Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Genome Res ; 31(11): 1983-1993, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34675070

RESUMO

Long terminal repeat (LTR) retrotransposons are widely distributed across the human genome. They have accumulated through retroviral integration into germline DNA and are latent genetic modules. Active LTR promoters are observed in germline cells; however, little is known about the mechanisms underlying their active transcription in somatic tissues. Here, by integrating our previous transcriptome data set with publicly available data sets, we show that the LTR families MLT2A1 and MLT2A2 are primarily expressed in human four-cell and eight-cell embryos and are also activated in some adult somatic tissues, particularly pineal gland. Three MLT2A elements function as the promoters and first exons of the protein-coding genes ABCE1, COL5A1, and GALNT13 specifically in the pineal gland of humans but not in that of macaques, suggesting that the exaptation of these LTRs as promoters occurred during recent primate evolution. This analysis provides insight into the possible transition from germline insertion to somatic expression of LTR retrotransposons.


Assuntos
Retroelementos , Sequências Repetidas Terminais , Animais , Primatas/genética , Regiões Promotoras Genéticas , Retroelementos/genética , Sequências Repetidas Terminais/genética
2.
Neuroendocrinology ; 111(1-2): 27-44, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-31991411

RESUMO

BACKGROUND: Male estrogen receptor beta (ERß) knockout (BERKO) mice display anxiety and aggression linked to, among others, altered serotonergic signaling in the basolateral amygdala and dorsal raphe, impaired cortical radial glia migration, and reduced GABAergic signaling. The effects on primary motor cortex (M1 cortex) and locomotor activity as a consequence of ERß loss have not been investigated. OBJECTIVE: The aim of this study was to determine whether locomotor activity is altered as a consequence of the changes in the M1 cortex. METHODS: The locomotor activity of male wild-type (WT) and BERKO mice was evaluated using the open-field and rotarod tests. Molecular changes in the M1 cortex were analyzed by RNA sequencing, electron microscopy, electrophysiology, and immunohistological techniques. In addition, we established oligodendrocyte (OL) cultures from WT and BERKO mouse embryonic stem cells to evaluate OL function. RESULTS: Locomotor profiling revealed that BERKO mice were more active than WT mice but had impaired motor coordination. Analysis of the M1 cortex pointed out differences in synapse function and myelination. There was a reduction in GABAergic signaling resulting in imbalanced excitatory and inhibitory neurotransmission as well as a defective OL differentiation accompanied by myelin defects. The effects of ERß loss on OL differentiation were confirmed in vitro. CONCLUSION: ERß is an important regulator of GABAergic interneurons and OL differentiation, which impacts on adult M1 cortex function and may be linked to increased locomotor activity and decreased motor coordination in BERKO mice.


Assuntos
Receptor beta de Estrogênio/genética , Locomoção/genética , Córtex Motor/fisiopatologia , Bainha de Mielina/fisiologia , Desempenho Psicomotor , Transmissão Sináptica , Animais , Expressão Gênica , Masculino , Camundongos , Camundongos Knockout , Córtex Motor/metabolismo , Oligodendroglia/patologia
3.
Proc Natl Acad Sci U S A ; 114(19): E3816-E3822, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28439009

RESUMO

As estrogen receptor ß-/- (ERß-/-) mice age, the ventral prostate (VP) develops increased numbers of hyperplastic, fibroplastic lesions and inflammatory cells. To identify genes involved in these changes, we used RNA sequencing and immunohistochemistry to compare gene expression profiles in the VP of young (2-mo-old) and aging (18-mo-old) ERß-/- mice and their WT littermates. We also treated young and old WT mice with an ERß-selective agonist and evaluated protein expression. The most significant findings were that ERß down-regulates androgen receptor (AR) signaling and up-regulates the tumor suppressor phosphatase and tensin homolog (PTEN). ERß agonist increased expression of the AR corepressor dachshund family (DACH1/2), T-cadherin, stromal caveolin-1, and nuclear PTEN and decreased expression of RAR-related orphan receptor c, Bcl2, inducible nitric oxide synthase, and IL-6. In the ERß-/- mouse VP, RNA sequencing revealed that the following genes were up-regulated more than fivefold: Bcl2, clusterin, the cytokines CXCL16 and -17, and a marker of basal/intermediate cells (prostate stem cell antigen) and cytokeratins 4, 5, and 17. The most down-regulated genes were the following: the antioxidant gene glutathione peroxidase 3; protease inhibitors WAP four-disulfide core domain 3 (WFDC3); the tumor-suppressive genes T-cadherin and caveolin-1; the regulator of transforming growth factor ß signaling SMAD7; and the PTEN ubiquitin ligase NEDD4. The role of ERß in opposing AR signaling, proliferation, and inflammation suggests that ERß-selective agonists may be used to prevent progression of prostate cancer, prevent fibrosis and development of benign prostatic hyperplasia, and treat prostatitis.


Assuntos
Envelhecimento/metabolismo , Regulação para Baixo , Receptor beta de Estrogênio/metabolismo , Próstata/metabolismo , Receptores Androgênicos/biossíntese , Transdução de Sinais , Envelhecimento/genética , Envelhecimento/patologia , Androgênios/metabolismo , Animais , Quimiocina CXCL16/biossíntese , Quimiocina CXCL16/genética , Quimiocinas CXC/biossíntese , Quimiocinas CXC/genética , Clusterina/biossíntese , Clusterina/genética , Receptor beta de Estrogênio/genética , Interleucina-6/genética , Interleucina-6/metabolismo , Queratinas/biossíntese , Queratinas/genética , Masculino , Camundongos , Camundongos Knockout , Ubiquitina-Proteína Ligases Nedd4/biossíntese , Ubiquitina-Proteína Ligases Nedd4/genética , PTEN Fosfo-Hidrolase/biossíntese , PTEN Fosfo-Hidrolase/genética , Próstata/patologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores Androgênicos/genética , Proteína Smad7/biossíntese , Proteína Smad7/genética
4.
Development ; 143(19): 3459-3469, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27578796

RESUMO

Leucine twenty homeobox (LEUTX) is a paired (PRD)-like homeobox gene that is expressed almost exclusively in human embryos during preimplantation development. We previously identified a novel transcription start site for the predicted human LEUTX gene based on the transcriptional analysis of human preimplantation embryos. The novel variant encodes a protein with a complete homeodomain. Here, we provide a detailed description of the molecular cloning of the complete homeodomain-containing LEUTX Using a human embryonic stem cell overexpression model we show that the complete homeodomain isoform is functional and sufficient to activate the transcription of a large proportion of the genes that are upregulated in human embryo genome activation (EGA), whereas the previously predicted partial homeodomain isoform is largely inactive. Another PRD-like transcription factor, DPRX, is then upregulated as a powerful repressor of transcription. We propose a two-stage model of human EGA in which LEUTX acts as a transcriptional activator at the 4-cell stage, and DPRX as a balancing repressor at the 8-cell stage. We conclude that LEUTX is a candidate regulator of human EGA.


Assuntos
Blastocisto/metabolismo , Células-Tronco Embrionárias/metabolismo , Proteínas de Homeodomínio/metabolismo , Isoformas de Proteínas/metabolismo , Animais , Linhagem Celular , Ensaio de Desvio de Mobilidade Eletroforética , Técnica Indireta de Fluorescência para Anticorpo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Reação em Cadeia da Polimerase , Isoformas de Proteínas/genética
5.
Reproduction ; 151(3): 253-9, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26700939

RESUMO

Estrogen receptor beta (ERß/ESR2) has a central role in mouse ovaries, as ERß knockout (BERKO) mice are subfertile due to an increase in fibrosis around the maturing follicle and a decrease in blood supply. This has a consequence that these follicles rarely rupture to release the mature oocyte. Matrix metalloproteinases (MMPs) are modulators of the extracellular matrix, and the expression of one specific MMP, MMP-19, is normally increased in granulosa cells during their maturation until ovulation. In this study, we demonstrate that MMP-19 levels are downregulated in BERKO mouse ovaries. Using human MCF-7 cells that overexpress ERß, we could identify MMP-19 to be a transcriptional target of ligand-bound activated ERß acting on a specificity protein-1 binding site. These data provide a molecular explanation for the observed follicle rupture defect that contributes to the subfertility of female BERKO mice.


Assuntos
Receptor beta de Estrogênio/metabolismo , Metaloproteinases da Matriz Secretadas/metabolismo , Ovário/metabolismo , Animais , Gonadotropina Coriônica , Regulação para Baixo , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Células MCF-7 , Metaloproteinases da Matriz Secretadas/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovulação , Fator de Transcrição Sp1/metabolismo
6.
Stem Cells ; 33(5): 1434-46, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25645121

RESUMO

Congenital heart defects (CHD) occur in approximately 50% of patients with Down syndrome (DS); the mechanisms for this occurrence however remain unknown. In order to understand how these defects evolve in early development in DS, we focused on the earliest stages of cardiogenesis to ascertain perturbations in development leading to CHD. Using a trisomy 21 (T21) sibling human embryonic stem cell (hESC) model of DS, we show that T21-hESC display many significant differences in expression of genes and cell populations associated with mesodermal, and more notably, secondary heart field (SHF) development, in particular a reduced number of ISL1(+) progenitor cells. Furthermore, we provide evidence for two candidate genes located on chromosome 21, ETS2 and ERG, whose overexpression during cardiac commitment likely account for the disruption of SHF development, as revealed by downregulation or overexpression experiments. Additionally, we uncover an abnormal electrophysiological phenotype in functional T21 cardiomyocytes, a result further supported by mRNA expression data acquired using RNA-Seq. These data, in combination, revealed a cardiomyocyte-specific phenotype in T21 cardiomyocytes, likely due to the overexpression of genes such as RYR2, NCX, and L-type Ca(2+) channel. These results contribute to the understanding of the mechanisms involved in the development of CHD. Stem Cells 2015;33:1434-1446.


Assuntos
Síndrome de Down/patologia , Síndrome de Down/fisiopatologia , Coração/embriologia , Coração/fisiopatologia , Células-Tronco Embrionárias Humanas/metabolismo , Miócitos Cardíacos/patologia , Potenciais de Ação , Diferenciação Celular , Linhagem Celular , Cromossomos Humanos Par 21/genética , Síndrome de Down/genética , Regulação da Expressão Gênica no Desenvolvimento , Estudos de Associação Genética , Cardiopatias Congênitas/genética , Humanos , Modelos Biológicos , Miócitos Cardíacos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sequência de RNA , Transcriptoma/genética
7.
Bioinformatics ; 29(22): 2943-5, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23995393

RESUMO

MOTIVATION: Recent transcriptome studies have revealed that total transcript numbers vary by cell type and condition; therefore, the statistical assumptions for single-cell transcriptome studies must be revisited. SAMstrt is an extension code for SAMseq, which is a statistical method for differential expression, to enable spike-in normalization and statistical testing based on the estimated absolute number of transcripts per cell for single-cell RNA-seq methods. AVAILABILITY AND IMPLEMENTATION: SAMstrt is implemented on R and available in github (https://github.com/shka/R-SAMstrt). CONTACT: shintaro.katayama@ki.se


Assuntos
Perfilação da Expressão Gênica/métodos , Análise de Sequência de RNA/métodos , Interpretação Estatística de Dados , Humanos , Análise de Célula Única
8.
Reproduction ; 147(6): 875-83, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24625359

RESUMO

The coxsackievirus and adenovirus receptor (CXADR (CAR)) is a cell adhesion molecule expressed mainly in epithelial cells. Numerous evidence indicate that CXADR has an important role in testis development and function of the blood-testis barrier (BTB) in vitro. The role of CXADR in testis physiology in vivo has, however, not been addressed. We therefore constructed a conditional CXADR knockout (cKO) mouse model in which CXADR can be depleted at any chosen timepoint by the administration of tamoxifen. We report for the first time that testicular depletion of CXADR in adult and pubertal mice does not alter BTB permeability or germ cell migration across the BTB during spermatogenesis. Adult cKO mice display normal junctional ultra-structure and localization of the junctional proteins claudin-3, occludin, junction-associated molecule-A (JAM-A), and ZO1. The BTB was intact with no leakage of biotin and lanthanum tracers into the tubular lumen. Adult CXADR cKO mice were fertile with normal sperm parameters and litter size. Breeding experiments and genotyping of the pups demonstrated that CXADR-negative sperm could fertilize WT eggs. In addition, knocking down CXADR from postnatal day 9 (P9) does not affect testicular development and BTB formation. These cKO mice were analyzed at P49 and P90 and display an intact barrier and uncompromised fertility. We conclude that CXADR possesses no direct role in testicular physiology in vivo.


Assuntos
Barreira Hematotesticular/metabolismo , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/deficiência , Espermatogênese , Espermatozoides/metabolismo , Fatores Etários , Animais , Barreira Hematotesticular/ultraestrutura , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/genética , Feminino , Fertilidade , Junções Intercelulares/metabolismo , Tamanho da Ninhada de Vivíparos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Permeabilidade , Gravidez , Maturidade Sexual , Proteínas de Junções Íntimas/metabolismo
9.
N Engl J Med ; 361(5): 489-95, 2009 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-19641205

RESUMO

The mitochondrial aspartate-glutamate carrier isoform 1 (AGC1), specific to neurons and muscle, supplies aspartate to the cytosol and, as a component of the malate-aspartate shuttle, enables mitochondrial oxidation of cytosolic NADH, thought to be important in providing energy for neurons in the central nervous system. We describe AGC1 deficiency, a novel syndrome characterized by arrested psychomotor development, hypotonia, and seizures in a child with a homozygous missense mutation in the solute carrier family 25, member 12, gene SLC25A12, which encodes the AGC1 protein. Functional analysis of the mutant AGC1 protein showed abolished activity. The child had global hypomyelination in the cerebral hemispheres, suggesting that impaired efflux of aspartate from neuronal mitochondria prevents normal myelin formation.


Assuntos
Sistemas de Transporte de Aminoácidos Acídicos/deficiência , Antiporters/deficiência , Cérebro/patologia , Epilepsia/genética , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/genética , Proteínas de Transporte da Membrana Mitocondrial/genética , Mutação de Sentido Incorreto , Transtornos Psicomotores/genética , Ácido Aspártico/metabolismo , Pré-Escolar , Feminino , Homozigoto , Humanos , Imageamento por Ressonância Magnética , Mitocôndrias/metabolismo , Hipotonia Muscular/genética , Isoformas de Proteínas , Análise de Sequência de DNA , Síndrome
10.
Hum Mol Genet ; 18(15): 2813-24, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19423553

RESUMO

The molecular mechanisms that control reproductive aging and menopausal age in females are poorly understood. Here, we provide genetic evidence that 3-phosphoinositide-dependent protein kinase-1 (PDK1) signaling in oocytes preserves reproductive lifespan by maintaining the survival of ovarian primordial follicles. In mice lacking the PDK1-encoding gene Pdk1 in oocytes, the majority of primordial follicles are depleted around the onset of sexual maturity, causing premature ovarian failure (POF) during early adulthood. We further showed that suppressed PDK1-Akt-p70 S6 kinase 1 (S6K1)-ribosomal protein S6 (rpS6) signaling in oocytes appears to be responsible for the loss of primordial follicles, and mice lacking the Rps6 gene in oocytes show POF similar to that in Pdk1-deficient mice. In combination with our earlier finding that phosphatase and tensin homolog deleted on chromosome 10 (PTEN) in oocytes suppresses follicular activation, we have now pinpointed the molecular network involving phosphatidylinositol 3 kinase (PI3K)/PTEN-PDK1 signaling in oocytes that controls the survival, loss and activation of primordial follicles, which together determine reproductive aging and the length of reproductive life in females. Underactivation or overactivation of this signaling pathway in oocytes is shown to cause pathological conditions in the ovary, including POF and infertility.


Assuntos
Envelhecimento/metabolismo , Oócitos/metabolismo , Folículo Ovariano/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Feminino , Humanos , Expectativa de Vida , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oócitos/citologia , Oócitos/crescimento & desenvolvimento , Folículo Ovariano/citologia , Folículo Ovariano/crescimento & desenvolvimento , Proteínas Serina-Treonina Quinases/genética , Proteína S6 Ribossômica/genética , Proteína S6 Ribossômica/metabolismo , Proteínas Quinases S6 Ribossômicas/genética , Proteínas Quinases S6 Ribossômicas/metabolismo
11.
Genome Med ; 13(1): 40, 2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33726816

RESUMO

BACKGROUND: We report the findings from 4437 individuals (3219 patients and 1218 relatives) who have been analyzed by whole genome sequencing (WGS) at the Genomic Medicine Center Karolinska-Rare Diseases (GMCK-RD) since mid-2015. GMCK-RD represents a long-term collaborative initiative between Karolinska University Hospital and Science for Life Laboratory to establish advanced, genomics-based diagnostics in the Stockholm healthcare setting. METHODS: Our analysis covers detection and interpretation of SNVs, INDELs, uniparental disomy, CNVs, balanced structural variants, and short tandem repeat expansions. Visualization of results for clinical interpretation is carried out in Scout-a custom-developed decision support system. Results from both singleton (84%) and trio/family (16%) analyses are reported. Variant interpretation is done by 15 expert teams at the hospital involving staff from three clinics. For patients with complex phenotypes, data is shared between the teams. RESULTS: Overall, 40% of the patients received a molecular diagnosis ranging from 19 to 54% for specific disease groups. There was heterogeneity regarding causative genes (n = 754) with some of the most common ones being COL2A1 (n = 12; skeletal dysplasia), SCN1A (n = 8; epilepsy), and TNFRSF13B (n = 4; inborn errors of immunity). Some causative variants were recurrent, including previously known founder mutations, some novel mutations, and recurrent de novo mutations. Overall, GMCK-RD has resulted in a large number of patients receiving specific molecular diagnoses. Furthermore, negative cases have been included in research studies that have resulted in the discovery of 17 published, novel disease-causing genes. To facilitate the discovery of new disease genes, GMCK-RD has joined international data sharing initiatives, including ClinVar, UDNI, Beacon, and MatchMaker Exchange. CONCLUSIONS: Clinical WGS at GMCK-RD has provided molecular diagnoses to over 1200 individuals with a broad range of rare diseases. Consolidation and spread of this clinical-academic partnership will enable large-scale national collaboration.


Assuntos
Atenção à Saúde , Doenças Raras/diagnóstico , Doenças Raras/genética , Sequenciamento Completo do Genoma , Estudos de Coortes , Variações do Número de Cópias de DNA/genética , Heterogeneidade Genética , Genômica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Disseminação de Informação , Padrões de Herança/genética , Repetições de Microssatélites/genética , Mutação/genética , Suécia , Dissomia Uniparental/genética
12.
Evol Dev ; 12(3): 329-42, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20565543

RESUMO

The cystatin family comprises a group of generally broadly expressed protease inhibitors. The Cres/Testatin subgroup (CTES) genes within the type 2 cystatins differs from the classical type 2 cystatins in having a strikingly reproductive tissue-specific expression, and putative functions in reproduction have therefore been discussed. We have performed evolutionary studies of the CTES genes based on gene searches in genomes from 11 species. Ancestors of the cystatin family can be traced back to plants. We have localized the evolutionary origin of the CTES genes to the split of marsupial and placental mammals. A model for the evolution of these genes illustrates that they constitute a dynamic group of genes, which has undergone several gene expansions and we find indications of a high degree of positive selection, in striking contrast to what is seen for the classical cystatin C. We show with phylogenetic relations that the CTES genes are clustered into three original groups, a testatin, a Cres, and a CstL1 group. We have further characterized the expression patterns of all human members of the subfamily. Of a total of nine identified human genes, four express putative functional transcripts with a predominant expression in the male reproductive system. Our results are compatible with a function of this gene family in reproduction.


Assuntos
Cistatinas/genética , Evolução Molecular , Sequência de Bases , Primers do DNA , DNA Complementar , Humanos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Mol Cell Biol ; 25(12): 4892-902, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15923608

RESUMO

The testatin gene was previously isolated in a screen focused on finding novel signaling molecules involved in sex determination and differentiation. testatin is specifically upregulated in pre-Sertoli cells in early fetal development, immediately after the onset of Sry expression, and was therefore considered a strong candidate for involvement in early testis development. testatin expression is maintained in the adult Sertoli cell, and it can also be found in a small population of germ cells. Testatin shows homology to family 2 cystatins, a group of broadly expressed small secretory proteins that are inhibitors of cysteine proteases in vitro but whose in vivo functions are unclear. testatin belongs to a novel subfamily among the cystatins, comprising genes that all show expression patterns that are strikingly restricted to reproductive tissue. To investigate a possible role of testatin in testis development and male reproduction, we have generated a mouse with targeted disruption of the testatin gene. We found no abnormalities in the testatin knockout mice with regard to fetal and adult testis morphology, cellular ultrastructure, body and testis weight, number of offspring, spermatogenesis, or hormonal parameters (testosterone, luteinizing hormone, and follicle-stimulating hormone).


Assuntos
Cistatinas/genética , Cistatinas/metabolismo , Fertilidade/fisiologia , Desenvolvimento Sexual/fisiologia , Testículo/crescimento & desenvolvimento , Sequência de Aminoácidos , Animais , Cistatinas/classificação , Feminino , Hormônio Foliculoestimulante/sangue , Marcação de Genes , Hormônio Luteinizante/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Filogenia , Gravidez , Reprodução/fisiologia , Alinhamento de Sequência , Análise para Determinação do Sexo , Espermatozoides/citologia , Espermatozoides/metabolismo , Testículo/metabolismo , Testículo/fisiologia , Testículo/ultraestrutura , Testosterona/sangue
14.
Reprod Biol Endocrinol ; 5: 8, 2007 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-17343741

RESUMO

BACKGROUND: Hypospadias is a common inborn error of the male urethral development, for which the aetiology is still elusive. Polymorphic variants in genes involved in the masculinisation of male genitalia, such as the androgen receptor, have been associated with some cases of hypospadias. Co-regulators of the androgen receptor start being acknowledged as possible candidates for hormone-resistance instances, which could account for hypospadias. One such molecule, the protein FKBP52, coded by the FKBP4 gene, has an important physiological role in up-regulating androgen receptor activity, an essential step in the development of the male external genitalia. The presence of hypospadias in mice lacking fkbp52 encouraged us to study the sequence and the expression of FKBP4 in boys with isolated hypospadias. PATIENTS AND METHODS: The expression of FKBP52 in the genital skin of boys with hypospadias and in healthy controls was tested by immunohistochemistry. Mutation screening in the FKBF4 gene was performed in ninety-one boys with non syndromic hypospadias. Additionally, two polymorphisms were typed in a larger cohort. RESULTS: Immunohistochemistry shows epithelial expression of FKBP52 in the epidermis of the penile skin. No apparent difference in the FKBP52 expression was detected in healthy controls, mild or severe hypospadias patients. No sequence variants in the FKBP4 gene have implicated in hypospadias in our study. CONCLUSION: FKBP52 is likely to play a role in growth and development of the male genitalia, since it is expressed in the genital skin of prepubertal boys; however alterations in the sequence and in the expression of the FKBP4 gene are not a common cause of non-syndromic hypospadias.


Assuntos
Hipospadia/genética , Chaperonas Moleculares/genética , Receptores Androgênicos/genética , Proteínas de Ligação a Tacrolimo/genética , Genitália Masculina/metabolismo , Genitália Masculina/patologia , Humanos , Hipospadia/metabolismo , Hipospadia/patologia , Masculino , Polimorfismo de Nucleotídeo Único/genética , Receptores Androgênicos/metabolismo , Síndrome , Proteínas de Ligação a Tacrolimo/biossíntese
15.
Sci Rep ; 6: 28995, 2016 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-27412763

RESUMO

PAIRED (PRD)-like homeobox genes belong to a class of predicted transcription factor genes. Several of these PRD-like homeobox genes have been predicted in silico from genomic sequence but until recently had no evidence of transcript expression. We found recently that nine PRD-like homeobox genes, ARGFX, CPHX1, CPHX2, DPRX, DUXA, DUXB, NOBOX, TPRX1 and TPRX2, were expressed in human preimplantation embryos. In the current study we characterized these PRD-like homeobox genes in depth and studied their functions as transcription factors. We cloned multiple transcript variants from human embryos and showed that the expression of these genes is specific to embryos and pluripotent stem cells. Overexpression of the genes in human embryonic stem cells confirmed their roles as transcription factors as either activators (CPHX1, CPHX2, ARGFX) or repressors (DPRX, DUXA, TPRX2) with distinct targets that could be explained by the amino acid sequence in homeodomain. Some PRD-like homeodomain transcription factors had high concordance of target genes and showed enrichment for both developmentally important gene sets and a 36 bp DNA recognition motif implicated in Embryo Genome Activation (EGA). Our data implicate a role for these previously uncharacterized PRD-like homeodomain proteins in the regulation of human embryo genome activation and preimplantation embryo development.


Assuntos
Blastocisto/metabolismo , Proteínas Fetais/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox , Proteínas de Homeodomínio/genética , Fatores de Transcrição/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Clonagem Molecular , Sequência Consenso , DNA Complementar/genética , Proteínas Fetais/biossíntese , Perfilação da Expressão Gênica , Biblioteca Gênica , Proteínas de Homeodomínio/biossíntese , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Família Multigênica , Especificidade de Órgãos , Células-Tronco Pluripotentes/metabolismo , Regiões Promotoras Genéticas , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/biossíntese
16.
Nat Commun ; 6: 8207, 2015 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-26360614

RESUMO

Transcriptional program that drives human preimplantation development is largely unknown. Here, by using single-cell RNA sequencing of 348 oocytes, zygotes and single blastomeres from 2- to 3-day-old embryos, we provide a detailed analysis of the human preimplantation transcriptome. By quantifying transcript far 5'-ends (TFEs), we include in our analysis transcripts that derive from alternative promoters. We show that 32 and 129 genes are transcribed during the transition from oocyte to four-cell stage and from four- to eight-cell stage, respectively. A number of identified transcripts originates from previously unannotated genes that include the PRD-like homeobox genes ARGFX, CPHX1, CPHX2, DPRX, DUXA, DUXB and LEUTX. Employing de novo promoter motif extraction on sequences surrounding TFEs, we identify significantly enriched gene regulatory motifs that often overlap with Alu elements. Our high-resolution analysis of the human transcriptome during preimplantation development may have important implications on future studies of human pluripotent stem cells and cell reprograming.


Assuntos
Blastocisto/metabolismo , Blastômeros/metabolismo , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Oócitos/metabolismo , RNA Mensageiro/metabolismo , Retroelementos/genética , Fatores de Transcrição/genética , Zigoto/metabolismo , Regiões 5' não Traduzidas , Perfilação da Expressão Gênica , Células HEK293 , Proteínas de Homeodomínio/metabolismo , Humanos , Análise de Sequência de RNA , Fatores de Transcrição/metabolismo
17.
PLoS One ; 9(8): e102949, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25089626

RESUMO

Infertility is a worldwide concern that can be treated with in vitro fertilization (IVF). Improvements in IVF and infertility treatment depend largely on better understanding of the molecular mechanisms for human preimplantation development. Several large-scale studies have been conducted to identify gene expression patterns for the first five days of human development, and many functional studies utilize mouse as a model system. We have identified genes of possible importance for this time period by analyzing human microarray data and available data from online databases. We selected 70 candidate genes for human preimplantation development and investigated their expression in the early mouse development from oocyte to the 8-cell stage. Maternally loaded genes expectedly decreased in expression during development both in human and mouse. We discovered that 25 significantly upregulated genes after fertilization in human included 13 genes whose orthologs in mouse behaved differently and mimicked the expression profile of maternally expressed genes. Our findings highlight many significant differences in gene expression patterns during mouse and human preimplantation development. We also describe four cancer-testis antigen families that are also highly expressed in human embryos: PRAME, SSX, GAGE and MAGEA.


Assuntos
Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Blastocisto/metabolismo , Análise por Conglomerados , Feminino , Humanos , Masculino , Camundongos , Família Multigênica
18.
Biomed Res Int ; 2013: 489032, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23956986

RESUMO

The aim of this study was to evaluate mitochondrial alteration and ATP content of germinal vesicle (GV) oocytes isolated from fresh and vitrified ovaries. After superovulation, the ovaries from adult mice were collected and divided into control and vitrified groups. GV oocytes were isolated mechanically from each group. Half were cultured for 24 hours and their maturation was assessed. Metaphase II oocytes were collected and submitted to in vitro fertilization and their fertilization rates and development to the blastocyst stage were evaluated. In the remaining GV oocytes, ATP levels were quantified, and mitochondrial distribution, mitochondrial membrane potential, and intracellular free calcium were detected with rhodamine 123, JC-1 and Flou-4 AM staining, using laser-scanning confocal microscopy. Maturation and fertilization rates of GV oocytes and the developmental rates of subsequent embryos were significantly lower in vitrified samples (P < 0.05). The ATP content and Ca(2+) levels differed significantly in fresh and vitrified GV oocytes (P < 0.05). Most mitochondria were seen as large and homogenous aggregates (66.6%) in fresh GV oocytes compared to vitrified oocytes (50%). No significant differences in mitochondrial membrane potential were found between the groups. The lower maturation and fertilization rates of GV oocytes from vitrified ovaries may be due to changes in their mitochondrial function and distribution.


Assuntos
Trifosfato de Adenosina/metabolismo , Criopreservação , Oócitos/fisiologia , Ovário/fisiologia , Animais , Blastocisto/citologia , Sobrevivência Celular/fisiologia , Feminino , Fertilização/fisiologia , Fertilização in vitro , Potencial da Membrana Mitocondrial/fisiologia , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/fisiologia , Oócitos/metabolismo , Ovário/citologia , Vitrificação
19.
Int J Dev Biol ; 55(3): 313-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21710437

RESUMO

During development there is a multitude of signaling events governing the assembly of the developing organism. Receptors for signaling molecules such as fibroblast growth factor receptor 2 (FGFR2) enable the embryo to communicate with the surrounding environment and activate downstream pathways. The neural cell adhesion molecule (NCAM) was first characterized as a cell adhesion molecule highly expressed in the nervous system, but recent studies have shown that it is also a signaling receptor. Using a novel single oocyte adaptation of the proximity ligation assay, we here show a close association between NCAM and FGFR2 in mouse oocytes and 2-cell embryos. Real-time PCR analyses revealed the presence of messenger RNA encoding key proteins in downstream signaling pathways in oocytes and early mouse embryos. In summary these findings show a co-localization of NCAM and FGFR2 in early vertebrate development with intracellular signaling pathways present to enable a cellular response.


Assuntos
Antígeno CD56/metabolismo , Embrião de Mamíferos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Antígeno CD56/genética , Diferenciação Celular , Membrana Celular/metabolismo , Embrião de Mamíferos/citologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oócitos , Reação em Cadeia da Polimerase , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA