Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
World J Clin Cases ; 9(2): 403-409, 2021 Jan 16.
Article in English | MEDLINE | ID: mdl-33521108

ABSTRACT

BACKGROUND: Most intrahepatic arterioportal fistulae (IAPF) are acquired. The few cases of congenital fistulae are diagnosed in infants and children. CASE SUMMARY: We report a 31-year-old female patient presenting with haematemesis and melena three weeks after delivering her second child. The patient had a 20-year history of abdominal distention and nausea. IAPF, along with splenomegaly and ascites, was found by Doppler sonography and confirmed by computed tomography angiography. The patient was treated with endovascular coil embolization, resulting in occlusion of the fistula. CONCLUSION: This was an unusual case of possible congenital IAPF that manifested during a second pregnancy and was complicated by portal hypertension.

3.
J Assist Reprod Genet ; 37(2): 471-475, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31916079

ABSTRACT

Non-obstructive azoospermia accounts for 10-15% of male infertility, resulting in 60% of all cases of azoospermia and affecting about 1% of the male population. About 30% of these cases are due to Y chromosome microdeletions, chromosome abnormalities, or hormonal disorders. Pathogenic variants in genes on the sex chromosomes have key roles in spermatogenic failure. The co-occurrence of azoospermia and congenital cataracts ranges between 1 in 165,000 and 1 in 500,000. Our 28-year-old patient with normal intelligence and abnormally shaped teeth presented with both disorders. A microarray revealed a microdeletion at Xp23.13 with a whole NHS gene deletion as well as a contiguous deletion of two other genes [SCML1 and RAI2]. This observation represents the first report of non-obstructive azoospermia with congenital cataracts and a contiguous deletion of the SCML1 gene, a transcript of which is exclusively expressed in the testis. SCML1 is the putative culprit gene, which requires functional study or animal experiments. Our analysis of 60 known spermatogenesis failure-related genes by whole-exome sequencing revealed no other candidate. The Nance-Horan syndrome due to pathogenic variants in the NHS gene at Xp23.13 including whole gene deletion does not have azoospermia as a feature. Our report adds to the completeness of genetic counseling for an individual with azoospermia and congenital cataracts.


Subject(s)
Azoospermia/genetics , Cataract/congenital , Cataract/genetics , Chromosome Aberrations , Genetic Diseases, X-Linked/genetics , Tooth Abnormalities/genetics , Adult , Azoospermia/pathology , Cataract/pathology , Chromosome Deletion , Chromosomes, Human, X/genetics , Genetic Diseases, X-Linked/pathology , Humans , Male , Oligospermia/genetics , Oligospermia/pathology , Spermatogenesis/genetics , Testis/growth & development , Testis/pathology , Tooth Abnormalities/pathology
4.
Genet Med ; 18(8): 850-4, 2016 08.
Article in English | MEDLINE | ID: mdl-26681313

ABSTRACT

PURPOSE: We evaluated the Exome Aggregation Consortium (ExAC) database as a control cohort to classify variants across a diverse set of genes spanning dominant and recessively inherited disorders. METHODS: The frequency of pathogenic variants in ExAC was compared with the estimated maximal pathogenic allele frequency (MPAF), based on the disease prevalence, penetrance, inheritance, allelic and locus heterogeneity of each gene. Additionally, the observed carrier frequency and the ethnicity-specific variant distribution were compared between ExAC and the published literature. RESULTS: The carrier frequency and ethnic distribution of pathogenic variants in ExAC were concordant with reported estimates. Of 871 pathogenic/likely pathogenic variants across 19 genes, only 3 exceeded the estimated MPAF. Eighty-four percent of variants with ExAC frequencies above the estimated MPAF were classified as "benign." Additionally, 20% of the cardiac and 19% of the Lynch syndrome gene variants originally classified as "VUS" occurred with ExAC frequencies above the estimated MPAF, making these suitable for reassessment. CONCLUSIONS: The ExAC database is a useful source for variant classification and is not overrepresented for pathogenic variants in the genes evaluated. However, the mutational spectrum, pseudogenes, genetic heterogeneity, and paucity of literature should be considered in deriving meaningful classifications using ExAC.Genet Med 18 8, 850-854.


Subject(s)
Databases, Genetic , Ethnicity/genetics , Genetic Variation , Exome , Gene Frequency , Genetic Predisposition to Disease , Humans
5.
J Cell Physiol ; 228(2): 313-21, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22886425

ABSTRACT

Epigenetic mechanisms mediating expression of the Runt-related transcription factor Runx2 are critical for controlling its osteogenic activity during skeletal development. Here, we characterized bona fide regulatory elements within 120 kbp of the endogenous bone-related Runx2 promoter (P1) in osteoblasts by genomic DNase I footprinting and chromatin immuno-precipitations (ChIPs). We identified a ~10 kbp genomic domain spanning the P1 promoter that interacts with acetylated histones H3 and H4 reflecting an open chromatin conformation in MC3T3 osteoblasts. This large chromatin domain contains a single major DNaseI hypersensitive (DHS) region that defines a 0.4 kbp "basal core" promoter. This region encompasses two endogenous genomic protein/DNA interaction sites (i.e., footprints at Activating Protein 1 [AP1], E-box and Runx motifs). Helix-Loop-Helix (HLH)/E-box occupancy and presence of the DHS region persists in several mesenchymal cell types, but AP1 site occupancy occurs only during S phase when Runx2 expression is minimal. Point-mutation of the HLH/E box dramatically reduces basal promoter activity. Our results indicate that the Runx2 P1 promoter utilizes two stable principal protein/DNA interaction domains associated with AP1 and HLH factors. These sites function together with dynamic and developmentally responsive sites in a major DHS region to support epigenetic control of bone-specific transcription when osteoblasts transition into a quiescent or differentiated state.


Subject(s)
Core Binding Factor Alpha 1 Subunit/genetics , Helix-Loop-Helix Motifs/genetics , Protein Interaction Domains and Motifs/genetics , Animals , Cell Line , Chromatin/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Deoxyribonuclease I/metabolism , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Helix-Loop-Helix Motifs/physiology , Histones/metabolism , Mesoderm/metabolism , Mice , Osteoblasts/metabolism , Point Mutation , Promoter Regions, Genetic , Protein Interaction Domains and Motifs/physiology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
6.
PLoS One ; 7(4): e35003, 2012.
Article in English | MEDLINE | ID: mdl-22558107

ABSTRACT

Identification of common molecular pathways affected by genetic variation in autism is important for understanding disease pathogenesis and devising effective therapies. Here, we test the hypothesis that rare genetic variation in the metabotropic glutamate-receptor (mGluR) signaling pathway contributes to autism susceptibility. Single-nucleotide variants in genes encoding components of the mGluR signaling pathway were identified by high-throughput multiplex sequencing of pooled samples from 290 non-syndromic autism cases and 300 ethnically matched controls on two independent next-generation platforms. This analysis revealed significant enrichment of rare functional variants in the mGluR pathway in autism cases. Higher burdens of rare, potentially deleterious variants were identified in autism cases for three pathway genes previously implicated in syndromic autism spectrum disorder, TSC1, TSC2, and SHANK3, suggesting that genetic variation in these genes also contributes to risk for non-syndromic autism. In addition, our analysis identified HOMER1, which encodes a postsynaptic density-localized scaffolding protein that interacts with Shank3 to regulate mGluR activity, as a novel autism-risk gene. Rare, potentially deleterious HOMER1 variants identified uniquely in the autism population affected functionally important protein regions or regulatory sequences and co-segregated closely with autism among children of affected families. We also identified rare ASD-associated coding variants predicted to have damaging effects on components of the Ras/MAPK cascade. Collectively, these findings suggest that altered signaling downstream of mGluRs contributes to the pathogenesis of non-syndromic autism.


Subject(s)
Autistic Disorder/genetics , Genetic Variation , Receptors, Metabotropic Glutamate/genetics , Signal Transduction/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Case-Control Studies , Female , High-Throughput Nucleotide Sequencing , Homer Scaffolding Proteins , Humans , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Polymorphism, Single Nucleotide/genetics , Risk Factors , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
7.
Immunol Allergy Clin North Am ; 28(2): 387-412, x, 2008 May.
Article in English | MEDLINE | ID: mdl-18424339

ABSTRACT

Gene testing in primary immune deficiencies (PIDs) once was limited to expert academic laboratories, but now is easily available to physicians with a broad range of clinical expertise. Such testing can establish or confirm a suspected diagnosis and also may predict future disease risk in advance of clinical signs and symptoms, inform reproductive decision making, and guide clinicians in selecting the most appropriate therapeutic options. This article, based on the authors' experience and a review of the published literature, discusses some of the advances and challenges currently encountered in the clinical molecular genetic diagnosis of PIDs.


Subject(s)
Genetic Testing , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Mutation , Genes , Humans , Immunologic Deficiency Syndromes/immunology , Sequence Analysis, DNA
8.
J Biol Chem ; 280(39): 33132-40, 2005 Sep 30.
Article in English | MEDLINE | ID: mdl-16043491

ABSTRACT

Both activating and null mutations of proteins required for canonical WNT signaling have revealed the importance of this pathway for normal skeletal development. However, tissue-specific transcriptional mechanisms through which WNT signaling promotes the differentiation of bone-forming cells have yet to be identified. Here, we address the hypothesis that canonical WNT signaling and the bone-related transcription factor RUNX2/CBFA1/AML3 are functionally linked components of a pathway required for the onset of osteoblast differentiation. Our findings show that, in bone of the SFRP1 (secreted frizzled-related protein-1)-null mouse, which exhibits activated WNT signaling and a high bone mass phenotype, there is a significant increase in expression of T-cell factor (TCF)-1, Runx2, and the RUNX2 target gene osteocalcin. We demonstrate by mutational analysis that a functional TCF regulatory element responsive to canonical WNT signaling resides in the promoter of the Runx2 gene (-97 to -93). By chromatin immunoprecipitation, recruitment of beta-catenin and TCF1 to the endogenous Runx2 gene is shown. Coexpression of TCF1 with canonical WNT proteins resulted in a 2-5-fold activation of Runx2 promoter activity and a 7-8-fold induction of endogenous mRNA in mouse pluripotent mesenchymal and osteoprogenitor cells. This enhancement was abrogated by SFRP1. Taken together, our results provide evidence for direct regulation of Runx2 by canonical WNT signaling and suggest that Runx2 is a target of beta-catenin/TCF1 for the stimulation of bone formation. We propose that WNT/TCF1 signaling, like bone morphogenetic protein/transforming growth factor-beta signaling, activates Runx2 gene expression in mesenchymal cells for the control of osteoblast differentiation and skeletal development.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Gene Expression Regulation, Developmental , Osteogenesis/genetics , Signal Transduction , Animals , Cell Differentiation , Cells, Cultured , Chromatin Immunoprecipitation , Fibroblasts/metabolism , Hepatocyte Nuclear Factor 1-alpha/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Models, Biological , Osteoblasts/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism
9.
J Biol Chem ; 280(21): 20274-85, 2005 May 27.
Article in English | MEDLINE | ID: mdl-15781466

ABSTRACT

The Runx2 (CBFA1/AML3/PEBP2alphaA) transcription factor promotes skeletal cell differentiation, but it also has a novel cell growth regulatory activity in osteoblasts. We addressed here whether Runx2 activity is functionally linked to cell cycle-related mechanisms that control normal osteoblast proliferation and differentiation. We found that the levels of Runx2 gene transcription, mRNA and protein, are each up-regulated with cessation of cell growth (i.e. G(0)/G(1) transition) in preconfluent MC3T3 osteoblastic cells that do not yet express mature bone phenotypic gene expression. Cell growth regulation of Runx2 is also observed in primary calvarial osteoblasts and other osteoblastic cells with relatively normal cell growth characteristics, but not in osteosarcoma cells (e.g. SAOS-2 and ROS17/2.8). Runx2 levels are cell cycle-regulated in MC3T3 cells with respect to the G(1)/S and M/G(1) transitions: oscillates from maximal expression levels during early G(1) to minimal levels during early S phase and mitosis. However, in normal or immortalized (e.g. ATDC5) chondrocytic cells, Runx2 expression is suppressed during quiescence, and Runx2 levels are not regulated during G(1) and S phase in ATDC5 cells. Antisense or small interfering RNA-mediated reduction of the low physiological levels of Runx2 in proliferating MC3T3 cells does not accelerate cell cycle progression. However, forced expression of Runx2 suppresses proliferation of MC3T3 preosteoblasts or C2C12 mesenchymal cells which have osteogenic potential. Forced elevation of Runx2 in synchronized MC3T3 cells causes a delay in G(1). We propose that Runx2 levels and function are biologically linked to a cell growth-related G(1) transition in osteoblastic cells.


Subject(s)
Cell Cycle/physiology , Cell Division/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation , Osteoblasts/cytology , Transcription Factors/genetics , Transcription Factors/physiology , Animals , Cell Line , Cell Line, Transformed , Cell Nucleus/chemistry , Core Binding Factor Alpha 1 Subunit , DNA-Binding Proteins/analysis , G1 Phase/physiology , Humans , Kinetics , Mice , Mitosis , Osteoblasts/chemistry , Osteoblasts/metabolism , Osteosarcoma/metabolism , Protein Isoforms/genetics , RNA, Messenger/analysis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor AP-2 , Transcription Factors/analysis , Transfection , Tumor Cells, Cultured
10.
Gene ; 342(1): 35-40, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15527963

ABSTRACT

Histones are the major protein component of nucleosomes, and de novo histone synthesis is essential for packaging newly replicated DNA into chromatin. As a result, histone gene expression is exquisitely and functionally coupled with DNA replication. Vastly divergent organisms such as yeast, fly and human all demonstrate the phylogenetically conserved propensity to maintain clustering of histone genes at one or more genomic loci. Although specific mechanisms are unclear, clustering is presumed to be important for common stringent transcriptional control of these genes at the G1/S phase transition. In this study, we describe a genomic duplication of the human histone gene cluster located at chromosome 1q21, which effectively doubles the previously known size and gene number of that cluster. The duplication persists in all examined tissues and cell lines, and the duplicated genes are transcriptionally active. Levels of messenger RNAs for duplicated histone H4 genes are high relative to those for non-duplicated H4 genes. Our data suggest that transcriptionally robust histone H4 genes may have been preferentially duplicated during evolution.


Subject(s)
Histones/genetics , Multigene Family/genetics , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Chromosomes, Human, Pair 1/genetics , Gene Duplication , Genes, Duplicate/genetics , Genome, Human , HL-60 Cells , HeLa Cells , Humans , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Nucleic Acid
19.
Mol Cell Biol ; 23(22): 8110-23, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14585971

ABSTRACT

At the G(1)/S phase cell cycle transition, multiple histone genes are expressed to ensure that newly synthesized DNA is immediately packaged as chromatin. Here we have purified and functionally characterized the critical transcription factor HiNF-P, which is required for E2F-independent activation of the histone H4 multigene family. Using chromatin immunoprecipitation analysis and ligation-mediated PCR-assisted genomic sequencing, we show that HiNF-P interacts with conserved H4 cell cycle regulatory sequences in vivo. Antisense inhibition of HiNF-P reduces endogenous histone H4 gene expression. Furthermore, we find that HiNF-P utilizes NPAT/p220, a substrate of the cyclin E/cyclin-dependent kinase 2 (CDK2) kinase complex, as a key coactivator to enhance histone H4 gene transcription. The biological role of HiNF-P is reflected by impeded cell cycle progression into S phase upon antisense-mediated reduction of HiNF-P levels. Our results establish that HiNF-P is the ultimate link in a linear signaling pathway that is initiated with the growth factor-dependent induction of cyclin E/CDK2 kinase activity at the restriction point and culminates in the activation of histone H4 genes through HiNF-P at the G(1)/S phase transition.


Subject(s)
Histones/genetics , S Phase/genetics , S Phase/physiology , Transcription Factors/metabolism , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Gene Expression Regulation , HeLa Cells , Humans , Molecular Sequence Data , Molecular Weight , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Repressor Proteins , Signal Transduction , Transcription Factors/genetics , Transcription Factors/isolation & purification , Zinc Fingers/genetics
20.
Mol Endocrinol ; 17(4): 743-56, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12554783

ABSTRACT

The remodeling of chromatin is required for tissue-specific gene activation to permit interactions of transcription factors and coregulators with their cognate elements. Here, we investigate the chromatin-mediated mechanisms by which the bone-specific osteocalcin (OC) gene is transcriptionally activated during cessation of cell growth in ROS 17/2.8 osteosarcoma cells and during normal osteoblast differentiation. Acetylation of histones H3 and H4 at the OC gene promoter was assayed during the proliferative and postproliferative stages of cell growth by using chromatin immunoprecipitation assays with antibodies that recognize different acetylated forms of histones H3 or H4. The results show that the promoter and coding regions of the OC gene contain very low levels of acetylated histones H3 and H4 during the proliferative period of osteoblast differentiation when the OC gene is inactive. Active expression of the OC gene in mature osteoblasts and confluent ROS 17/2.8 cells is functionally linked to preferential acetylation of histone H4 and, to a lesser extent, to acetylation of histone H3. Histone acetylation at the loci for RUNX2 (CBFA1), alkaline phosphatase, bone sialoprotein, osteopontin, and the cell growth regulator p21, which are expressed throughout osteoblast differentiation, is not altered postproliferatively. We conclude that acetylation of histones H3 and H4 is functionally coupled to the chromatin remodeling events that mediate the developmental induction of OC gene transcription in bone cells.


Subject(s)
Cell Differentiation/genetics , Histones/metabolism , Neoplasm Proteins , Osteoblasts/cytology , Osteocalcin/genetics , Transcription, Genetic , Acetylation , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Amino Acid Sequence , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Differentiation/drug effects , Cells, Cultured , Cholecalciferol/pharmacology , Chromatin/genetics , Chromatin/immunology , Chromatin/metabolism , Core Binding Factor Alpha 1 Subunit , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , Gene Expression Regulation , Integrin-Binding Sialoprotein , Molecular Sequence Data , Osteoblasts/drug effects , Osteoblasts/physiology , Osteocalcin/metabolism , Osteopontin , Osteosarcoma/genetics , Osteosarcoma/pathology , Promoter Regions, Genetic , Rats , Sialoglycoproteins/genetics , Sialoglycoproteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...