Next Article in Journal
An Expanding Toolkit for Heterochromatin Repair Studies
Next Article in Special Issue
Circulating miRNAs as Potential Biomarkers for Patient Stratification in Bipolar Disorder: A Combined Review and Data Mining Approach
Previous Article in Journal
PTCHD1: Identification and Neurodevelopmental Contributions of an Autism Spectrum Disorder and Intellectual Disability Susceptibility Gene
Previous Article in Special Issue
Identification of Key Modules and Genes Associated with Major Depressive Disorder in Adolescents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Commentary

Phenotypic Variability in Phelan–McDermid Syndrome and Its Putative Link to Environmental Factors

1
Healthcare Genetics Program, School of Nursing, College of Behavioral, Social and Health Sciences, Clemson University, Clemson, SC 29634, USA
2
Laboratory of Neuropsychology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
3
Department of Health Sciences, University Magna Graecia, 88100 Catanzaro, Italy
4
Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Indiana University School of Medicine/Riley Hospital for Children, Indianapolis, IN 46202, USA
5
Greenwood Genetic Center, Greenwood, SC 29646, USA
6
Genetics Laboratory, Florida Cancer Specialists &Research Institute, Fort Myers, FL 33916, USA
*
Author to whom correspondence should be addressed.
Genes 2022, 13(3), 528; https://doi.org/10.3390/genes13030528
Submission received: 5 March 2022 / Revised: 14 March 2022 / Accepted: 15 March 2022 / Published: 17 March 2022
(This article belongs to the Special Issue Gene-Exposome Interactions in Mental Illness)

Abstract

:
Phelan–McDermid syndrome (PMS) is a multi-systemic disorder characterized by both genetic and phenotypic variability. Genetic abnormalities causing PMS span from pathogenic variants of the SHANK3 gene to chromosomal rearrangements affecting the 22q13 region and leading to the loss of up to over nine megabases. The clinical presentation of individuals with PMS includes intellectual disability, neonatal hypotonia, delayed or absent speech, developmental delay, and minor dysmorphic facial features. Several other features may present with differences in age of onset and/or severity: seizures, autism, regression, sleep disorders, gastrointestinal problems, renal disorders, dysplastic toenails, and disrupted thermoregulation. Among the causes of this phenotypic variability, the size of the 22q13 deletion has effects that may be influenced by environmental factors interacting with haploinsufficiency or hemizygous variants of certain genes. Another mechanism linking environmental factors and phenotypic variability in PMS involves the loss of one copy of genes like BRD1 or CYP2D6, located at 22q13 and involved in the regulation of genomic methylation or pharmacokinetics, which are also influenced by external agents, such as diet and drugs. Overall, several non-mutually exclusive genetic and epigenetic mechanisms interact with environmental factors and may contribute to the clinical variability observed in individuals with PMS. Characterization of such factors will help to better manage this disorder.

1. Introduction

Defining the contribution of environmental factors to the clinical presentation of a genetic disorder is often challenging. Many phenotypic features can be caused either by genetic alterations alone, environmental factors alone, or a combination of both. One example is the decreased growth rate in infancy, which can be due to failure to thrive (generally, a sign of genetic abnormalities), malnourishment (the result of a poor diet), or both. However, certain genetic disorders caused by large genomic alterations, with disruption or loss of several genes, leave little or no margin for a putative contribution of environmental factors. Theoretically, one of these disorders is Phelan–McDermid syndrome (PMS) (OMIM #606232): most cases are caused by chromosomal rearrangements leading to the loss of portions of the 22q13 region ranging from less than 100 kb to over 9 Mb [1,2,3,4]. In addition to chromosomal deletions of variable sizes, PMS can also be caused by pathogenic variants of the SHANK3 gene, mapping in the telomeric region of 22q13.33 [5,6]. Such genetic heterogeneity is further complicated by cases with interstitial 22q13 deletions (not encompassing SHANK3) presenting with clinical features compatible with the PMS phenotype [7]. The clinical presentation of PMS is also variable; it can include moderate to profound developmental delay, neonatal hypotonia, delayed or absent speech, autistic traits, motor impairment, seizures, sleep disorders, gastrointestinal problems, renal disorders, dysplastic toenails, disrupted thermoregulation, and minor dysmorphic traits [1,2,3,4,5,6]. The difficulties of sorting the clinical profiles of individuals with interstitial 22q13 deletions from those with terminal deletions have led to a new classification of this disorder as PMS-SHANK3 related, for cases with deletions or pathogenic variants affecting SHANK3, and PMS-SHANK3 unrelated, for the remaining cases where SHANK3 is preserved [8]. Although this classification promises to increase the diagnostic yield for this syndrome, it still does not explain the complexity of the genotype-phenotype correlation. Militating against easy genotype-phenotype assignment, some cases of PMS with large deletions present with relatively milder phenotypes and some cases with small deletions or pathogenic variants of SHANK3 show severe features.

2. Possible Gene-Environment Interactions in Phelan–McDermid Syndrome

The widening of the investigational approach to 22q13 genes beyond the terminal region [9,10] and even to a genomic scale [11] has highlighted the potential contribution of various genes that can exert a modifying effect on PMS phenotypes. Moreover, the contribution of some of these genes can be, in turn, influenced by certain environmental factors.
In this commentary paper, we focus on a limited number of examples of the myriad of possible gene-environment interactions that can affect the PMS clinical presentation and we propose two major, but not necessarily mutually exclusive, mechanisms: (1) the loss of one copy of a 22q13 gene leads to haploinsufficiency or unmasks a pathogenic variant (hemizygosity), altering interactions with environmental factors in specific nodes of metabolic pathways; (2) the loss of a gene has a broad downstream effect, involving numerous genetic and/or epigenetic regulatory processes and affecting multiple pathways simultaneously.

2.1. Haploinsufficiency or Hemizygous Variants of 22q13 Genes

2.1.1. PNPLA3 and Liver Disease

The PNPLA3 gene maps almost 7 Mb from the 22q telomere and can therefore be involved in both large terminal deletions [3,11] and interstitial deletions [7,9]. Consequently, its mechanism of gene-environment interaction may apply to both PMS-SHANK3 related and PMS-SHANK3 unrelated cases. The protein encoded by this gene—patatin-like phospholipase domain-containing protein 3 (PNPLA3), or adiponutrin—is a lipase that mediates triacylglycerol hydrolysis in adipocytes and hepatocytes; its expression is induced in the liver after feeding and during insulin resistance [12].
PNPLA3 variants, particularly the p.Ile148Met change, have been associated with both alcoholic and non-alcoholic liver disease, with a higher predisposing effect in homozygous subjects than in heterozygous ones [12,13]. It has been reported that both homozygous and hemizygous PNPLA3 variants can be detected in subjects with PMS, leading to steatohepatitis, hepatic fibrosis, and liver metabolic dysfunction, which in turn may cause a lack of response to pharmaceutical treatments [14].
The pathogenic effects of PNPLA3 variants cause accumulation of lipids in the hepatocytes (macrovesicular steatosis) and may be amplified by environmental factors such as a high-calorie diet, alcohol, or oxidants (Table 1).
A similar mechanism can be proposed for the PPARA gene, mapping on 22q13.31 and encoding the peroxisome proliferator-activated receptor α: the complete deficiency of the Ppara protein in mouse hepatocytes leads to non-alcoholic fatty liver disease and liver inflammation in combination with a high-fat diet [15,16]. The abnormal lipid peroxidation induced by the lack of Ppara promotes the progression of liver injury to steatohepatitis and was proven to play a pivotal role in the adaptation to sepsis [16,17].
For this reason, individuals with PMS who have lost a copy of PNPLA3 and/or PPARA should be screened for variants in the remaining copy and eventually follow a low-calorie, alcohol-free dietary regimen to reduce the risk for liver disease.

2.1.2. SCO2, TYMP, SULT4A1, and Mitochondrial Dysfunction

Numerous genes involved in mitochondrial functions are located within the 22q13 region [18], and several studies have suggested that the haploinsufficiency of some of these genes, particularly SCO2, TYMP, and SULT4A1 may contribute to the clinical presentation of PMS [7,9,10,11,19]. SCO2 encodes a copper metallochaperone involved in the synthesis and maturation of the subunit II of cytochrome C oxidase, whose deficiency results in an increase in reactive oxygen species [10,18]. Pathogenic variants of this gene, such as the rare nonsense change p.Gln53* or the missense substitutions p.Ala259Val and p.Arg114His, can cause a severe form of autosomal dominant myopia (OMIM #608908) [20] or a fatal infantile cardioencephalomyopathy (OMIM #604377) [21]. TYMP’s product is a thymidine phosphorylase that promotes growth, angiogenic and chemotactic activities in endothelial cells. The reversible phosphorylase of thymidine catalysed by TYMP is also crucial for the rescue of pyrimidines for nucleotide synthesis. The lack of activity of this enzyme causes the autosomal recessive disorder mitochondrial DNA depletion syndrome 1 (MNGIE type) [10].
The neuron-specific sulfotransferase SULT4A1 localizes to the mitochondrial outer membrane and negatively regulates the interaction between SULT1A1 and SULT1A3, and through this mechanism, dopamine toxicity [22]. SULT4A1 protects against mitochondrial dysfunction induced by oxidative stress, presumably via the same mechanism [23]. Knockout of Sult4a1 in mice causes progressive neurological degeneration and early death [24]. Loss of one copy of the SULT4A1 gene has been implicated in PMS-SHANK3 unrelated cases [7,9], but it may as well occur in PMS-SHANK3 related cases carrying terminal deletions larger than 7 Mb [3,11].
Abnormal levels of reactive oxygen species and a decreased capacity of mitochondria to produce DNA and therefore adjust to different metabolic conditions can be further worsened by certain drugs, like chemotherapeutic agents or Nrf2-inhibitors, and lack of anti-oxidants introduced via diet and supplements [25,26] (Table 1).

2.1.3. SHANK3 and Inflammation

SHANK3 maps in the subtelomeric region of chromosome 22q (22q13.33), encodes a multidomain scaffold protein of the post-synaptic density of several types of excitatory synapses, and its pathogenic alterations have been associated with several disorders, including, in addition to PMS-SHANK3 related [5,6,8], autism spectrum disorder (ASD), schizophrenia, a Rett syndrome-like phenotype, and intellectual disability [27].
Disruption of protein-protein interactions at the postsynaptic density is a well-characterized model for the pathogenesis of ASD. SHANK proteins fit this model, as they are critical for the stability of glutamate receptors and cell adhesion proteins [28]. For this reason, several in vivo studies have targeted Shank3 to investigate genetic and environmental factors that could affect synaptic stability and function, leading to ASD or other neurobehavioral disorders associated with this gene. One of such studies has shown that acute inflammation from a lipopolysaccharide challenge can acutely unmask behavioural deficits in Shank3 haploinsufficient mice. The loss of social preference can be reversed by Trpv4 inhibition in the Nucleus Accumbens (NA) [29]. Early postnatal downregulation of NA Shank3 creates a similar behavioural deficit by shifting the membrane properties of dopamine type 1-expressing medium spiny neurons. The behavioural deficits are also rescued by Trpv4 inhibition, suggesting a mechanism in common between developmental and environmental influences.
Interestingly, the inflammatory response does not target directly SHANK3 but rather exerts its influence via a gene-gene-environment model, where the environmental factor—the immune response—targets a gene, TRPV4, which maps on a different chromosome from SHANK3 (12q24.11), and via its activation disrupts SHANK3 function at the post-synaptic density causing eventually reversible behavioural deficits. This model provides a valuable example of the intricacy of connections between genes and environmental factors and suggests that indirect influence on gene function may play an important role in the variability in genotype-phenotype correlation for PMS and other conditions.

2.2. Genome-Wide Effects

2.2.1. BRD1 and Epigenomic Dysregulation

Bromodomain-containing protein 1 (BRD1) is a component of the MOZ/MORF complex, which has a histone H3 acetyltransferase activity [10]. BRD1 pathogenic variants have been associated with schizophrenia and functional studies on haploinsufficient animal models showed that this gene’s product modulates, via epigenetic regulation, behaviour, neurotransmission, and expression of genes involved in pathways associated with mental illness and molecular signaling processes [30]. Homozygous Brd1 knockout mice have failure to thrive, seizures, decreased levels of histone H3 acetylation (H3K9ac, H3K14ac, and H3K18ac), and increased N-tail clipping [31].
Loss of one copy of BRD1 alters genome-wide methylation, likely disrupting the epigenetic regulation of the expression of numerous genes (Table 1): the downstream effects are so broad that they significantly change genome-wide methylation and metabolic profiles in individuals with PMS [32]. For example, Thomford et al. [33] demonstrated how the methylation status of the promoter of another 22q13 gene previously discussed, SCO2, affects the penetrance of the effects of pathogenic variants in the non-deleted allele, suggesting a gene-epigene interaction leading to a potentially more severe phenotype. Epigenetic alterations have been associated with various conditions, including ASD [34], often present in individuals with PMS. Moreover, environmental factors such as medications have been shown to influence the risk for ASD and other neurodevelopmental conditions [35], plausibly via epigenetic modifications. It is plausible that epigenetic-mediated effects predisposing to ASD may play an additive or even synergistic role in PMS in combination with the disruption of SHANK3, the main 22q13 candidate gene for ASD and other neurological issues.
In conclusion, patients with 22q13 deletions encompassing BRD1 may benefit from pharmacogenomic counseling to identify and eventually avoid drugs that can alter their epigenetic profile.

2.2.2. CYP2D6 and Pharmacogenomics

The CYP2D6 gene encodes the cytochrome P450DB1 protein and maps in 22q13.2, therefore, just as discussed for PNPAL3 and SULT4A1, the gene-environment interaction involving this gene can apply to large terminal deletions (PMS-SHANK3 related cases) as well as interstitial deletions preserving the telomeric regions (PMS-SHANK3 unrelated cases).
Cytochrome p450 proteins (CYP) constitute a superfamily of enzymes containing heme as a cofactor that function as monooxygenases and are involved in the metabolization of numerous types of drugs. For this reason, extensive research has been focused on the effect of variants in genes encoding CYP proteins on the pharmacokinetics of several compounds. Genes for CYP enzymes, such as CYP2C9, CYP2C19, CYP2D6, CYP3A4, and CYP3A5, have been classified as important pharmacogenes [36].
For CYP2D6 in particular, established genotype-phenotype relationships allow for patients to be categorized into four groups based on the variant status: ultra-rapid metabolizers, extensive (normal) metabolizers, intermediate metabolizers, and poor metabolizers [37]. Among the classes of pharmaceutics whose efficacy is influenced by CYP2D6 variants, there are compounds that can be used in the clinical management of individuals with PMS, like antidepressants, antipsychotics, and epileptics (Table 1). The potential co-occurrence of variants in other pharmacogenes may further affect the efficacy of these classes of drugs by increasing the disruption of the cytochrome p450 activity.
Therefore, physicians of subjects with PMS who carry deletions encompassing CYP2D6 should consider genetic screening for variants in the preserved copy of this gene and other pharmacogenes, and eventually explore the possibility of pharmacogenomic counselling.

3. Critical Period Effects

Certain gene-environment interactions are limited to specific times of growth and development and thus not yet fully characterized. It is well understood, for example, that neurodevelopment undergoes critical periods and that these critical periods are temporal windows sensitive to disruptive influences from environmental factors [38]. Critical periods are associated with manifestations of neurodevelopmental disorders, affecting cognitive, social, and motor performance. Although environmental factors affecting critical periods may play an important role in PMS phenotype variability, the mechanistic details of this role remain largely unexplored. A step towards identifying potential gene-environment interactions is to identify genes of 22q13 that play a major, transient role in neurodevelopment, and that are likely sensitive to haploinsufficiency. An example is the PHF21B gene, whose sequence is highly constrained, indicating likely sensitivity to haploinsufficiency [9]. A change in isoform expression of this gene triggers the transition from neuronal progenitor cell to neuron during cortical neurogenesis through an epigenetic mechanism [39]. Neuronal differentiation has been associated with a wide variety of disruptive environmental factors [38], making PHF21B an obvious candidate for further investigation.

4. Conclusions

Although environmental factors play no causative role in the pathogenesis of PMS, there is evidence that they may affect the onset and/or severity of certain features. Characterization of their contribution to the syndrome’s phenotypes may improve clinical management and prognosis. This commentary paper provides some examples of gene-environment interactions that bear the potential of highlighting validated or hypothetical models for future investigation, suggesting molecules or pathways that could be used as biomarkers for early screening or prognosis, and identifying novel targets for precision treatments. The investigation of the impact of environmental factors on genetic disorders is still limited by the lack of information collected via functional studies: the complexity of the multiple interactions between different genes and various environmental factors is difficult to replicate in models designed to assess the impact of single variables. Future approaches will benefit from the data collected by omic studies, which will probably broaden our knowledge on the interactions between genes, proteins, and metabolites, identifying novel targets for environmental factors as well. Moreover, investigation of the gene-environment interactions in PMS provides valuable information on pathogenic mechanisms involving genes within and outside the 22q13 region and suggests an approach applicable to other multigenic disorders.

Author Contributions

Conceptualization, L.B. and A.M.; methodology, S.M.S.; validation, C.R., B.D. and K.P.; data curation, L.A. and W.B.; writing—original draft preparation, L.B. and A.M.; writing—review and editing, L.A., S.M.S., C.R., B.D. and K.P.; visualization, L.A. and W.B.; supervision, L.B., S.M.S., C.R. and K.P.; project administration, L.B.; funding acquisition, A.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported (in part) by the Intramural Research Program of the National Institute of Mental Health, National Institutes of Health (Annual Report numbers: ZICMH002961 and ZIAMH002886).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Phelan, K.; Rogers, R.C.; Boccuto, L. Phelan-McDermid Syndrome. In GeneReviews®; Adam, M.P., Ardinger, H.H., Pagon, R.A., Wallace, S.E., Bean, L.J.H., Gripp, K.W., Mirzaa, G.M., Amemiya, A., Eds.; University of Washington: Seattle, DC, USA, 1993. [Google Scholar]
  2. Kolevzon, A.; Angarita, B.; Bush, L.; Wang, A.T.; Frank, Y.; Yang, A.; Rapaport, R.; Saland, J.; Srivastava, S.; Farrell, C.; et al. Phelan-McDermid syndrome: A review of the literature and practice parameters for medical assessment and monitoring. J. Neurodev. Disord. 2014, 6, 39. [Google Scholar] [CrossRef] [PubMed]
  3. Phelan, K.; Rogers, R.C.; Boccuto, L. Deletion 22q13 syndrome: Phelan–McDermid syndrome. In Cassidy and Allanson’s Management of Genetic Syndromes; Wiley: New York, NY, USA, 2021; pp. 317–334. [Google Scholar]
  4. Sarasua, S.M.; Boccuto, L.; Sharp, J.L.; Dwivedi, A.; Chen, C.F.; Rollins, J.D.; Rogers, R.C.; Phelan, K.; DuPont, B.R. Clinical and genomic evaluation of 201 patients with Phelan-McDermid syndrome. Hum. Genet. 2014, 133, 847–859. [Google Scholar] [CrossRef]
  5. Soorya, L.; Kolevzon, A.; Zweifach, J.; Lim, T.; Dobry, Y.; Schwartz, L.; Frank, Y.; Wang, A.T.; Cai, G.; Parkhomenko, E.; et al. Prospective investigation of autism and genotype-phenotype correlations in 22q13 deletion syndrome and SHANK3 deficiency. Mol. Autism 2013, 4, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. De Rubeis, S.; Siper, P.M.; Durkin, A.; Weissman, J.; Muratet, F.; Halpern, D.; Trelles, M.; Frank, Y.; Lozano, R.; Wang, A.T.; et al. Delineation of the genetic and clinical spectrum of Phelan-McDermid syndrome caused by SHANK3 point mutations. Mol. Autism 2018, 9, 31. [Google Scholar] [CrossRef] [PubMed]
  7. Disciglio, V.; Lo Rizzo, C.; Mencarelli, M.A.; Mucciolo, M.; Marozza, A.; Di Marco, C.; Massarelli, A.; Canocchi, V.; Baldassarri, M.; Ndoni, E.; et al. Interstitial 22q13 deletions not involving SHANK3 gene: A new contiguous gene syndrome. Am. J. Med. Genet. Part A 2014, 164, 1666–1676. [Google Scholar] [CrossRef]
  8. Phelan, K.; Boccuto, L.; Powell, C.M.; Boeckers, T.M.; van Ravenswaaij-Arts, C.; Rogers, R.C.; Sala, C.; Verpelli, C.; Thurm, A.; Bennett, W.E., Jr.; et al. Phelan-McDermid syndrome: A classification system after 30 years of experience. Orphanet J. Rare Dis. 2022, 17, 27. [Google Scholar] [CrossRef]
  9. Mitz, A.R.; Philyaw, T.J.; Boccuto, L.; Shcheglovitov, A.; Sarasua, S.M.; Kaufmann, W.E.; Thurm, A. Identification of 22q13 genes most likely to contribute to Phelan McDermid syndrome. Eur. J. Hum. Genet. 2018, 26, 293–302. [Google Scholar] [CrossRef] [Green Version]
  10. Ricciardello, A.; Tomaiuolo, P.; Persico, A.M. Genotype-phenotype correlation in Phelan-McDermid syndrome: A comprehensive review of chromosome 22q13 deleted genes. Am. J. Med. Genet. Part A 2021, 185, 2211–2233. [Google Scholar] [CrossRef]
  11. Tabet, A.C.; Rolland, T.; Ducloy, M.; Lévy, J.; Buratti, J.; Mathieu, A.; Haye, D.; Perrin, L.; Dupont, C.; Passemard, S.; et al. A framework to identify contributing genes in patients with Phelan-McDermid syndrome. NPJ Genom. Med. 2017, 2, 32. [Google Scholar] [CrossRef] [PubMed]
  12. Boccuto, L.; Abenavoli, L. Genetic and Epigenetic Profile of Patients With Alcoholic Liver Disease. Ann. Hepatol. 2017, 16, 490–500. [Google Scholar] [CrossRef] [PubMed]
  13. Dongiovanni, P.; Donati, B.; Fares, R.; Lombardi, R.; Mancina, R.M.; Romeo, S.; Valenti, L. PNPLA3 I148M polymorphism and progressive liver disease. World J. Gastroenterol. 2013, 19, 6969–6978. [Google Scholar] [CrossRef] [PubMed]
  14. Boccuto, L.; Abenavoli, L.; Cascio, L.; Srikanth, S.; DuPont, B.; Mitz, A.R.; Rogers, R.C.; Phelan, K. Variability in Phelan-McDermid syndrome: The impact of the PNPLA3 p.I148M polymorphism. Clin. Genet. 2018, 94, 590–591. [Google Scholar] [CrossRef] [PubMed]
  15. Régnier, M.; Polizzi, A.; Smati, S.; Lukowicz, C.; Fougerat, A.; Lippi, Y.; Fouché, E.; Lasserre, F.; Naylies, C.; Bétoulières, C.; et al. Hepatocyte-specific deletion of Pparα promotes NAFLD in the context of obesity. Sci. Rep. 2020, 10, 6489. [Google Scholar] [CrossRef] [PubMed]
  16. Ronis, M.; Mercer, K.; Engi, B.; Pulliam, C.; Zimniak, P.; Hennings, L.; Shearn, C.; Badger, T.; Petersen, D. Global Deletion of Glutathione S-Transferase A4 Exacerbates Developmental Nonalcoholic Steatohepatitis. Am. J. Pathol. 2017, 187, 418–430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Paumelle, R.; Haas, J.T.; Hennuyer, N.; Baugé, E.; Deleye, Y.; Mesotten, D.; Langouche, L.; Vanhoutte, J.; Cudejko, C.; Wouters, K.; et al. Hepatic PPARα is critical in the metabolic adaptation to sepsis. J. Hepatol. 2019, 70, 963–973. [Google Scholar] [CrossRef] [PubMed]
  18. Frye, R.E.; Cox, D.; Slattery, J.; Tippett, M.; Kahler, S.; Granpeesheh, D.; Damle, S.; Legido, A.; Goldenthal, M.J. Mitochondrial Dysfunction may explain symptom variation in Phelan-McDermid Syndrome. Sci. Rep. 2016, 6, 19544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Srikanth, S.; Jain, L.; Zepeda-Mendoza, C.; Cascio, L.; Jones, K.; Pauly, R.; DuPont, B.; Rogers, C.; Sarasua, S.; Phelan, K.; et al. Position effects of 22q13 rearrangements on candidate genes in Phelan-McDermid syndrome. PLoS ONE 2021, 16, e0253859. [Google Scholar] [CrossRef] [PubMed]
  20. Tran-Viet, K.N.; Powell, C.; Barathi, V.A.; Klemm, T.; Maurer-Stroh, S.; Limviphuvadh, V.; Soler, V.; Ho, C.; Yanovitch, T.; Schneider, G.; et al. Mutations in SCO2 are associated with autosomal-dominant high-grade myopia. Am. J. Hum. Genet. 2013, 92, 820–826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Papadopoulou, L.C.; Sue, C.M.; Davidson, M.M.; Tanji, K.; Nishino, I.; Sadlock, J.E.; Krishna, S.; Walker, W.; Selby, J.; Glerum, D.M.; et al. Fatal infantile cardioencephalomyopathy with COX deficiency and mutations in SCO2, a COX assembly gene. Nat. Genet. 1999, 23, 333–337. [Google Scholar] [CrossRef] [PubMed]
  22. Idris, M.; Mitchell, D.J.; Gordon, R.; Sidharthan, N.P.; Butcher, N.J.; Minchin, R.F. Interaction of the Brain-Selective Sulfotransferase SULT4A1 with Other Cytosolic Sulfotransferases: Effects on Protein Expression and Function. Drug Metab. Dispos. Biol. Fate Chem. 2020, 48, 337–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Hossain, M.I.; Marcus, J.M.; Lee, J.H.; Garcia, P.L.; Gagné, J.P.; Poirier, G.G.; Falany, C.N.; Andrabi, S.A. SULT4A1 Protects Against Oxidative-Stress Induced Mitochondrial Dysfunction in Neuronal Cells. Drug Metab. Dispos. Biol. Fate Chem. 2019, 47, 949–953. [Google Scholar] [CrossRef] [PubMed]
  24. Garcia, P.L.; Hossain, M.I.; Andrabi, S.A.; Falany, C.N. Generation and Characterization of SULT4A1 Mutant Mouse Models. Drug Metab. Dispos. Biol. Fate Chem. 2018, 46, 41–45. [Google Scholar] [CrossRef] [Green Version]
  25. Cores, Á.; Piquero, M.; Villacampa, M.; León, R.; Menéndez, J.C. NRF2 Regulation Processes as a Source of Potential Drug Targets against Neurodegenerative Diseases. Biomolecules 2020, 10, 904. [Google Scholar] [CrossRef] [PubMed]
  26. Kim, M.J.; Jeon, J.H. Recent Advances in Understanding Nrf2 Agonism and Its Potential Clinical Application to Metabolic and Inflammatory Diseases. Int. J. Mol. Sci. 2022, 23, 2846. [Google Scholar] [CrossRef] [PubMed]
  27. Li, Y.; Jia, X.; Wu, H.; Xun, G.; Ou, J.; Zhang, Q.; Li, H.; Bai, T.; Hu, Z.; Zou, X.; et al. Genotype and phenotype correlations for SHANK3 de novo mutations in neurodevelopmental disorders. Am. J. Med. Genet. Part A 2018, 176, 2668–2676. [Google Scholar] [CrossRef] [PubMed]
  28. Almandil, N.B.; Alkuroud, D.N.; AbdulAzeez, S.; AlSulaiman, A.; Elaissari, A.; Borgio, J.F. Environmental and Genetic Factors in Autism Spectrum Disorders: Special Emphasis on Data from Arabian Studies. Int. J. Environ. Res. Public Health 2019, 16, 658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Tzanoulinou, S.; Musardo, S.; Contestabile, A.; Bariselli, S.; Casarotto, G.; Magrinelli, E.; Jiang, Y.H.; Jabaudon, D.; Bellone, C. Inhibition of Trpv4 rescues circuit and social deficits unmasked by acute inflammatory response in a Shank3 mouse model of Autism. Mol. Psychiatry 2022, 1–15. [Google Scholar] [CrossRef] [PubMed]
  30. Qvist, P.; Christensen, J.H.; Vardya, I.; Rajkumar, A.P.; Mørk, A.; Paternoster, V.; Füchtbauer, E.M.; Pallesen, J.; Fryland, T.; Dyrvig, M.; et al. The Schizophrenia-Associated BRD1 Gene Regulates Behavior, Neurotransmission, and Expression of Schizophrenia Risk Enriched Gene Sets in Mice. Biol. Psychiatry 2017, 82, 62–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Paternoster, V.; Edhager, A.V.; Qvist, P.; Donskov, J.G.; Shliaha, P.; Jensen, O.N.; Mors, O.; Nielsen, A.L.; Børglum, A.D.; Palmfeldt, J.; et al. Inactivation of the Schizophrenia-associated BRD1 gene in Brain Causes Failure-to-thrive, Seizure Susceptibility and Abnormal Histone H3 Acetylation and N-tail Clipping. Mol. Neurobiol. 2021, 58, 4495–4505. [Google Scholar] [CrossRef] [PubMed]
  32. Schenkel, L.C.; Aref-Eshghi, E.; Rooney, K.; Kerkhof, J.; Levy, M.A.; McConkey, H.; Rogers, R.C.; Phelan, K.; Sarasua, S.M.; Jain, L.; et al. DNA methylation epi-signature is associated with two molecularly and phenotypically distinct clinical subtypes of Phelan-McDermid syndrome. Clin. Epigenet. 2021, 13, 2. [Google Scholar] [CrossRef]
  33. Thomford, N.E.; Dzobo, K.; Yao, N.A.; Chimusa, E.; Evans, J.; Okai, E.; Kruszka, P.; Muenke, M.; Awandare, G.; Wonkam, A.; et al. Genomics and Epigenomics of Congenital Heart Defects: Expert Review and Lessons Learned in Africa. Omics J. Integr. Biol. 2018, 22, 301–321. [Google Scholar] [CrossRef] [PubMed]
  34. Tseng, C.J.; McDougle, C.J.; Hooker, J.M.; Zürcher, N.R. Epigenetics of Autism Spectrum Disorder: Histone Deacetylases. Biol. Psychiatry 2021. [Google Scholar] [CrossRef] [PubMed]
  35. Isaksson, J.; Ruchkin, V.; Aho, N.; Lundin Remnélius, K.; Marschik, P.B.; Bölte, S. Nonshared environmental factors in the aetiology of autism and other neurodevelopmental conditions: A monozygotic co-twin control study. Mol. Autism 2022, 13, 8. [Google Scholar] [CrossRef] [PubMed]
  36. Dyar, B.; Meaddough, E.; Sarasua, S.M.; Rogers, C.; Phelan, K.; Boccuto, L. Genetic Findings as the Potential Basis of Personalized Pharmacotherapy in Phelan-McDermid Syndrome. Genes 2021, 12, 1192. [Google Scholar] [CrossRef] [PubMed]
  37. Taylor, C.; Crosby, I.; Yip, V.; Maguire, P.; Pirmohamed, M.; Turner, R.M. A Review of the Important Role of CYP2D6 in Pharmacogenomics. Genes 2020, 11, 1295. [Google Scholar] [CrossRef] [PubMed]
  38. Rice, D.; Barone, S. Critical periods of vulnerability for the developing nervous system: Evidence from humans and animal models. Environ. Health Perspect. 2000, 108 (Suppl. 3), 511–533. [Google Scholar] [CrossRef] [PubMed]
  39. Basu, A.; Mestres, I.; Sahu, S.K.; Tiwari, N.; Khongwir, B.; Baumgart, J.; Singh, A.; Calegari, F.; Tiwari, V.K. Phf21b imprints the spatiotemporal epigenetic switch essential for neural stem cell differentiation. Genes Dev. 2020, 34, 1190–1209. [Google Scholar] [CrossRef] [PubMed]
Table 1. Possible contributions of environmental factors to Phelan–McDermid syndrome clinical presentation.
Table 1. Possible contributions of environmental factors to Phelan–McDermid syndrome clinical presentation.
Gene (s)Environmental Factor (s)MechanismReferences
PNPLA3, PPARADiet, drugs, inflammationHemizygous variants or complete loss of protein lead to liver disease, worsened by high-calorie foods, alcohol, drugs, and inflammation [14,16,17]
SCO2, TYMP, SULT4A1Diet, oxidantsHaploinsufficiency or hemizygous variants disrupt mitochondrial function, which can also be affected by certain nutrients/supplements and oxidants[7,9,10,11,18,23]
SHANK3InflammationLoss of a copy plus inflammation lead to hyperexcitability of medium spiny neurons[28,29]
BRD1DrugsLoss of a copy alters epigenomic regulation, which is also influenced by certain drugs [32,33,34,35]
CYP2D6DrugsInteractions with other components of cytochrome p450 affect responses to several drugs [37]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Boccuto, L.; Mitz, A.; Abenavoli, L.; Sarasua, S.M.; Bennett, W.; Rogers, C.; DuPont, B.; Phelan, K. Phenotypic Variability in Phelan–McDermid Syndrome and Its Putative Link to Environmental Factors. Genes 2022, 13, 528. https://doi.org/10.3390/genes13030528

AMA Style

Boccuto L, Mitz A, Abenavoli L, Sarasua SM, Bennett W, Rogers C, DuPont B, Phelan K. Phenotypic Variability in Phelan–McDermid Syndrome and Its Putative Link to Environmental Factors. Genes. 2022; 13(3):528. https://doi.org/10.3390/genes13030528

Chicago/Turabian Style

Boccuto, Luigi, Andrew Mitz, Ludovico Abenavoli, Sara M. Sarasua, William Bennett, Curtis Rogers, Barbara DuPont, and Katy Phelan. 2022. "Phenotypic Variability in Phelan–McDermid Syndrome and Its Putative Link to Environmental Factors" Genes 13, no. 3: 528. https://doi.org/10.3390/genes13030528

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop