Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Biochem Biophys Res Commun ; 721: 150124, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38776833

ABSTRACT

Prader-Willi syndrome (PWS) is a complex epigenetic disorder caused by the deficiency of paternally expressed genes in chromosome 15q11-q13. This syndrome also includes endocrine dysfunction, leading to short stature, hypogonadism, and obscure hyperphagia. Although recent progress has been made toward understanding the genetic basis for PWS, the molecular mechanisms underlying its pathology in obesity remain unclear. In this study, we examined the adipocytic characteristics of two PWS-induced pluripotent stem cell (iPSC) lines: those with the 15q11-q13 gene deletion (iPWS cells) and those with 15q11-q13 abnormal methylation (M-iPWS cells). The transcript levels of the lipid-binding protein aP2 were decreased in iPWS and M-iPWS adipocytes. Flow-cytometry analysis showed that PWS adipocytes accumulated more lipid droplets than did normal individual adipocytes. Furthermore, glucose uptake upon insulin stimulation was attenuated compared to that in normal adipocytes. Overall, our results suggest a significantly increased lipid content and defective in glucose metabolism in PWS adipocytes.


Subject(s)
Adipocytes , Induced Pluripotent Stem Cells , Prader-Willi Syndrome , Prader-Willi Syndrome/pathology , Prader-Willi Syndrome/metabolism , Prader-Willi Syndrome/genetics , Adipocytes/metabolism , Adipocytes/pathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Glucose/metabolism , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , Fatty Acid-Binding Proteins/metabolism , Fatty Acid-Binding Proteins/genetics , Cell Line , DNA Methylation , Gene Deletion , Lipid Metabolism , Insulin/metabolism
2.
Int J Mol Sci ; 24(14)2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37511433

ABSTRACT

N-acetylglucosamine kinase (NAGK) has been identified as an anchor protein that facilitates neurodevelopment with its non-canonical structural role. Similarly, small nuclear ribonucleoprotein polypeptide N (SNRPN) regulates neurodevelopment and cognitive ability. In our previous study, we revealed the interaction between NAGK and SNRPN in the neuron. However, the precise role in neurodevelopment is elusive. In this study, we investigate the role of NAGK and SNRPN in the axodendritic development of neurons. NAGK and SNRPN interaction is significantly increased in neurons at the crucial stages of neurodevelopment. Furthermore, overexpression of the NAGK and SNRPN proteins increases axodendritic branching and neuronal complexity, whereas the knockdown inhibits neurodevelopment. We also observe the interaction of NAGK and SNRPN with the dynein light-chain roadblock type 1 (DYNLRB1) protein variably during neurodevelopment, revealing the microtubule-associated delivery of the complex. Interestingly, NAGK and SNRPN proteins rescued impaired axodendritic development in an SNRPN depletion model of Prader-Willi syndrome (PWS) patient-derived induced pluripotent stem cell neurons. Taken together, these findings are crucial in developing therapeutic approaches for neurodegenerative diseases.


Subject(s)
Prader-Willi Syndrome , Ribonucleoproteins, Small Nuclear , Humans , Autoantigens/metabolism , Chromosomes, Human, Pair 15/metabolism , Cytoplasmic Dyneins/metabolism , Dyneins/metabolism , Microtubules/metabolism , Neurons/metabolism , Peptides/metabolism , Ribonucleoproteins, Small Nuclear/genetics , snRNP Core Proteins
3.
J Cutan Pathol ; 48(2): 285-289, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32519331

ABSTRACT

Dermatofibrosarcoma protuberans (DFSP) is a rare sarcoma of the skin arising from the dermis. Its location is most commonly presented on the trunk of middle-aged adults and rarely on the face. The characteristic genetic aberration in the form of a reciprocal translocation t(17;22)(q21;q13) or a ring fusing the COL1A1 and PDGFB genes is found in 90% of DFSP. We present a case of a 42-year-old man who presented with a DFSP on the left cheek with foci of myxoid-fibrosarcomatous transformation. A conventional chromosomal analysis revealed a complex karyotype without a supernumerary ring chromosome or a linear translocation t(17;22). Comparative genome hybridization and fluorescence in-situ hybridization revealed the fusion of COL1A1 and PDGFB probes inserted in chromosome 15. This is a unique case of DFSP characterized by a rare body location, unique histopathological features, and novel chromosome COL1A1-PDGFB insertion, and may help guide future diagnostic and patient care modalities.


Subject(s)
Chromosomes, Human, Pair 15 , Facial Neoplasms , Fibrosarcoma , Mutagenesis, Insertional , Oncogene Proteins, Fusion , Skin Neoplasms , Adult , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 17/metabolism , Chromosomes, Human, Pair 22/genetics , Chromosomes, Human, Pair 22/metabolism , Facial Neoplasms/genetics , Facial Neoplasms/metabolism , Facial Neoplasms/pathology , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Humans , Male , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Translocation, Genetic
4.
Neurobiol Dis ; 141: 104879, 2020 07.
Article in English | MEDLINE | ID: mdl-32344153

ABSTRACT

Epilepsy affects millions of individuals worldwide and many cases are pharmacoresistant. Duplication 15q syndrome (Dup15q) is a genetic disorder caused by duplications of the 15q11.2-q13.1 region. Phenotypes include a high rate of pharmacoresistant epilepsy. We developed a Dup15q model in Drosophila melanogaster that recapitulates seizures in Dup15q by over-expressing fly Dube3a or human UBE3A in glial cells, but not neurons, implicating glia in the Dup15q epilepsy phenotype. We compared Dube3a overexpression in glia (repo>Dube3a) versus neurons (elav>Dube3a) using transcriptomics and proteomics of whole fly head extracts. We identified 851 transcripts differentially regulated in repo>Dube3a, including an upregulation of glutathione S-transferase (GST) genes that occurred cell autonomously within glial cells. We reliably measured approximately 2,500 proteins by proteomics, most of which were also quantified at the transcript level. Combined transcriptomic and proteomic analysis revealed an enrichment of 21 synaptic transmission genes downregulated at the transcript and protein in repo>Dube3a indicating synaptic proteins change in a cell non-autonomous manner in repo>Dube3a flies. We identified 6 additional glia originating bang-sensitive seizure lines and found upregulation of GSTs in 4 out of these 6 lines. These data suggest GST upregulation is common among gliopathic seizures and may ultimately provide insight for treating epilepsy.


Subject(s)
Drosophila Proteins/metabolism , Epilepsy/metabolism , Neuroglia/metabolism , Neurons/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Chromosomes, Human, Pair 15/metabolism , Disease Models, Animal , Drosophila melanogaster , Gene Expression Profiling , Humans , Proteome , Proteomics , Transcriptome , Trisomy
5.
J Cutan Pathol ; 47(1): 70-75, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31437301

ABSTRACT

Pigmented epithelioid melanocytoma (PEM) represents a group of rare, heavily pigmented melanocytic tumors encompassing lesions previously designated as "animal-type melanomas" and "epithelioid blue nevi." Despite the association of multiple such tumors in the setting of Carney complex, most cases of PEM occur spontaneously as solitary neoplasms in otherwise healthy patients. PEM may arise in both children and adults, and has a known propensity to spread to the regional lymph nodes. Despite this latter finding, recurrence at the biopsy site or spread beyond the lymph node basin is exceptionally uncommon. Although the molecular basis for PEM continues to be characterized, findings to date suggest that this category of melanocytic neoplasia has genetic alterations distinct from those seen in common nevi, dysplastic nevi, Spitz nevi, and melanoma. Herein, we present an in-depth clinical, histopathologic, and molecular analysis of a case of PEM occurring on the scalp of a young African American girl found to have a novel NTRK3-SCAPER gene fusion.


Subject(s)
Carrier Proteins , Chromosome Aberrations , Chromosomes, Human, Pair 15 , Chromosomes, Human, Pair 17 , Discoidin Domain Receptor 2 , Head and Neck Neoplasms , Nevus, Blue , Oncogene Proteins, Fusion , Carrier Proteins/genetics , Carrier Proteins/metabolism , Child, Preschool , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 17/metabolism , Discoidin Domain Receptor 2/genetics , Discoidin Domain Receptor 2/metabolism , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Nevus, Blue/genetics , Nevus, Blue/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
6.
Cytokine ; 126: 154879, 2020 02.
Article in English | MEDLINE | ID: mdl-31629107

ABSTRACT

Carriers of the human 15q13.3 microdeletion (MD) present with a variable spectrum of neuropathological phenotypes that range from asymptomatic to severe clinical outcomes, suggesting an interplay of genetic and non-genetic factors. The most common 2 MB 15q13.3 MD encompasses six genes (MTMR10, FAN1, TRPM1, KLF13, OTUD7A, and CHRNA7), which are expressed in neuronal and non-neuronal tissues. The nicotinic acetylcholine receptor (nAChR) α7, encoded by CHRNA7, is a key player in the cholinergic anti-inflammatory pathway, and the transcription factor KLF13 is also involved in immune responses. Using a mouse model with a heterozygous deletion of the orthologous region of the human 15q13.3 (Df[h15q13]/+), the present study examined peripheral and central innate immune responses to an acute intraperitoneal (i.p.) injection of the bacteriomimetic, lipopolysaccharide (LPS) (100 µg/kg) in adult heterozygous (Het) and wildtype (WT) mice. Serum levels of inflammatory markers were measured 2 h post injection using a Multiplex assay. In control saline injected animals, all measured cytokines were at or below detection limits, whereas LPS significantly increased serum levels of interleukin 1beta (IL-1ß), tumor necrosis factor alpha (TNF-α), IL-6 and IL-10, but not interferon-γ. There was no effect of genotype but a sexual dimorphic response for TNF-α, with females exhibiting greater LPS-induced TNF-α serum levels than males. In situ hybridization revealed similar increases in LPS-induced c-fos mRNA expression in the dorsal vagal complex in all groups. The hippocampal expression of the pro-inflammatory cytokines was evaluated by real-time quantitative PCR. LPS-treatment resulted in significantly increased mRNA expression for IL-1ß, IL-6, and TNF-α compared to saline controls, with no effect of genotype, but a significant sex-effect was detected for IL-1ß. The present study provided no evidence for interactive effects between the heterozygous 15q13.3 MD and a low-dose LPS immune challenge in innate peripheral or central immune responses, although, sex-differential effects in males and females were detected.


Subject(s)
Chromosome Disorders/metabolism , Cytokines/blood , Gene Expression Regulation/drug effects , Immunity, Innate , Intellectual Disability/metabolism , Seizures/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/blood , Basic Helix-Loop-Helix Transcription Factors/genetics , Chromosome Deletion , Chromosome Disorders/genetics , Chromosome Disorders/immunology , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/immunology , Chromosomes, Human, Pair 15/metabolism , Cytokines/genetics , Disease Models, Animal , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Hippocampus/metabolism , Immunity , Immunity, Innate/drug effects , Immunity, Innate/genetics , In Situ Hybridization , Inflammation/blood , Inflammation/genetics , Intellectual Disability/genetics , Intellectual Disability/immunology , Interferon-gamma/blood , Interferon-gamma/genetics , Interleukin-10/blood , Interleukin-10/genetics , Interleukin-1beta/blood , Interleukin-1beta/genetics , Interleukin-6/blood , Interleukin-6/genetics , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Real-Time Polymerase Chain Reaction , Seizures/genetics , Seizures/immunology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics
7.
Int J Mol Sci ; 20(12)2019 Jun 14.
Article in English | MEDLINE | ID: mdl-31207912

ABSTRACT

The 15q11.2 BP1-BP2 microdeletion (Burnside-Butler) syndrome is an emerging disorder that encompasses four genes (NIPA1, NIPA2, CYFIP1, and TUBGCP5). When disturbed, these four genes can lead to cognitive impairment, language and/or motor delay, psychiatric/behavioral problems (attention-deficit hyperactivity, autism, dyslexia, schizophrenia/paranoid psychosis), ataxia, seizures, poor coordination, congenital anomalies, and abnormal brain imaging. This microdeletion was reported as the most common cytogenetic finding when using ultra-high- resolution chromosomal microarrays in patients presenting for genetic services due to autism with or without additional clinical features. Additionally, those individuals with Prader-Willi or Angelman syndromes having the larger typical 15q11-q13 type I deletion which includes the 15q11.2 BP1-BP2 region containing the four genes, show higher clinical severity than those having the smaller 15q11-q13 deletion where these four genes are intact. Two of the four genes (i.e., NIPA1 and NIPA2) are expressed in the brain and encode magnesium transporters. Magnesium is required in over 300 enzyme systems that are critical for multiple cellular functions, energy expenditure, protein synthesis, DNA transcription, and muscle and nerve function. Low levels of magnesium are found in those with seizures, depression, and acute or chronic brain diseases. Anecdotally, parents have administered magnesium supplements to their children with the 15q11.2 BP1-BP2 microdeletion and have observed improvement in behavior and clinical presentation. These observations require more attention from the medical community and should include controlled studies to determine if magnesium supplements could be a treatment option for this microdeletion syndrome and also for a subset of individuals with Prader-Willi and Angelman syndromes.


Subject(s)
Intellectual Disability/drug therapy , Magnesium/therapeutic use , Chromosome Aberrations , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , Dietary Supplements , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Magnesium/administration & dosage
8.
J Hum Genet ; 64(7): 647-652, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30988409

ABSTRACT

Prader-Willi syndrome (PWS) is a well-known imprinting disorder arising from a loss of paternally imprinted gene(s) at 15q11.2-q13. We report a typical PWS patient with a balanced reciprocal translocation, 46, XY, t(15;19)(q11.2;q13.3). After Illumina whole-genome sequencing, we used BreakDancer-1.45 software to predict candidate breakpoints and manually investigated via the Integrated Genome Viewer. Breakpoint PCR followed by Sanger sequencing determined the t(15;19) breakpoints. We investigated the expression of upstream/centromeric and downstream/telomeric genes of the 15q11.2 breakpoint by reverse transcriptase PCR, using total RNA extracted from the patient's lymphoblasts. Of note, the expression of paternally expressed genes PWAR6, SNORD109A/B, SNORD116, IPW, and PWAR1, downstream of the breakpoint, was abolished. Interestingly, the breakpoint did not destroy protein coding genes or individual snoRNAs. These results indicate that these genes may play a major role in the PWS phenotype.


Subject(s)
Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 19/genetics , Prader-Willi Syndrome/genetics , RNA, Long Noncoding/genetics , Translocation, Genetic , Adolescent , Chromosome Breakpoints , Chromosomes, Human, Pair 15/metabolism , Chromosomes, Human, Pair 19/metabolism , Genomic Imprinting/genetics , Humans , Male , Phenotype , Prader-Willi Syndrome/diagnosis , Prader-Willi Syndrome/metabolism , RNA, Small Nucleolar/biosynthesis , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , Whole Genome Sequencing
9.
PLoS One ; 14(2): e0211799, 2019.
Article in English | MEDLINE | ID: mdl-30707743

ABSTRACT

Genomic imprinting is important for normal brain development and aberrant imprinting has been associated with impaired cognition. We studied the imprinting status in selected imprints (H19, IGF2, SNRPN, PEG3, MEST1, NESPAS, KvDMR, IG-DMR and ZAC1) by pyrosequencing in blood samples from longitudinal cohorts born in 1936 (n = 485) and 1921 (n = 223), and anterior hippocampus, posterior hippocampus, periventricular white matter, and thalamus from brains donated to the Aberdeen Brain Bank (n = 4). MEST1 imprint methylation was related to childhood cognitive ability score (-0.416 95% CI -0.792,-0.041; p = 0.030), with the strongest effect evident in males (-0.929 95% CI -1.531,-0.326; p = 0.003). SNRPN imprint methylation was also related to childhood cognitive ability (+0.335 95%CI 0.008,0.663; p = 0.045). A significant association was also observed for SNRPN methylation and adult crystallised cognitive ability (+0.262 95%CI 0.007,0.517; p = 0.044). Further testing of significant findings in a second cohort from the same region, but born in 1921, resulted in similar effect sizes and greater significance when the cohorts were combined (MEST1; -0.371 95% CI -0.677,-0.065; p = 0.017; SNRPN; +0.361 95% CI 0.079,0.643; p = 0.012). For SNRPN and MEST1 and four other imprints the methylation levels in blood and in the five brain regions were similar. Methylation of the paternally expressed, maternally methylated genes SNRPN and MEST1 in adult blood was associated with cognitive ability in childhood. This is consistent with the known importance of the SNRPN containing 15q11-q13 and the MEST1 containing 7q31-34 regions in cognitive function. These findings, and their sex specific nature in MEST1, point to new mechanisms through which complex phenotypes such as cognitive ability may be inherited. These mechanisms are potentially relevant to both the heritable and non-heritable components of cognitive ability. The process of epigenetic imprinting-within SNRPN and MEST1 in particular-and the factors that influence it, are worthy of further study in relation to the determinants of cognitive ability.


Subject(s)
Brain/metabolism , Cognition/physiology , Genomic Imprinting/physiology , Proteins/metabolism , snRNP Core Proteins/blood , Adult , Aged , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , Chromosomes, Human, Pair 7/genetics , Chromosomes, Human, Pair 7/metabolism , Female , Follow-Up Studies , Humans , Male , Middle Aged , Proteins/genetics , snRNP Core Proteins/genetics
10.
Mol Genet Genomic Med ; 6(5): 811-818, 2018 09.
Article in English | MEDLINE | ID: mdl-30003711

ABSTRACT

BACKGROUND: Prader-Willi syndrome (PWS) is a multisystem genetic disorder characterized by lack of satiety leading to morbid obesity, variable degrees of mental retardation, behavior disorders, short stature, and hypogonadism. The underlying genetic cause for PWS is an imprinting defect resulting from a lack of expression of several paternally inherited genes embedded within the 15q11.2-q13 region. Although the clinical expression of hypogonadism in PWS is variable, there are no known cases of fertility in PWS men. In this paper, we described a pure, nearly diploid seminoma in an apparently 32 year-old infertile man with PWS due to maternal uniparental disomy (UPD) on chromosome 15. The development of a germ cell tumor in this subject was an unanticipated result. The aim of this study was to explore the origin of the germ cell tumor in this PWS male patient. METHODS: To explain the origin of the germ cell tumor (seminoma) in our PWS patient we have characterized the tumor for cell morphology and tumor type by pathological examination (H&E and immuno-stainings), evaluated its karyotype by chromosomal microarray analysis and confirmed its UPD origin by haplotype analysis. In addition, DNA methylation status of the PWS- and H19- imprinting centers in wild-type and affected fibroblasts, patient derived induced pluripotent stem cells (iPSCs), and PWS seminoma were determined by bisulfite DNA colony sequencing. RESULTS: To explain the apparent contradiction between the existence of a germ cell tumor and hypogonadism we first confirmed the germ cell origin of the tumor. Next, we determined the tumor chromosomal composition, and validated the presence of a maternal UPD in all examined cell types from this patient. Finally, we characterized the maternal imprints in the PWS and H19 imprinting centers in the tumor and compared them with patient's fibroblasts and iPSCs derived from them. Unpredictably, methylation was reduced to 50% in the tumor, while preserved in the other cell types. CONCLUSION: We infer from this assay that the loss of methylation in the PWS-IC specifically in the tumor of our patient is most likely a locus-specific event resulting from imprint relaxation rather than from general resetting of the imprints throughout the genome during germ line specification.


Subject(s)
Chromosomes, Human, Pair 15 , DNA Methylation , DNA, Neoplasm , Prader-Willi Syndrome , Seminoma , Testicular Neoplasms , Adult , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Humans , Male , Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/metabolism , Prader-Willi Syndrome/pathology , Seminoma/genetics , Seminoma/metabolism , Seminoma/pathology , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology
12.
Mol Autism ; 9: 6, 2018.
Article in English | MEDLINE | ID: mdl-29423132

ABSTRACT

Background: The inability to analyze gene expression in living neurons from Angelman (AS) and Duplication 15q (Dup15q) syndrome subjects has limited our understanding of these disorders at the molecular level. Method: Here, we use dental pulp stem cells (DPSC) from AS deletion, 15q Duplication, and neurotypical control subjects for whole transcriptome analysis. We identified 20 genes unique to AS neurons, 120 genes unique to 15q duplication, and 3 shared transcripts that were differentially expressed in DPSC neurons vs controls. Results: Copy number correlated with gene expression for most genes across the 15q11.2-q13.1 critical region. Two thirds of the genes differentially expressed in 15q duplication neurons were downregulated compared to controls including several transcription factors, while in AS differential expression was restricted primarily to the 15q region. Here, we show significant downregulation of the transcription factors FOXO1 and HAND2 in neurons from 15q duplication, but not AS deletion subjects suggesting that disruptions in transcriptional regulation may be a driving factor in the autism phenotype in Dup15q syndrome. Downstream analysis revealed downregulation of the ASD associated genes EHPB2 and RORA, both genes with FOXO1 binding sites. Genes upregulated in either Dup15q cortex or idiopathic ASD cortex both overlapped significantly with the most upregulated genes in Dup15q DPSC-derived neurons. Conclusions: Finding a significant increase in both HERC2 and UBE3A in Dup15q neurons and significant decrease in these two genes in AS deletion neurons may explain differences between AS deletion class and UBE3A specific classes of AS mutation where HERC2 is expressed at normal levels. Also, we identified an enrichment for FOXO1-regulated transcripts in Dup15q neurons including ASD-associated genes EHPB2 and RORA indicating a possible connection between this syndromic form of ASD and idiopathic cases.


Subject(s)
Angelman Syndrome/genetics , Chromosome Deletion , Neural Stem Cells/metabolism , Transcriptome , Trisomy/genetics , Angelman Syndrome/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cells, Cultured , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , Dental Pulp/cytology , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
13.
Pediatr Blood Cancer ; 65(4)2018 Apr.
Article in English | MEDLINE | ID: mdl-29286563

ABSTRACT

BACKGROUND: Congenital mesoblastic nephroma (MN) is a rare pediatric renal tumor representing approximately 5% of all pediatric renal tumors. Three different types of MN are distinguished histologically: classical, cellular, and mixed. A frequent genetic alteration is the translocation t(12;15) resulting in a fusion of the ETV6 gene on 12p13 and the NTRK3 gene on 15p15 that occurs almost exclusively in cellular MN. The aim of this study was to determine translocation status of a large cohort of MN with respect to tumor subtype and outcome. PROCEDURE: In total, clinical data from 111 patients were available. Sixty-seven tumors were classical MN (51%), 29 cellular MN (31%), and 15 were mixed MN (18%). From these 111 cases, 79 were analyzed by FISH and RT-PCR. RESULTS: All classical and mixed MN were translocation negative. Seventeen out of 29 (58%) cellular MN harbored the ETV6-NTRK3 translocation. Five-year relapse-free survival (RFS) and overall survival (OS) were 93.2% and 96.8% for the complete cohort. All seven relapses occurred in translocation negative tumors. Five-year RFS was significantly inferior for cellular and mixed MN compared to classic MN (89%, 80%, and 98%), whereas 5-year OS was similar (93%, 96%, and 98%). Within the group of cellular MN, patients having translocation-positive tumors had a significantly superior RFS (5-year RFS: 100% vs. 73%). CONCLUSION: The majority of cellular MNs harbor the ETV6-NTKR3 gene fusion, whereas all classic- and mixed-type MNs were translocation negative. Within the cellular subgroup, patients having translocation-positive tumors had a significantly superior RFS.


Subject(s)
Chromosomes, Human, Pair 12 , Chromosomes, Human, Pair 15 , Nephroma, Mesoblastic , Oncogene Proteins, Fusion , Translocation, Genetic , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 12/metabolism , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , Disease-Free Survival , Female , Humans , In Situ Hybridization, Fluorescence , Male , Nephroma, Mesoblastic/genetics , Nephroma, Mesoblastic/metabolism , Nephroma, Mesoblastic/mortality , Nephroma, Mesoblastic/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
14.
Hum Mol Genet ; 26(20): 3995-4010, 2017 10 15.
Article in English | MEDLINE | ID: mdl-29016856

ABSTRACT

Maternally derived copy number gains of human chromosome 15q11.2-q13.3 (Dup15q syndrome or Dup15q) cause intellectual disability, epilepsy, developmental delay, hypotonia, speech impairments, and minor dysmorphic features. Dup15q syndrome is one of the most common and penetrant chromosomal abnormalities observed in individuals with autism spectrum disorder (ASD). Although ∼40 genes are located in the 15q11.2-q13.3 region, overexpression of the ubiquitin-protein E3A ligase (UBE3A) gene is thought to be the predominant molecular cause of the phenotypes observed in Dup15q syndrome. The UBE3A gene demonstrates maternal-specific expression in neurons and loss of maternal UBE3A causes Angelman syndrome, a neurodevelopmental disorder with some overlapping neurological features to Dup15q. To directly test the hypothesis that overexpression of UBE3A is an important underlying molecular cause of neurodevelopmental dysfunction, we developed and characterized a mouse overexpressing Ube3a isoform 2 in excitatory neurons. Ube3a isoform 2 is conserved between mouse and human and known to play key roles in neuronal function. Transgenic mice overexpressing Ube3a isoform 2 in excitatory forebrain neurons exhibited increased anxiety-like behaviors, learning impairments, and reduced seizure thresholds. However, these transgenic mice displayed normal social approach, social interactions, and repetitive motor stereotypies that are relevant to ASD. Reduced forebrain, hippocampus, striatum, amygdala, and cortical volume were also observed. Altogether, these findings show neuronal overexpression of Ube3a isoform 2 causes phenotypes translatable to neurodevelopmental disorders.


Subject(s)
Intellectual Disability/enzymology , Neurons/enzymology , Ubiquitin-Protein Ligases/biosynthesis , Animals , Antisocial Personality Disorder/genetics , Antisocial Personality Disorder/metabolism , Anxiety/genetics , Anxiety/metabolism , Chromosome Aberrations , Chromosomes, Human, Pair 15/enzymology , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Phenotype , Seizures/genetics , Seizures/metabolism , Ubiquitin-Protein Ligases/genetics
15.
Neurobiol Dis ; 108: 238-248, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28888970

ABSTRACT

Duplication 15q syndrome (Dup15q) is an autism-associated disorder co-incident with high rates of pediatric epilepsy. Additional copies of the E3 ubiquitin ligase UBE3A are thought to cause Dup15q phenotypes, yet models overexpressing UBE3A in neurons have not recapitulated the epilepsy phenotype. We show that Drosophila endogenously expresses Dube3a (fly UBE3A homolog) in glial cells and neurons, prompting an investigation into the consequences of glial Dube3a overexpression. Here we expand on previous work showing that the Na+/K+ pump ATPα is a direct ubiquitin ligase substrate of Dube3a. A robust seizure-like phenotype was observed in flies overexpressing Dube3a in glial cells, but not neurons. Glial-specific knockdown of ATPα also produced seizure-like behavior, and this phenotype was rescued by simultaneously overexpressing ATPα and Dube3a in glia. Our data provides the basis of a paradigm shift in Dup15q research given that clinical phenotypes have long been assumed to be due to neuronal UBE3A overexpression.


Subject(s)
Drosophila Proteins/metabolism , Neuroglia/metabolism , Seizures/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Synapses/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Animals, Genetically Modified , Chromosomes, Human, Pair 15/metabolism , Disease Models, Animal , Disease Progression , Down-Regulation , Drosophila , Drosophila Proteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Photoreceptor Cells, Invertebrate/metabolism , Photoreceptor Cells, Invertebrate/pathology , Potassium/metabolism , Seizures/pathology , Sodium-Potassium-Exchanging ATPase/genetics , Synapses/pathology , Trisomy/pathology , Ubiquitin-Protein Ligases/genetics
16.
Methods ; 112: 39-45, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27394668

ABSTRACT

Automated imaging flow cytometry integrates flow cytometry with digital microscopy to produce high-resolution digital imaging with quantitative analysis. This enables cell identification based on morphology (cell size, shape), antigen expression, quantification of fluorescence signal intensity and localisation of detected signals (i.e. surface, cytoplasm, nuclear). We describe applications of imaging flow cytometry for the diagnostic assessment of acute leukaemia. These bone marrow malignancies are traditionally diagnosed and classified by cell morphology, phenotype and cytogenetic abnormalities. Traditionally morphology is assessed by light microscopy, phenotyping by conventional flow cytometry and genetics by karyotype and fluorescence in situ hybridisation (FISH) on interphase nuclei/metaphase spreads of cells on slides. Imaging flow cytometry adds a new dimension to the diagnostic assessment of these neoplasms. We describe three specific applications: From this we conclude that imaging flow cytometry offers benefits over conventional diagnostic methods. Specifically the ability to visualise the cells of interest, the pattern and localisation of expressed antigens and assess cytogenetic abnormalities in one integrated automated high-throughput test. Imaging flow cytometry presents a new paradigm for the diagnostic assessment of leukaemia.


Subject(s)
Chromosomes, Human, Pair 15/ultrastructure , Chromosomes, Human, Pair 17/ultrastructure , Flow Cytometry/methods , Image Cytometry/methods , Leukemia, Promyelocytic, Acute/diagnostic imaging , Translocation, Genetic , Aneuploidy , Automation, Laboratory , Chromosomes, Human, Pair 15/metabolism , Chromosomes, Human, Pair 17/metabolism , Flow Cytometry/instrumentation , Gene Expression , Humans , Image Cytometry/instrumentation , In Situ Hybridization, Fluorescence/methods , Interphase , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phenotype
18.
Eur Neuropsychopharmacol ; 26(7): 1150-60, 2016 07.
Article in English | MEDLINE | ID: mdl-27061851

ABSTRACT

The 15q13.3 microdeletion copy number variation is strongly associated with schizophrenia and epilepsy. The CHRNA7 gene, encoding nicotinic acetylcholine alpha 7 receptors (nAChA7Rs), is hypothesized to be one of the main genes in this deletion causing the neuropsychiatric phenotype. Here we used a recently developed 15q13.3 microdeletion mouse model to explore whether an established schizophrenia-associated connectivity phenotype is replicated in a murine model, and whether positive modulation of nAChA7 receptor might pharmacologically normalize the connectivity patterns. Resting-state fMRI data were acquired from male mice carrying a hemizygous 15q13.3 microdeletion (N=9) and from wild-type mice (N=9). To study the connectivity profile of 15q13.3 mice and test the effect of nAChA7 positive allosteric modulation, the 15q13.3 mice underwent two imaging sessions, one week apart, receiving a single intraperitoneal injection of either 15mg/kg Lu AF58801 or saline. The control group comprised wild-type mice treated with saline. We performed seed-based functional connectivity analysis to delineate aberrant connectivity patterns associated with the deletion (15q13.3 mice (saline treatment) versus wild-type mice (saline treatment)) and their modulation by Lu AF58801 (15q13.3 mice (Lu AF58801 treatment) versus 15q13.3 mice (saline treatment)). Compared to wild-type mice, 15q13.3 mice evidenced a predominant hyperconnectivity pattern. The main effect of Lu AF58801 was a normalization of elevated functional connectivity between prefrontal and frontal, hippocampal, striatal, thalamic and auditory regions. The strongest effects were observed in brain regions expressing nAChA7Rs, namely hippocampus, cerebral cortex and thalamus. These effects may underlie the antiepileptic, pro-cognitive and auditory gating deficit-reversal effects of nAChA7R stimulation.


Subject(s)
Brain/drug effects , Cholinergic Agents/pharmacology , Chromosome Disorders/drug therapy , Intellectual Disability/drug therapy , Schizophrenia/drug therapy , Seizures/drug therapy , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Brain/diagnostic imaging , Brain/metabolism , Brain Mapping , Chromosome Deletion , Chromosome Disorders/genetics , Chromosome Disorders/metabolism , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , Cyclopropanes/pharmacology , Disease Models, Animal , Endophenotypes , Intellectual Disability/genetics , Intellectual Disability/metabolism , Magnetic Resonance Imaging , Male , Mice, Transgenic , Neural Pathways/diagnostic imaging , Neural Pathways/drug effects , Neural Pathways/metabolism , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Rest , Schizophrenia/metabolism , Seizures/genetics , Seizures/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics
19.
Gene ; 575(2 Pt 1): 353-8, 2016 Jan 10.
Article in English | MEDLINE | ID: mdl-26335514

ABSTRACT

Prader-Willi and Angelman syndromes are two distinct neurogenetic disorders caused by chromosomal deletions, uniparental disomy or loss of the imprinted gene expression in the 15q11-q13 region. PWS results from the lack of the paternally expressed gene contribution in the region. The aim of our study was to compare a new molecular approach based on the quantification of the expression of non-imprinted bi-allelic gene (NIPA1 and OCA2) with in house MS-PCR and the MS-MLPA test. Blood samples were collected from 12 patients, clinical criteria positives for Prader-Willi syndrome. DNA and RNA samples were isolated from white blood cells. Epigenetic changes at SNRPN gene locus were evaluated by MS-PCR technique. The expression levels of two non-imprinted genes (NIPA1 and OCA2) were evaluated in qReal-Time PCR, in order to identify type 1 and type 2 deletions. SALSA MS-MLPA kit ME028 was used to detect copy number changes and to analyze CpG islands methylation of the 15q11 region. MS-MLPA test confirmed that 8/12 patients presented different types of deletion at the SNRPN gene level (promoter, introns, and exons) and 4/8 displayed type 1 or type 2 deletion. In children with 15q11-13 deletions, the decreased level of NIPA1and OCA2 gene expression is related to chromosomal abnormality in the investigated area. The deletions were confirmed by MS-MLPA analysis, thus recommending NIPA1 and OCA2 gene expression as an alternate method to investigate deletions.


Subject(s)
Chromosomes, Human, Pair 15 , Gene Expression Regulation , Membrane Proteins , Membrane Transport Proteins , Prader-Willi Syndrome , Sequence Deletion , Child , Child, Preschool , Chromosome Aberrations , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , CpG Islands , DNA Methylation , Female , Genetic Loci , Humans , Infant , Male , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Transport Proteins/biosynthesis , Membrane Transport Proteins/genetics , Prader-Willi Syndrome/diagnosis , Prader-Willi Syndrome/genetics , Prader-Willi Syndrome/metabolism , Real-Time Polymerase Chain Reaction
20.
Epigenomics ; 7(7): 1213-28, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26585570

ABSTRACT

The E3 ubiquitin ligase UBE3A, also known as E6-AP, has a multitude of ascribed functions and targets relevant to human health and disease. Epigenetic regulation of the UBE3A gene by parentally imprinted noncoding transcription within human chromosome 15q11.2-q13.3 is responsible for the maternal-specific effects of 15q11.2-q13.3 deletion or duplication disorders. Here, we review the evidence for diverse and emerging roles for UBE3A in the proteasome, synapse and nucleus in regulating protein stability and transcription as well as the current mechanistic understanding of UBE3A imprinting in neurons. Angelman and Dup15q syndromes as well as experimental models of these neurodevelopmental disorders are highlighted as improving understanding of UBE3A and its complex regulation for improving therapeutic strategies.


Subject(s)
Angelman Syndrome/genetics , Epigenesis, Genetic , Genomic Imprinting , Trisomy/genetics , Ubiquitin-Protein Ligases/genetics , Angelman Syndrome/metabolism , Angelman Syndrome/pathology , Cell Nucleus/metabolism , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 15/metabolism , Female , Humans , Male , Neurons/metabolism , Neurons/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis , Synapses/metabolism , Synapses/pathology , Transcription, Genetic , Trisomy/pathology , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...