Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Behav Res Ther ; 178: 104553, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38728832

ABSTRACT

Previous studies showed that glucose has beneficial effects on memory function and can enhance contextual fear learning. To derive potential therapeutic interventions, further research is needed regarding the effects of glucose on fear extinction. In two experimental studies with healthy participants (Study 1: N = 68, 39 females; Study 2: N = 89, 67 females), we investigated the effects of glucose on fear extinction learning and its consolidation. Participants completed a differential fear conditioning paradigm consisting of acquisition, extinction, and return of fear tests: reinstatement, and extinction recall. US-expectancy ratings, skin conductance response (SCR), and fear potentiated startle (FPS) were collected. Participants were pseudorandomized and double-blinded to one of two groups: They received either a drink containing glucose or saccharine 20 min before (Study 1) or immediately after extinction (Study 2). The glucose group showed a significantly stronger decrease in differential FPS during extinction (Study 1) and extinction recall (Study 2). Additionally, the glucose group showed a significantly lower contextual anxiety at test of reinstatement (Study 2). Our findings provide first evidence that glucose supports the process of fear extinction, and in particular the consolidation of fear extinction memory, and thus has potential as a beneficial adjuvant to extinction-based treatments. Registered through the German Clinical Trials Registry (https://www.bfarm.de/EN/BfArM/Tasks/German-Clinical-Trials-Register/_node.html; Study 1: DRKS00010550; Study 2: DRKS00018933).


Subject(s)
Conditioning, Classical , Extinction, Psychological , Fear , Galvanic Skin Response , Glucose , Humans , Extinction, Psychological/drug effects , Fear/drug effects , Fear/psychology , Female , Male , Adult , Young Adult , Double-Blind Method , Conditioning, Classical/drug effects , Galvanic Skin Response/drug effects , Reflex, Startle/drug effects , Reflex, Startle/physiology , Adolescent , Mental Recall/drug effects
2.
Genesis ; 62(1): e23580, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37974491

ABSTRACT

Bop1 can promote cell proliferation and is a component of the Pes1-Bop1-WDR12 (PeBoW) complex that regulates ribosomal RNA processing and biogenesis. In embryos, however, bop1 mRNA is highly enriched in the neural plate, cranial neural crest and placodes, and potentially may interact with Six1, which also is expressed in these tissues. Recent work demonstrated that during development, Bop1 is required for establishing the size of the tadpole brain, retina and cranial cartilages, as well as controlling neural tissue gene expression levels. Herein, we extend this work by assessing the effects of Bop1 knockdown at neural plate and larval stages. Loss of Bop1 expanded neural plate gene expression domains (sox2, sox11, irx1) and reduced neural crest (foxd3, sox9), placode (six1, sox11, irx1, sox9) and epidermal (dlx5) expression domains. At larval stages, Bop1 knockdown reduced the expression of several otic vesicle genes (six1, pax2, irx1, sox9, dlx5, otx2, tbx1) and branchial arch genes that are required for chondrogenesis (sox9, tbx1, dlx5). The latter was not the result of impaired neural crest migration. Together these observations indicate that Bop1 is a multifunctional protein that in addition to its well-known role in ribosomal biogenesis functions during early development to establish the craniofacial precursor domains.


Subject(s)
Neural Crest , Transcription Factors , Neural Crest/metabolism , Transcription Factors/metabolism , Head , Skull/metabolism , Ribosomes/metabolism , Gene Expression Regulation, Developmental
3.
Curr Opin Pediatr ; 35(6): 620-630, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37791807

ABSTRACT

PURPOSE OF REVIEW: This review will focus on the current knowledge of the diagnosis and management of overgrowth syndromes with specific focus on mosaic conditions and treatment strategies. RECENT FINDINGS: With the implementation of massively parallel sequencing, the genetic etiology of many classically described overgrowth syndromes have been identified. More recently, the role of mosaic genetic changes has been well described in numerous syndromes. Furthermore, the role of imprinting and methylation, especially of the 11p15 region, has been shown to be instrumental for growth. Perhaps most importantly, many overgrowth syndromes carry an increased risk of neoplasm formation especially in the first 10 years of life and possibly beyond. The systematic approach to the child with overgrowth will aide in timely diagnosis and efficiently align them with appropriate screening strategies. In some cases, precision medical interventions are available to target the perturbed growth signaling pathways. SUMMARY: The systematic approach to the child with overgrowth aids in the standardization of the diagnostic pathway for these young patients, thereby expediting the diagnostic timeline, enabling rigorous monitoring, and delivering tailored therapeutic interventions.


Subject(s)
Signal Transduction , Child , Humans , Syndrome
4.
Dev Dyn ; 252(12): 1407-1427, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37597164

ABSTRACT

BACKGROUND: Members of the sulfotransferase superfamily (SULT) influence the activity of a wide range of hormones, neurotransmitters, metabolites and xenobiotics. However, their roles in developmental processes are not well characterized even though they are expressed during embryogenesis. We previously found in a microarray screen that Six1 up-regulates LOC100037047, which encodes XB5850668.L, an uncharacterized sulfotransferase. RESULTS: Since Six1 is required for patterning the embryonic ectoderm into its neural plate, neural crest, preplacodal and epidermal domains, we used loss- and gain-of function assays to characterize the role of XB5850668.L during this process. Knockdown of endogenous XB5850668.L resulted in the reduction of epidermal, neural crest, cranial placode and otic vesicle gene expression domains, concomitant with neural plate expansion. Increased levels had minimal effects, but infrequently expanded neural plate and neural crest gene domains, and infrequently reduced cranial placode and otic vesicle gene domains. Mutation of two key amino acids in the sulfotransferase catalytic domain required for PAPS binding and enzymatic activity tended to reduce the effects of overexpressing the wild-type protein. CONCLUSIONS: Our analyses indicates that XB5850668.L is a member of the SULT2 family that plays important roles in patterning the embryonic ectoderm. Some aspects of its influence likely depend on sulfotransferase activity.


Subject(s)
Ectoderm , Neural Crest , Neural Crest/metabolism , Skull/metabolism , Embryonic Development/genetics , Sulfotransferases/genetics , Sulfotransferases/metabolism , Gene Expression Regulation, Developmental
5.
Cancers (Basel) ; 15(9)2023 Apr 29.
Article in English | MEDLINE | ID: mdl-37174013

ABSTRACT

Patients with Beckwith-Wiedemann syndrome (BWS), an epigenetic imprinting disorder involving alterations in genes at the 11p15 chromosomal location, are predisposed to develop hepatoblastomas (HBs), which are rare embryonal liver tumors. Tumors can develop after a BWS diagnosis or, conversely, can be the presenting feature leading to a subsequent diagnosis. While HBs are the cardinal tumors of BWS, not all patients with the BWS spectrum will develop HBs. This observation has led to many hypotheses, including genotype-associated risk, tissue mosaicism, and tumor-specific second hits. To explore these hypotheses, we present the largest cohort of patients with BWS and HBs to date. Our cohort comprised 16 cases, and we broadened our sample size by searching the literature for all cases of BWS with HBs. From these isolated case studies, we amassed another 34 cases, bringing the total number to 50 cases of BWS-HB. We observed that paternal uniparental isodisomy (upd(11)pat) was the most common genotype, representing 38% of cases. The next most common genotype was IC2 LOM, representing 14% of cases. Five patients had clinical BWS without a molecular diagnosis. To investigate the potential mechanism of HBs in BWS, we analyzed normal liver and HB samples from eight cases and isolated tumor samples from another two cases. These samples underwent methylation testing, and 90% of our tumor samples underwent targeted cancer next-generation sequencing (NGS) panels. These matched samples provided novel insights into the oncogenesis of HBs in BWS. We found that 100% of the HBs that underwent NGS panel testing had variants in the CTNNB1 gene. We further identified three distinct groups of BWS-HB patients based on epigenotype. We also demonstrated epigenotype mosaicism, where 11p15 alterations can differ between the blood, HB, and normal liver. In light of this epigenotype mosaicism, tumor risk assessment based on blood profiling may not be accurate. Therefore, universal screening is recommended for all patients with BWS.

6.
Int J Mol Sci ; 24(6)2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36982709

ABSTRACT

Point mutations in the ß2 (N265S) and ß3 (N265M) subunits of γ-amino butyric acid type A receptors (GABAARs) that render them insensitive to the general anesthetics etomidate and propofol have been used to link modulation of ß2-GABAARs to sedation and ß3-GABAARs to surgical immobility. These mutations also alter GABA sensitivity, and mice carrying the ß3-N265M mutation have been reported to have impaired baseline memory. Here, we tested the effects of the ß2-N265M and ß3-N265M mutations on memory, movement, hotplate sensitivity, anxiety, etomidate-induced sedation, and intrinsic kinetics. We found that both ß2-N265M and ß3-N265M mice exhibited baseline deficits in the Context Preexposure Facilitation Effect learning paradigm. Exploratory activity was slightly greater in ß2-N265M mice, but there were no changes in either genotype in anxiety or hotplate sensitivity. ß2-N265M mice were highly resistant to etomidate-induced sedation, and heterozygous mice were partially resistant. In rapid solution exchange experiments, both mutations accelerated deactivation two- to three-fold compared to wild type receptors and prevented modulation by etomidate. This degree of change in the receptor deactivation rate is comparable to that produced by an amnestic dose of etomidate but in the opposite direction, indicating that intrinsic characteristics of GABAARs are optimally tuned under baseline conditions to support mnemonic function.


Subject(s)
Etomidate , Propofol , Mice , Animals , Etomidate/pharmacology , Point Mutation , Receptors, GABA-A/genetics , Propofol/pharmacology , gamma-Aminobutyric Acid/genetics
7.
Front Cell Dev Biol ; 10: 786052, 2022.
Article in English | MEDLINE | ID: mdl-35198557

ABSTRACT

The embryonic ectoderm is composed of four domains: neural plate, neural crest, pre-placodal region (PPR) and epidermis. Their formation is initiated during early gastrulation by dorsal-ventral and anterior-posterior gradients of signaling factors that first divide the embryonic ectoderm into neural and non-neural domains. Next, the neural crest and PPR domains arise, either via differential competence of the neural and non-neural ectoderm (binary competence model) or via interactions between the neural and non-neural ectoderm tissues to produce an intermediate neural border zone (NB) (border state model) that subsequently separates into neural crest and PPR. Many previous gain- and loss-of-function experiments demonstrate that numerous TFs are expressed in initially overlapping zones that gradually resolve into patterns that by late neurula stages are characteristic of each of the four domains. Several of these studies suggested that this is accomplished by a combination of repressive TF interactions and competence to respond to local signals. In this study, we ectopically expressed TFs that at neural plate stages are characteristic of one domain in a different domain to test whether they act cell autonomously as repressors. We found that almost all tested TFs caused reduced expression of the other TFs. At gastrulation these effects were strictly within the lineage-labeled cells, indicating that the effects were cell autonomous, i.e., due to TF interactions within individual cells. Analysis of previously published single cell RNAseq datasets showed that at the end of gastrulation, and continuing to neural tube closure stages, many ectodermal cells express TFs characteristic of more than one neural plate stage domain, indicating that different TFs have the opportunity to interact within the same cell. At neurula stages repression was observed both in the lineage-labeled cells and in adjacent cells not bearing detectable lineage label, suggesting that cell-to-cell signaling has begun to contribute to the separation of the domains. Together, these observations directly demonstrate previous suggestions in the literature that the segregation of embryonic ectodermal domains initially involves cell autonomous, repressive TF interactions within an individual cell followed by the subsequent advent of non-cell autonomous signaling to neighbors.

8.
Article in English | MEDLINE | ID: mdl-34697083

ABSTRACT

Beckwith-Wiedemann syndrome (BWS) is a rare overgrowth disorder caused by epigenetic alterations on Chromosome 11p15.5. Most molecular changes are sporadic and are thought to occur in a mosaic pattern. Thereby, the distribution of affected cells differs between tissues for each individual, which can complicate genotype-phenotype correlations. In two of the BWS molecular subtypes, tissue mosaicism has been demonstrated; however, mosaicism has not been specifically studied in the most common cause of BWS, loss of methylation (LOM) at KCNQ1OT1:TSS differentially methylated region (DMR) imprinting center 2 (IC2) LOM. The increased prevalence of twinning associated with the IC2 LOM subtype and the discordant phenotypes between the twins previously led to the proposal of diffused epigenetic mosaicism, leading to asymmetric distribution of affected cells during embryonic development. In this study, we evaluated the level of methylation detected in 64 samples collected from 30 individuals with IC2 LOM. We demonstrate that the IC2 LOM defect can occur in mosaic and nonmosaic patterns, and tissues from the same individual can show variable patterns, which suggests that this asymmetric distribution occurs during development. We further suggest that the clinical phenotype in individuals with BWS IC2 LOM is correlated with the epigenetic burden of affected cells in each tissue type. This series is the first report to demonstrate tissue mosaicism within the IC2 LOM epigenotype, and consideration of this mosaicism is necessary to understanding the pathogenesis of BWS.


Subject(s)
Beckwith-Wiedemann Syndrome , Beckwith-Wiedemann Syndrome/genetics , DNA Methylation , Female , Genomic Imprinting , Humans , Mosaicism , Phenotype , Pregnancy
9.
Sci Rep ; 11(1): 11295, 2021 05 28.
Article in English | MEDLINE | ID: mdl-34050248

ABSTRACT

MBD5-associated neurodevelopmental disorder (MAND) is an autism spectrum disorder (ASD) characterized by intellectual disability, motor delay, speech impairment and behavioral problems; however, the biological role of methyl-CpG-binding domain 5, MBD5, in neurodevelopment and ASD remains largely undefined. Hence, we created neural progenitor cells (NPC) derived from individuals with chromosome 2q23.1 deletion and conducted RNA-seq to identify differentially expressed genes (DEGs) and the biological processes and pathways altered in MAND. Primary skin fibroblasts from three unrelated individuals with MAND and four unrelated controls were converted into induced pluripotent stem cell (iPSC) lines, followed by directed differentiation of iPSC to NPC. Transcriptome analysis of MAND NPC revealed 468 DEGs (q < 0.05), including 20 ASD-associated genes. Comparison of DEGs in MAND with SFARI syndromic autism genes revealed a striking significant overlap in biological processes commonly altered in neurodevelopmental phenotypes, with TGFß, Hippo signaling, DNA replication, and cell cycle among the top enriched pathways. Overall, these transcriptome deviations provide potential connections to the overlapping neurocognitive and neuropsychiatric phenotypes associated with key high-risk ASD genes, including chromatin modifiers and epigenetic modulators, that play significant roles in these disease states.


Subject(s)
Autism Spectrum Disorder/genetics , DNA-Binding Proteins/genetics , Neurodevelopmental Disorders/genetics , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Autism Spectrum Disorder/metabolism , Autistic Disorder/genetics , Cell Differentiation/genetics , Chromosome Deletion , Chromosomes, Human, Pair 2/genetics , Chromosomes, Human, Pair 2/metabolism , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , DNA-Binding Proteins/metabolism , Gene Expression/genetics , Gene Expression Profiling/methods , Humans , Induced Pluripotent Stem Cells/metabolism , Intellectual Disability/genetics , Intellectual Disability/metabolism , Neural Stem Cells/metabolism , Neurodevelopmental Disorders/metabolism , Phenotype , Primary Cell Culture , RNA-Seq , Signal Transduction/genetics , Transcriptome/genetics
10.
J Shoulder Elbow Surg ; 30(1): 113-119, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32807371

ABSTRACT

BACKGROUND: Despite rapid increases in the demand for total shoulder arthroplasty, data describing cost trends are scarce. We aim to (1) describe variation in the cost of shoulder arthroplasty performed by different surgeons at multiple hospitals and (2) determine the driving factors of such variation. METHODS: A standardized, highly accurate cost accounting method, time-driven activity-based costing, was used to determine the cost of 1571 shoulder arthroplasties performed by 12 surgeons at 4 high-volume institutions between 2016 and 2018. Costs were broken down into supply costs (including implant price and consumables) and personnel costs, including physician fees. Cost parameters were compared with total cost for surgical episodes and case volume. RESULTS: Across 4 institutions and 12 surgeons, surgeon volume and hospital volume did not correlate with episode-of-care cost. Average cost per case of each institution varied by factors of 1.6 (P = .47) and 1.7 (P = .06) for anatomic total shoulder arthroplasty (TSA) and reverse total shoulder arthroplasty (RSA), respectively. Implant (56% and 62%, respectively) and personnel costs from check-in through the operating room (21% and 17%, respectively) represented the highest percentages of cost and highly correlated with the cost of the episode of care for TSA and RSA. CONCLUSIONS: Variation in episode-of-care total costs for both TSA and RSA had no association with hospital or surgeon case volume at 4 high-volume institutions but was driven primarily by variation in implant and personnel costs through the operating room. This analysis does not address medium- or long-term costs.


Subject(s)
Arthroplasty, Replacement, Shoulder , Orthopedic Surgeons/economics , Shoulder Joint , Arthroplasty, Replacement, Shoulder/economics , Arthroplasty, Replacement, Shoulder/instrumentation , Arthroplasty, Replacement, Shoulder/statistics & numerical data , Costs and Cost Analysis , Economics, Hospital/statistics & numerical data , Episode of Care , Hospital Costs/statistics & numerical data , Hospitals/statistics & numerical data , Hospitals, High-Volume/statistics & numerical data , Humans , Orthopedic Surgeons/statistics & numerical data , Retrospective Studies , Shoulder Joint/surgery , Shoulder Prosthesis/economics , United States/epidemiology
11.
Dev Biol ; 462(2): 165-179, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32259520

ABSTRACT

Xenopus laevis frogs from laboratory stocks normally lay eggs exhibiting extensive size variability. We find that these initial size differences subsequently affect the size of the embryos prior to the onset of growth, and the size of tadpoles during the growth period. Even though these tadpoles differ in size, their tissues, organs, and structures always seem to be properly proportioned, i.e. they display static allometry. Initial axial patterning events in Xenopus occur in a spherical embryo, allowing easy documentation of their size-dependent features. We examined the size distribution of early Xenopus laevis embryos and measured diameters that differed by about 38% with a median of about 1.43 â€‹mm. This range of embryo sizes corresponds to about a 1.9-fold difference in surface area and a 2.6-fold difference in volume. We examined the relationship between embryo size and gene expression and observed a significant correlation between diameter and RNA content during gastrula stages. In addition, we investigated the expression levels of genes that pattern the mesoderm, induce the nervous system and mediate the progression of ectodermal cells to neural precursors in large and small embryos. We found that most of these factors were expressed at levels that scaled with the different embryo sizes and total embryo RNA content. In agreement with the changes in transcript levels, the expression domains in larger embryos increased proportionally with the increase in surface area, maintaining their relative expression domain size in relation to the total size of the embryo. Thus, our study identified a mechanism for adapting gene expression domains to embryo size by adjusting the transcript levels of the genes regulating mesoderm induction and patterning. In the neural plate, besides the scaling of the expression domains, we observed similar cell sizes and cell densities in small and large embryos suggesting that additional cell divisions took place in large embryos to compensate for the increased size. Our results show in detail the size variability among Xenopus laevis embryos and the transcriptional adaptation to scale gene expression with size. The observations further support the involvement of BMP/ADMP signaling in the scaling process.


Subject(s)
Body Patterning/physiology , Gene Expression Regulation, Developmental/genetics , Morphogenesis/physiology , Animals , Bone Morphogenetic Proteins/metabolism , Cell Size , Embryo, Nonmammalian/metabolism , Embryonic Development/physiology , Gastrula/metabolism , Gene Expression/genetics , Gene Expression Regulation, Developmental/physiology , Mesoderm/metabolism , Morphogenesis/genetics , Signal Transduction/physiology , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/metabolism
12.
Microrna ; 9(1): 70-80, 2020.
Article in English | MEDLINE | ID: mdl-31232238

ABSTRACT

BACKGROUND: We have previously described mosaic mutations in the RNase IIIb domain of DICER1that display global developmental delays, lung cysts, somatic overgrowth, macrocephaly and Wilms tumor. This constellation of phenotypes was classified as GLOW syndrome. Due to the phenotypic overlap between GLOW and syndromes caused by mutations in the PI3K/AKT/mTOR pathway, we hypothesized that alterations in miRNA regulation of this pathway cause its specific constellation of phenotypes. OBJECTIVE: To test the hypothesis that DICER1 "hot spot" mutations associated with GLOW syndrome activate PI3K/AKT/mTOR signaling. METHODS: We developed HEK293T cells with loss of exon 25 in DICER1, a genetic modification that is synonymous with the "hot spot" RNAseIIIb mutations that cause GLOW syndrome. We assayed the cells for activation of the PI3K/AKT/mTOR signaling pathway. RESULTS: We observed activation of the PI3K/AKT/mTOR pathway as demonstrated by increased pS6Kinase, p4EBP1 and pTSC2 levels. Additionally, these cells demonstrate a striking cellular phenotype, with the ability to form spheres when the serum is removed from their growth medium. The cells in these spheres are Oct4 and Sox2 positive and exhibit the property of reversion with the addition of serum. We queried miRNA expression data and identified a population of miRNAs that increase due to these mutations and target negative regulators of the PI3K/AKT/mTOR pathway. CONCLUSION: This work identifies the delicate and essential role for miRNA control of the PI3K/AKT/mTOR pathway. We conclude that the phenotypes observed in the GLOW syndrome are the result of PI3K/AKT/mTOR activation.


Subject(s)
DEAD-box RNA Helicases/genetics , Kidney Neoplasms/genetics , Megalencephaly/genetics , MicroRNAs/genetics , Ribonuclease III/genetics , Wilms Tumor/genetics , Cell Line , HEK293 Cells , Humans , Kidney Neoplasms/pathology , Megalencephaly/pathology , Octamer Transcription Factor-3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases/metabolism , SOXB1 Transcription Factors/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein/metabolism , Wilms Tumor/pathology
13.
Am J Med Genet A ; 179(12): 2517-2531, 2019 12.
Article in English | MEDLINE | ID: mdl-31639285

ABSTRACT

The hedgehog (Hh) pathway is highly conserved and required for embryonic patterning and determination. Mutations in the Hh pathway are observed in sporadic tumors as well as under syndromic conditions. Common to these syndromes are the findings of polydactyly/syndactyly and brain overgrowth. The latter is also a finding most commonly observed in the cases of mutations in the PI3K/AKT/mTOR pathway. We have identified novel Hh pathway mutations and structural copy number variations in individuals with somatic overgrowth, macrocephaly, dysmorphic facial features, and developmental delay, which phenotypically closely resemble patients with phosphatase and tensin homolog (PTEN) mutations. We hypothesized that brain overgrowth and phenotypic overlap with syndromic overgrowth syndromes in these cases may be due to crosstalk between the Hh and PI3K/AKT/mTOR pathways. To test this, we modeled disease-associated variants by generating PTCH1 and Suppressor of Fused (SUFU) heterozygote cell lines using the CRISPR/Cas9 system. These cells demonstrate activation of PI3K signaling and increased phosphorylation of its downstream target p4EBP1 as well as a distinct cellular phenotype. To further investigate the mechanism underlying this crosstalk, we treated human neural stem cells with sonic hedgehog (SHH) ligand and performed transcriptional analysis of components of the mTOR pathway. These studies identified decreased expression of a set of mTOR negative regulators, leading to its activation. We conclude that there is a significant crosstalk between the SHH and PI3K/AKT/mTOR. We propose that this crosstalk is responsible for why mutations in PTCH1 and SUFU lead to macrocephaly phenotypes similar to those observed in PTEN hamartoma and other overgrowth syndromes associated with mutations in PI3K/AKT/mTOR pathway genes.


Subject(s)
Hedgehog Proteins/metabolism , Megalencephaly/genetics , Megalencephaly/metabolism , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , CRISPR-Cas Systems , Cell Line , Child, Preschool , Female , Gene Deletion , Haploinsufficiency , Humans , Infant , Male , Megalencephaly/diagnosis , Models, Biological , Neural Stem Cells
14.
Mol Genet Genomic Med ; 7(2): e00501, 2019 02.
Article in English | MEDLINE | ID: mdl-30447054

ABSTRACT

BACKGROUND: The cohesin complex is a multi-subunit protein complex which regulates sister chromatid cohesion and separation during cellular division. In addition, this evolutionarily conserved protein complex plays an integral role in DNA replication, DNA repair, and the regulation of transcription. The core complex is composed of four subunits: RAD21, SMC1A, SMC3, and STAG1/2. Mutations in these proteins have been implicated in human developmental disorders collectively termed "cohesinopathies." METHODS: Using clinical exome sequencing, we have previously identified three female cases with heterozygous STAG2 mutations and overlapping syndromic phenotypes. Subsequently, a familial missense variant was identified in five male family members. RESULTS: We now present the case of a 4-year-old male with developmental delay, failure to thrive, short stature, and polydactyly with a likely pathogenic STAG2 de novo missense hemizygous variant, c.3027A>T, p.Lys1009Asn. Furthermore, we compare the phenotypes of the four previously reported STAG2 variants with our case. CONCLUSION: We conclude that mutations in STAG2 cause a novel constellation of sex-specific cohesinopathy-related phenotypes and are furthermore, essential for neurodevelopment, human growth, and behavioral development.


Subject(s)
Antigens, Nuclear/genetics , Developmental Disabilities/genetics , Genetic Diseases, X-Linked/genetics , Growth Disorders/genetics , Phenotype , Polydactyly/genetics , Cell Cycle Proteins , Child, Preschool , Developmental Disabilities/pathology , Genetic Diseases, X-Linked/pathology , Growth Disorders/pathology , Humans , Male , Mutation, Missense , Polydactyly/pathology , Syndrome
15.
Child Psychiatry Hum Dev ; 49(6): 897-905, 2018 12.
Article in English | MEDLINE | ID: mdl-29681000

ABSTRACT

We investigated the role of self-reports and behavioral measures of interpretation biases and their content-specificity in children with varying levels of spider fear and/or social anxiety. In total, 141 selected children from a community sample completed an interpretation bias task with scenarios that were related to either spider threat or social threat. Specific interpretation biases were found; only spider-related interpretation bias and self-reported spider fear predicted unique variance in avoidance behavior on the Behavior Avoidance Task for spiders. Likewise, only social-threat related interpretation bias and self-reported social anxiety predicted anxiety during the Social Speech Task. These findings support the hypothesis that fearful children display cognitive biases that are specific to particular fear-relevant stimuli. Clinically, this insight might be used to improve treatments for anxious children by targeting content-specific interpretation biases related to individual disorders.


Subject(s)
Anxiety/psychology , Fear/psychology , Phobia, Social/psychology , Phobic Disorders/psychology , Child , Female , Humans , Male , Self Report
17.
Am J Med Genet A ; 173(5): 1319-1327, 2017 May.
Article in English | MEDLINE | ID: mdl-28296084

ABSTRACT

The cohesin complex is an evolutionarily conserved multi-subunit protein complex which regulates sister chromatid cohesion during mitosis and meiosis. Additionally, the cohesin complex regulates DNA replication, DNA repair, and transcription. The core of the complex consists of four subunits: SMC1A, SMC3, RAD21, and STAG1/2. Loss-of-function mutations in many of these proteins have been implicated in human developmental disorders collectively termed "cohesinopathies." Through clinical exome sequencing (CES) of an 8-year-old girl with a clinical history of global developmental delay, microcephaly, microtia with hearing loss, language delay, ADHD, and dysmorphic features, we describe a heterozygous de novo variant (c.205C>T; p.(Arg69*)) in the integral cohesin structural protein, STAG2. This variant is associated with decreased STAG2 protein expression. The analyses of metaphase spreads did not exhibit premature sister chromatid separation; however, delayed sister chromatid cohesion was observed. To further support the pathogenicity of STAG2 variants, we identified two additional female cases from the DECIPHER research database with mutations in STAG2 and phenotypes similar to our patient. Interestingly, the clinical features of these three cases are remarkably similar to those observed in other well-established cohesinopathies. Herein, we suggest that STAG2 is a dosage-sensitive gene and that heterozygous loss-of-function variants lead to a cohesinopathy.


Subject(s)
Antigens, Nuclear/genetics , Congenital Abnormalities/genetics , Developmental Disabilities/genetics , Microcephaly/genetics , Antigens, Nuclear/biosynthesis , Cell Cycle Proteins/genetics , Child , Chromosomal Proteins, Non-Histone/genetics , Congenital Abnormalities/physiopathology , Developmental Disabilities/physiopathology , Female , Gene Expression Regulation , Heterozygote , Humans , Microcephaly/physiopathology , Cohesins
18.
Sci Rep ; 6: 36916, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27845378

ABSTRACT

The mechanisms by which sex differences in the mammalian brain arise are poorly understood, but are influenced by a combination of underlying genetic differences and gonadal hormone exposure. Using a mouse embryonic neural stem cell (eNSC) model to understand early events contributing to sexually dimorphic brain development, we identified novel interactions between chromosomal sex and hormonal exposure that are instrumental to early brain sex differences. RNA-sequencing identified 103 transcripts that were differentially expressed between XX and XY eNSCs at baseline (FDR = 0.10). Treatment with testosterone-propionate (TP) reveals sex-specific gene expression changes, causing 2854 and 792 transcripts to become differentially expressed on XX and XY genetic backgrounds respectively. Within the TP responsive transcripts, there was enrichment for genes which function as epigenetic regulators that affect both histone modifications and DNA methylation patterning. We observed that TP caused a global decrease in 5-methylcytosine abundance in both sexes, a transmissible effect that was maintained in cellular progeny. Additionally, we determined that TP was associated with residue-specific alterations in acetylation of histone tails. These findings highlight an unknown component of androgen action on cells within the developmental CNS, and contribute to a novel mechanism of action by which early hormonal organization is initiated and maintained.


Subject(s)
Epigenesis, Genetic/drug effects , Testosterone/pharmacology , Transcriptome/drug effects , Acetylation/drug effects , Animals , Cell Lineage/drug effects , DNA Methylation/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Female , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , RNA/chemistry , RNA/isolation & purification , RNA/metabolism , Sequence Analysis, RNA , Sex Characteristics , Sex Chromosomes
19.
Plast Reconstr Surg Glob Open ; 4(9): e1030, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27757346

ABSTRACT

There is an exponential rise of patients with massive weight loss because of bariatric surgery or lifestyle changes. The result is an increase of patients with folds of redundant skin that may cause physical and psychological problems. The lower body lift is a procedure to correct deformities in the abdomen, mons, flanks, lateral thighs, and buttocks. Complication rates are quite high and could negatively affect the positive outcomes. The purpose of this study is to assess complication rates and to identify predictors of complications to optimize outcomes for patients after lower body lift surgery. METHODS: A retrospective analysis of 100 patients who underwent a lower body lift procedure was performed. The patients were reviewed for complications, demographic data, comorbidities, smoking, highest lifetime body mass index, body mass index before lower body lift surgery, percentage of excess weight loss, and amount of tissue excised. RESULTS: The overall complication rate was 78%. Twenty-two percent of the patients had major complications and 56% had minor complications. There is a linear relationship between body mass index before lower body lift surgery and complications (P = 0.03). The percentage of excess weight loss (odds ratio [OR] 0.97; 95% confidence interval [CI] 0.92-1.00), highest lifetime body mass index (OR 1.08; 95% CI 1.01-1.15), body mass index before lower body lift surgery (OR 1.17; 95% CI 1.02-1.33), and smoking (OR 7.74; CI 0.98-61.16) are significantly associated with the development of complications. CONCLUSIONS: This study emphasizes the importance of a good weight status before surgery and cessation of smoking to minimize the risk of complications.

20.
Ann Neurol ; 80(3): 456-60, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27422356

ABSTRACT

Dominant missense mutations in the amyloid ß (Aß) precursor protein (APP) gene have been implicated in early onset Alzheimer disease. These mutations alter protein structure to favor the pathologic production of Aß. We report that homozygous nonsense mutations in APP are associated with decreased somatic growth, microcephaly, hypotonia, developmental delay, thinning of the corpus callosum, and seizures. We compare the phenotype of this case to those reported in mouse models and demonstrate multiple similarities, strengthening the role of amyloid precursor protein in normal brain function and development. Ann Neurol 2016;80:456-460.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Corpus Callosum/pathology , Developmental Disabilities/genetics , Microcephaly/genetics , Muscle Hypotonia/genetics , Seizures/genetics , Consanguinity , Humans , Infant , Male , Mutation , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...