome. 

'''2.     Resources for Families''' 

1.     Phelan McDermid Foundation: 22q13.org

2.     <nowiki>http://www.shank3gene.org/</nowiki>

3.     Association Française du Syndrome Phelan-McDermid (France): <nowiki>http://22q13.fr</nowiki>


4.     Unique for Phelan McDermid Syndrome (UK):
==Incidence==
The incidence of the 22q13 deletion syndrome is uncertain. The [[National Institutes of Health]] Office of Rare Diseases (http://rarediseases.info.nih.gov/) lists 22q13 deletion syndrome as a rare disease.


<nowiki>http://www.rarechromo.org/information/Chromosome%2022/22q13%20deletions%20Phelan%20McDermid%20syndrome%20FTNW.pdf</nowiki>
== See also ==
* [[22q11.2 deletion syndrome]]
* [[SHANK3]]


==Notes==
==Notes==

Revision as of 02:47, 4 August 2016

22q13 deletion syndrome
SpecialtyGenetics Edit this on Wikidata

Phelan-McDermid syndrome (PMS), also called 22q13 deletion syndrome, is a rare genetic condition characterized by global developmental delay, intellectual disability (ID), absent or delayed speech, autism spectrum disorder (ASD), hypotonia and mild dysmorphic features[1][2][3].   Affected individuals present in addition with an array of medical, neurological and behavioral symptoms (tables 1 and 2)[4],[5].  The core clinical and neurobehavioral symptoms in PMS syndrome are caused by the loss of one functional copy of the SHANK3 gene (haploinsufficiency)1, which is located in the distal segment of the long arm of chromosome 22[6] (figure 1).   The SHANK3 gene encodes a protein that is critical for neuronal communication and synapse formation[7][8].    About 80% of cases result from a spontaneous, or de novo mutation, and is not inherited from parents.

History

The first case of PMS was described in 1985 by Watt et al., who described a 14 year old boy with severe intellectual disability, mild dysmorphic features and absent speech, which was associated with terminal loss the distal arm of chromosome 22(9).  In 1988, Phelan et al. described a similar clinical presentation associated with a de novo deletion in 22q13.3(4).  Subsequent cases where described in the following years with a similar clinical presentation.  Phelan et al (2001)(4), compared 37 subjects with 22q13 deletions with features of 24 cases described in the literature finding that the most common features were global developmental delay, absent or delayed speech and hypotonia.  In 2001, Bonaglia et al.(10), described a case that associated the 22q.13 deletion syndrome with a disruption of the SHANK3 gene (also called ProSAP2).  The following year, Anderlid et al. (2002)(11), refined the area in 22q13 presumably responsible for the common phenotypic presentation of the syndrome to a 100kb  in 22q13.3 (image).  Out of the three genes affected, SHANK3 was identified as the critical gene due to its expression pattern and function.  Wilson et al. (2003)(12) evaluated 56 patients with the  clinical presentation of PMS, all of whom had a functional loss of one copy of the SHANK3 gene, further confirming the role of SHANK3 in the major neuropsychiatric manifestations of the syndrome.  Since then multiple other studies have reported similar findings with smaller deletions of the region or mutations of the gene(13).

Epidemiology

The true prevalence of PMS has not been determined. More than 1200 people have been identified worldwide according the Phelan-McDermid Syndrome Foundation. However, it is believed to be underdiagnosed due to inadequate genetic testing and lack of specific clinical features. It is known to occur with equal frequency in males and females(7).   Studies using chromosomal microarray for diagnosis indicate that at least 0.5% of cases of ASD can be explained by mutations or deletions in the SHANK3 gene(14). In addition when ASD is associated with ID, SHANK3 mutations or deletions have been found in up to 2% of individuals(15,16).  The rate of SHANK3 insufficiency in ID and ASD is still being determined; however, these findings imply that it is one of the most common single gene causes.

Etiology

The loss of one copy of SHANK3 can result from chromosomal abnormalities in the terminal end of chromosome 22 or from a mutation in SHANK3 leading to a loss of function in the protein.  The SHANK3 gene codes for a protein (SHANK3) that is vital for synaptic formation and plasticity(17,18) (figure 2). It is expressed widely in the brain early in the postnatal period and its expression peaks during synaptic formation and maturation(13).   The protein is found in the hippocampus, the striatum, cerebellar granular cells, olfactory bulb and the thalamic nuclei(13,19).  SHANK proteins are critical for the architectural make-up of the dendritic spines and their expression is essential for cognition and learning(17).  SHANK3 is a scaffolding protein located in the postsynaptic density of excitatory synapses (figure 2)(7,13).  Genetic manipulation of Shank3 in animal model systems leads to changes in the key components of glutamatergic synapse and dendritic spine density.

Chromosomal abnormalities leading to the loss of the terminal arm of chromosome 22 include unbalanced chromosomal translocations, terminal or interstitial deletions involving the 22q13.3 region, a ring chromosome, or other structural changes (figure 3). Terminal deletions account for about 75% of the cases of PMS(7).  Some studies suggest a relationship between deletion size and the extent or severity of certain clinical features, but this has not been well established and remains the focus of clinical research(1,20–24).    

Clinical Description

Affected individuals present with a broad array of medical and behavioral manifestations (tables 1 and 2).  Patients are consistently characterized by global developmental delay, intellectual disability, speech abnormalities, ASD, hypotonia and mild dysmorphic features.    Table 1 summarizes the dysmorphic and medical conditions that have been reported in individuals with PMS.  Table 2 summarize the psychiatric and neurological associated with SHANK3 deficiency and PMS (1,4,21,23–33).  Most of the studies include small samples or relied on parental report or medical record review to collect information, which can account in part for the variability in the presentation of some of the presenting features.  Larger prospective studies are needed to further characterize the disorder genetic. It is likely that as the awareness, of PMS and the role of SHANK3 deficiency in ASD and ID, increases among the medical community more individuals will be identified which will provide a better understanding of the disorder.

Dysmorphic Feature Percentage (%) Medical Comorbidities Percentage (%)
Macrocephaly 7-31 Hypothyroidism 3-6
Microcephaly 11-14 Sleep disturbance 41-46
Dolichocephaly 23-86 Gastroesophageal reflux 42-44
Long eyelashes 43-93 Increased pain threshold 10-88
Bulbous nose 47-80 Constipation/diarrhea 38-41
High arched palate 25-47 Brain imaging abnormalities 7-75
Malocclusion/widely spaced teeth 19 Recurrent upper respiratory infections 8-53
Full cheeks 25 Renal abnormalities 17-26
Pointed chin 22-62 Lymphedema 8-53
Large fleshy hands 33-68 Seizures 14-41
Hypoplastic/dysplastic nails 3-78 Strabismus 6-26
Hyper-extensibility 25-61 Short stature 11-13
Abnormal spine curvature 22 Tall stature/accelerated growth 3-18
Sacral dimple 13-37 Cardiac defects 3-25
Syndactyly of toes 2 and 3 9-48 Precocious or delayed puberty 12

Table 1: Dysmorphic features and medical comorbid conditions that have been reported in individuals with Phelan McDermid Syndrome.

Psychiatric and Neurologic Manifestations Percentage (%)
Autism Spectrum disorder >50
Intellectual Disability >75
Global Developmental Delay >75
Absent or severely affected speech >75
Sensory seeking behaviors (mouthing of objects) >25
Teeth grinding >25
Hyperactivity and inattention >50
Stereotypical movements >50
Hypotonia >50
Fine and gross motor abnormalities >90
Poor fine motor coordination >90
Gait Abnormalities >90
 Visual tracking abnormalities >85
Seizure disorder 17-41
Brain structural abnormalities 44-100
Sleep problems >40

Table 2: Psychiatric and Neurologic Manifestations associated with Phelan McDermid Syndrome

1.     Diagnosis and Management

a.       Clinical Genetics and Genetic Testing

Genetic testing is necessary to confirm SHANK3 deficiency and the diagnosis of PMS.  A prototypical terminal deletion of 22q13 can be uncovered by karyotype analysis, but many terminal and interstitial deletions are too small to detect with this method(14,33). Chromosomal microarray should be ordered in children with suspected developmental delays or ASD(34,35).  Most cases will be identified by microarray; however, small variations in the SHANK3 gene might be missed.  If SHANK3 is highly suspected, Sanger or next generation sequencing or autism specific panels that include SHANK3 are warranted.  The falling cost for whole exome sequencing may replace DNA microarray technology for candidate gene evaluation.  Biological parents should be tested with fluorescence in situ hybridization (FISH) to rule out balanced translocations or inversions.  Balanced translocation in a parent increases the risk for recurrence and heritability within families (figure 3)(36).

Clinical genetic evaluations and dysmorphology exams should be done to evaluate growth, pubertal development, dysmorphic features (table 1) and screen for organ defects (table 2).

b.      Cognitive and Behavioral Assessment

All patients should undergo comprehensive developmental, cognitive and behavioral assessments by clinicians with experience in developmental disorders(1). Special emphasis should be placed on evaluating autism spectrum disorder (ASD) symptoms.  Gold standard assessment tools such as the Autism Standard Observation Schedule (ADOS), and the Autism Diagnostic Interview (ADI) are recommended; however, given the limitations of these tests in patients with profound intellectual disability results should be carefully integrated with expert clinical evaluation and results of cognitive testing.  Cognitive evaluation should be tailored for individuals with significant language and developmental delays(33). All patients should be referred for specialized speech/language, occupation and physical therapy evaluations.

c.       Neurological Management

Individuals with PMS should be followed by a pediatric neurologist regularly to monitor motor development, coordination and gait, as well as conditions that might be associated with hypotonia, like neuromuscular scoliosis and feeding problems(1,4,37).   Head circumference should be performed routinely up until 36 months.  Given the high rate of seizure disorders (up to 41% of patients) reported in the literature in patients with PMS and its overall negative impact on development, an overnight video EEG should be considered early to rule out seizure activity.  In addition, a baseline structural brain MRI should be considered to rule out the presence of structural abnormalities(22).

d.      Endocrinology

Growth should be monitored routinely due to reports of short stature and accelerated growth.  In addition, all patients should have baseline assessment of thyroid function(33).  If clinically indicated, nutritional assessments should be contemplated(5,31).

e.       Nephrology

All patients should have a baseline renal and bladder ultrasonography and a voiding cystourethrogram should be considered to rule out structural and functional abnormalities.  Renal abnormalities are reported in up to 38% of patients with PMS(1,38,39). Vesicouretral reflux, hydronephrosis, renal agenesis, dysplasic kidney, polycystic kidney and recurrent urinary tract infections have all been reported in patients with PMS.

f.       Cardiology

Congenital heart defects (CHD) are reported in samples of children with PMS with varying frequency (up to 25%)(29,36).  The most common CHD include tricuspid valve regurgitation, atrial septal defects and patent ductus arteriousus.  Cardiac evaluation, including echocardiography and electrocardiogram, should be performed to evaluate for CHD and cardiac function(33). 

g.      Gastroenterology

Gastrointestinal symptoms are common in individuals with PMS. Gastroesophageal reflux, constipation, diarrhea and cyclic vomiting are frequently described(1,5,40).

Medical Specialty Assessment Recommended
Primary Care/Development Pediatrics Careful and routine monitoring
Hearing Assessment
Visual Assessment
Monitoring of height, weight and BMI
Otolaryngology (ENT)
Pediatric dentistry
Physiatrist/physical therapy
Psychiatric and Psychology Psychiatric evaluation with focus on autism spectrum disorder
Autism Diagnostic Observation Schedule (ADOS)
Cognitive or Developmental Assessment
Speech and Language Evaluation/Therapy
Adaptive Function Testing
Educational Assessment
Occupational Therapy
Neurology Motor development, coordination and gait monitoring, as well as conditions that might be associated with hypotonia, like neuromuscular scoliosis and feeding problems
Overnight video EEG
Structural brain MRI
Head circumference up to 36 months
Nephrology Renal and bladder ultrasonography
Cardiology Echocardiogram
Electrocardiogram
Endocrinology Thyroid function
Nutritional assessment

Table 3: Clinical Assessment Recommendations in Phelan McDermid Syndrome. 

2.     Resources for Families 

1.     Phelan McDermid Foundation: 22q13.org

2.     http://www.shank3gene.org/

3.     Association Française du Syndrome Phelan-McDermid (France): http://22q13.fr

4.     Unique for Phelan McDermid Syndrome (UK):

http://www.rarechromo.org/information/Chromosome%2022/22q13%20deletions%20Phelan%20McDermid%20syndrome%20FTNW.pdf

Notes

  1. ^ Soorya, Latha; Kolevzon, Alexander; Zweifach, Jessica; Lim, Teresa; Dobry, Yuriy; Schwartz, Lily; Frank, Yitzchak; Wang, A Ting; Cai, Guiqing (2013-06-11). "Prospective investigation of autism and genotype-phenotype correlations in 22q13 deletion syndrome and SHANK3 deficiency". Molecular Autism. 4: 18. doi:10.1186/2040-2392-4-18. ISSN 2040-2392. PMC 3707861. PMID 23758760.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  2. ^ Phelan, K.; McDermid, H.E. "The 22q13.3 Deletion Syndrome (Phelan-McDermid Syndrome)". Molecular Syndromology. doi:10.1159/000334260. PMC 3366702. PMID 22670140.
  3. ^ Kolevzon, Alexander; Angarita, Benjamin; Bush, Lauren; Wang, A. Ting; Frank, Yitzchak; Yang, Amy; Rapaport, Robert; Saland, Jeffrey; Srivastava, Shubhika (2014-01-01). "Phelan-McDermid syndrome: a review of the literature and practice parameters for medical assessment and monitoring". Journal of Neurodevelopmental Disorders. 6 (1): 39. doi:10.1186/1866-1955-6-39. ISSN 1866-1947. PMC 4362650. PMID 25784960.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  4. ^ Phelan, M. C.; Rogers, R. C.; Saul, R. A.; Stapleton, G. A.; Sweet, K.; McDermid, H.; Shaw, S. R.; Claytor, J.; Willis, J. (2001-06-15). "22q13 deletion syndrome". American Journal of Medical Genetics. 101 (2): 91–99. ISSN 0148-7299. PMID 11391650.
  5. ^ Sarasua, Sara M.; Boccuto, Luigi; Sharp, Julia L.; Dwivedi, Alka; Chen, Chin-Fu; Rollins, Jonathan D.; Rogers, R. Curtis; Phelan, Katy; DuPont, Barbara R. (2014-07-01). "Clinical and genomic evaluation of 201 patients with Phelan-McDermid syndrome". Human Genetics. 133 (7): 847–859. doi:10.1007/s00439-014-1423-7. ISSN 1432-1203. PMID 24481935.
  6. ^ Costales, Jesse; Kolevzon, Alexander (2016-04-01). "The therapeutic potential of insulin-like growth factor-1 in central nervous system disorders". Neuroscience & Biobehavioral Reviews. 63: 207–222. doi:10.1016/j.neubiorev.2016.01.001. PMC 4790729. PMID 26780584.
  7. ^ Harony-Nicolas, Hala; Rubeis, Silvia De; Kolevzon, Alexander; Buxbaum, Joseph D. (2015-12-01). "Phelan McDermid Syndrome From Genetic Discoveries to Animal Models and Treatment". Journal of Child Neurology. 30 (14): 1861–1870. doi:10.1177/0883073815600872. ISSN 0883-0738. PMID 26350728.
  8. ^ Carbonetto, Salvatore (2014-02-01). "A blueprint for research on Shankopathies: a view from research on autism spectrum disorder". Developmental Neurobiology. 74 (2): 85–112. doi:10.1002/dneu.22150. ISSN 1932-846X. PMID 24218108.

References

External links