Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Biol Reprod ; 82(4): 751-8, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20032283

RESUMO

Maternal hyperglycemia is believed to be the metabolic derangement associated with both early pregnancy loss and congenital malformations in a diabetic pregnancy. Using an in vitro model of embryo exposure to hyperglycemia, this study questioned if increased flux through the hexosamine signaling pathway (HSP), which results in increased embryonic O-linked glycosylation (O-GlcNAcylation), underlies the glucotoxic effects of hyperglycemia during early embryogenesis. Mouse zygotes were randomly allocated to culture treatment groups that included no glucose (no flux through HSP), hyperglycemia (27 mM glucose, excess flux), 0.2 mM glucosamine (GlcN) in the absence of glucose (HSP flux alone), and O-GlcNAcylation levels monitored immunohistochemically. The impact of HSP manipulation on the first differentiation in development, blastocyst formation, was assessed, as were apoptosis and cell number in individual embryos. The enzymes regulating O-GlcNAcylation, and therefore hexosamine signaling, are the beta-linked-O-GlcNAc transferase (OGT) and an O-GlcNAc-selective beta-N-acetylglucosaminidase (O-GlcNAcase). Inhibition of these enzymes has a negative impact on blastocyst formation, demonstrating the importance of this signaling system to developmental potential. The ability of the OGT inhibitor benzyl-2-acetamido-2-deoxy-alpha-D-galactopyranoside (BADGP) to reverse the glucotoxic effects of hyperglycemia on these parameters was also sought. Excess HSP flux arising from a hyperglycemic environment or glucosamine supplementation reduced cell proliferation and blastocyst formation, confirming the criticality of this signaling pathway during early embryogenesis. Inhibition of OGT using BADGP blocked the negative impact of hyperglycemia on blastocyst formation, cell number, and apoptosis. Our results suggest that dysregulation of HSP and O-GlcNAcylation is the mechanism by which the embryotoxic effects of hyperglycemia are manifested during preimplantation development.


Assuntos
Embrião de Mamíferos/metabolismo , Hexosaminas/metabolismo , Hiperglicemia/complicações , Hiperglicemia/metabolismo , Acetilglucosaminidase/imunologia , Acetilglucosaminidase/metabolismo , Animais , Anticorpos/farmacologia , Metabolismo dos Carboidratos/efeitos dos fármacos , Metabolismo dos Carboidratos/fisiologia , Embrião de Mamíferos/efeitos dos fármacos , Desenvolvimento Embrionário/efeitos dos fármacos , Desenvolvimento Embrionário/fisiologia , Ativação Enzimática/efeitos dos fármacos , Feminino , Idade Gestacional , Glicosilação/efeitos dos fármacos , Hiperglicemia/patologia , Troca Materno-Fetal/fisiologia , Camundongos , Modelos Biológicos , N-Acetilglucosaminiltransferases/imunologia , N-Acetilglucosaminiltransferases/metabolismo , Gravidez , Complicações na Gravidez/metabolismo , Complicações na Gravidez/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
2.
Reproduction ; 138(3): 493-505, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19531609

RESUMO

Ex vivo two-cell mouse embryos deprived of glucose in vitro can develop to blastocysts by increasing their pyruvate consumption; however, zygotes when glucose-deprived cannot adapt this metabolic profile and degenerate as morulae. Prior to their death, these glucose-deprived morulae exhibit upregulation of the H+-monocarboxylate co-transporter SLC16A7 and catalase, which partly co-localize in peroxisomes. SLC16A7 has been linked to redox shuttling for peroxisomal beta-oxidation. Peroxisomal function is unclear during preimplantation development, but as a peroxisomal transporter in embryos, SLC16A7 may be involved and influenced by peroxisome proliferators such as peroxisome proliferator-activated receptor-alpha (PPARA). PCR confirmed Ppara mRNA expression in mouse embryos. Zygotes were cultured with or without glucose and with the PPARA-selective agonist WY14643 and the developing embryos assessed for expression of PPARA and phospho-PPARA in relation to the upregulation of SLC16A7 and catalase driven by glucose deprivation, indicative of peroxisomal proliferation. Reactive oxygen species (ROS) production and relationship to PPARA expression were also analysed. In glucose-deprived zygotes, ROS was elevated within 2 h, as were PPARA expression within 8 h and catalase and SLC16A7 after 12-24 h compared with glucose-supplied embryos. Inhibition of ROS production prevented this induction of PPARA and SLC16A7. Selective PPARA agonism with WY14643 also induced SLC16A7 and catalase expression in the presence of glucose. These data suggest that glucose-deprived cleavage stage embryos, although supplied with sufficient monocarboxylate-derived energy, undergo oxidative stress and exhibit elevated ROS, which in turn upregulates PPARA, catalase and SLC16A7 in a classical peroxisomal proliferation response.


Assuntos
Blastocisto/metabolismo , Glucose/deficiência , Estresse Oxidativo/fisiologia , PPAR alfa/fisiologia , Peroxissomos/metabolismo , Animais , Blastocisto/efeitos dos fármacos , Catalase/genética , Catalase/metabolismo , Células Cultivadas , Embrião de Mamíferos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glucose/metabolismo , Camundongos , Camundongos Endogâmicos CBA , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , PPAR alfa/genética , PPAR alfa/metabolismo , Proliferadores de Peroxissomos/farmacologia , Peroxissomos/efeitos dos fármacos , Pirimidinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo
3.
Gene Expr Patterns ; 4(6): 621-31, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15465484

RESUMO

In this study we report the cloning and characterisation of the mouse Glut12 gene and examine for the first time its expression pattern in the earliest stages of development. Mouse Glut12 (mGlut12) was cloned from preimplantation embryos by 5'RACE RT-PCR using primers designed from an EST clone corresponding to a human GLUT12 antigenic sequence after positive immunoreactivity was observed in mouse two-cell embryos by western immunoblotting. The mGlut12 gene contains an open reading frame of 1869 base pairs, potentially encoding a polypeptide of 622 amino acids. The predicted mGLUT12 protein bears all the hallmarks of the SLC2A family of hexose transporters and shares an 83% sequence homology to human GLUT12. Consistent with its human homolog mGlut12 mRNA is found highly expressed in skeletal and cardiac muscle and fat. Additionally, it was also found in the uterus and during early embryogenesis. During early development in the mouse, Glut12 expression is clearly apparent in ovulated oocytes and two-cell embryos but declines in day 3 morulae. With the exception of some Glut12 expression apparent in blastocysts, Glut12 mRNA remains at low to undetectable levels until E11.


Assuntos
Blastocisto , Proteínas de Transporte de Monossacarídeos/biossíntese , Proteínas de Transporte de Monossacarídeos/genética , Sequência de Aminoácidos , Animais , Transporte Biológico , Blastocisto/metabolismo , Clonagem Molecular , DNA Complementar/metabolismo , Éxons , Etiquetas de Sequências Expressas , Feminino , Glucose/metabolismo , Proteínas Facilitadoras de Transporte de Glucose , Hexoses/metabolismo , Humanos , Immunoblotting , Íntrons , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Fases de Leitura Aberta , Filogenia , Estrutura Terciária de Proteína , RNA/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Distribuição Tecidual
4.
Cell Reprogram ; 13(5): 391-401, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21827322

RESUMO

Manipulation of mammalian embryos and gametes in vitro reduces viability. Specific causes for these reductions are still largely undetermined. Accumulating evidence suggests that survival rates and developmental competency may be reduced following disruptions in the epigenetic regulation of gene expression. Chromatin-based epigenetics can regulate the transcriptome through the establishment of different transcriptionally permissive and repressive chromatin environments. Recently, support has been gathering for the hypothesis that the in vitro embryo displays reduced viability due to abnormal remodelling of the paternal chromatin, which is hypothesized to result in global transcriptional repression. In this study, we have used quantitative real-time PCR to document the effect of in vitro culture on the transcription of genes that code for proteins that are directly involved in the establishment of chromatin environments. We compare in vitro embryos to embryos generated through parthenogenetic activation to determine how the absence of paternal chromatin remodeling affects transcriptional activity. Through these studies, we show that the expression of many genes encoding for histone proteins and other modifiers involved in chromatin-based epigenetic regulation are perturbed by in vitro culture. In addition, we show that the expression of many candidate genes was reduced in in vitro embryos but not in parthenogenetic embryos. These results support the hypothesis that events linked to remodeling of paternal chromatin may influence transcriptional activity in the in vitro embryo and that chromatin-based reprogramming events in developing embryos are dynamically responsive to prevailing conditions.


Assuntos
Blastocisto/metabolismo , Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Histonas/biossíntese , Transcrição Gênica , Animais , Epigênese Genética , Feminino , Masculino , Camundongos
5.
Gene Expr Patterns ; 10(6): 299-305, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20601166

RESUMO

The histone code is an epigenetic regulatory system thought to play a crucial role in cellular events such as development, differentiation and in the maintenance of pluripotency. In order to gain an insight into the role variant histones may play during mammalian development; we studied gene expression of histone variants and remodelling enzymes in mouse embryonic stem (ES) cells and during mouse preimplantation development. Using quantitative reverse-transcription PCR (qRT-PCR) we document the gene expression pattern of 12 histone variants and 2 of their associated remodelling enzymes in undifferentiated ES cells and during preimplantation embryo development. All histone variants were detected in undifferentiated ES cells, with H2AZ showing the highest expression levels of all the histone variants tested. The results also show that H2A variant levels tend to increase later in embryo development whilst H3 variant levels are elevated in early preimplantation stages. In addition, the expression of SWI/SNF, a remodeler protein involved in specifically remodelling H2A-H2B dimers, mirrors the expression of H2B and H2A variants, and the H3-H4 specific chaperone CAF-1 expression mirrors H3 variant expression. These results provide a foundation for further studies on the functions of histone variants during development, differentiation and in pluripotency.


Assuntos
Blastocisto/metabolismo , Histonas/genética , Histonas/metabolismo , Proteínas Nucleares/genética , Células-Tronco Pluripotentes/metabolismo , Animais , Diferenciação Celular/genética , Células Cultivadas , Desenvolvimento Embrionário/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Variação Genética/fisiologia , Masculino , Camundongos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Nucleares/metabolismo , Células-Tronco Pluripotentes/fisiologia , Gravidez , Ligação Proteica
6.
Biol Reprod ; 79(1): 84-92, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18385447

RESUMO

Concurrent with compaction, preimplantation mouse embryos switch from the high pyruvate consumption that prevailed during cleavage stages to glucose consumption against a constant background of pyruvate uptake. However, zygotes exposed to and subsequently deprived of glucose can form blastocysts by increasing pyruvate uptake. This metabolic switch requires cleavage-stage exposure to glucose and is one aspect of metabolic differentiation that normally occurs in vivo. Monocarboxylates, such as pyruvate and lactate, are transported across membranes via the SLC16 family of H(+)-monocarboxylate cotransporter (MCT) proteins. Thus, the increase in pyruvate uptake in embryos developing without glucose must involve changes in activity and localization of MCT. In mouse embryos, continued expression of Slc16a1 (MCT1) requires glucose supply. Messenger RNA for Slc17a7 (MCT2) and Slc16a3 (MCT4) has been detected in mouse preimplantation embryos; however, protein function, localization, and regulation of expression at the basis of these net pyruvate uptake changes remain unclear. The expression and localization of SLC16A7 and SLC16A3 have therefore been examined to clarify their respective roles in embryos derived from the reproductive tract and cultured under varied conditions. SLC16A3 appears localized to the plasma membrane until the morula stage and also maintains a nuclear distribution throughout preimplantation development. However, continued Slc16a3 mRNA expression is dependent on prior exposure to glucose. SLC16A7 localizes to apical cortical regions with punctate, vesicular expression throughout blastomeres, partially colocalizing in peroxisomes with peroxisomal catalase (CAT). In contrast to SLC16A3 and SLC16A1, SLC16A7 and CAT demonstrate upregulation in the absence of glucose. These striking differences between the two isoforms in expression localization and regulation suggest unique roles for each in monocarboxylate transport and pH regulation during preimplantation development, and implicate peroxisomal SLC16A7 as an important redox regulator in the absence of glucose.


Assuntos
Proteínas de Transporte/genética , Desenvolvimento Embrionário/genética , Transportadores de Ácidos Monocarboxílicos/genética , Animais , Blastocisto/metabolismo , Proteínas de Transporte/metabolismo , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Glucose/metabolismo , Concentração de Íons de Hidrogênio , Camundongos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Oxirredução , RNA Mensageiro/metabolismo , Simportadores , Distribuição Tecidual
7.
Biol Reprod ; 78(4): 595-600, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18046015

RESUMO

Although mouse oocytes and cleavage-stage embryos are unable to utilize glucose as a metabolic fuel, they have a specific requirement for a short exposure to glucose prior to compaction. The reason for this requirement has been unclear. In this study we confirm that cleavage-stage exposure to glucose is required for blastocyst formation and show that the absence of glucose between 18-64 h after hCG causes an irreversible decrease in cellular proliferation and an increase in apoptosis. More importantly, this glucose signals to activate expression of Slc2a3 transcript and SLC2A3 protein, a facilitative glucose transporter (previously known as GLUT3) associated with developmental competence and increased glucose uptake used to fuel blastocyst formation. Glucosamine could substitute for glucose in these roles, suggesting that hexosamine biosynthesis may be a nutrient-sensing mechanism involved in metabolic differentiation. Inhibition of the rate-limiting enzyme in this pathway, glutamine-fructose-6-phosphate amidotransferase (GFPT), inhibited expression of the SLC2A3 transporter protein and blastocyst formation. Glucosamine, a substrate that enters this pathway downstream of GFPT, was able to overcome this inhibition and support SLC2A3 expression. These data suggest that early embryos rely on hexosamine biosynthesis as a glucose-sensing pathway to initiate metabolic differentiation.


Assuntos
Desenvolvimento Embrionário/fisiologia , Glucose/metabolismo , Hexosaminas/biossíntese , Transdução de Sinais , Animais , Apoptose , Azasserina/farmacologia , Blastocisto/fisiologia , Inibidores Enzimáticos/farmacologia , Expressão Gênica/efeitos dos fármacos , Glucosamina/administração & dosagem , Glucose/administração & dosagem , Transportador de Glucose Tipo 3/genética , Glutamina-Frutose-6-Fosfato Transaminase (Isomerizante)/antagonistas & inibidores , Camundongos
8.
Reproduction ; 134(1): 41-9, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17641087

RESUMO

Although IGF-I and insulin are important modulators of preimplantation embryonic physiology, the signalling pathways activated during development remain to be elucidated. As a model of preimplantation embryos, pluripotent mouse embryonic stem cells were used to investigate which receptor mediated actions of physiological concentrations of IGF-I and insulin on growth measured by protein synthesis. Exposure of mouse embryonic stem (ES) cells to 1.7 pM IGF-I or 1.7 nM insulin for 4 h caused approximately 25% increase in protein synthesis when compared with cells cultured in basal medium containing BSA. Dose-response studies showed 100-fold higher potency of IGF-I that pointed to the type 1 IGF receptor as the mediating receptor for both ligands. This was confirmed using an anti-type 1 IGF receptor-blocking antibody (alphaIR3). Both 1.7 pM IGF-I and 1.7 nM insulin increased phosphorylation of the type 1 IGF receptor and this increase was blocked by alphaIR3, but the insulin receptor was not phosphorylated. Finally, binding of either agonist led to downstream phosphorylation of ERK1/2 mitogen-activated protein kinase (MAPK) also via IGF-1R as this was blocked by alphaIR3. Together, these results suggest that IGF-I and insulin modulate ES cell physiology through binding to the type 1 IGF receptor and subsequent activation of MAPK pathway.


Assuntos
Fator de Crescimento Insulin-Like I/farmacologia , Insulina/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células-Tronco Pluripotentes/metabolismo , Receptor IGF Tipo 1/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Bioensaio , Linhagem Celular , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosforilação , Células-Tronco Pluripotentes/efeitos dos fármacos , Biossíntese de Proteínas , Receptor IGF Tipo 1/imunologia , Estimulação Química
9.
Reproduction ; 131(3): 469-79, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16514190

RESUMO

Cleavage-stage embryos have an absolute requirement for pyruvate and lactate, but as the morula compacts, it switches to glucose as the preferred energy source to fuel glycolysis. Substrates such as glucose, amino acids, and lactate are moved into and out of cells by facilitated diffusion. In the case of lactate and pyruvate, this occurs via H+-monocarboxylate cotransporter (MCT) proteins. To clarify the role of MCT in development, transport characteristics for DL-lactate were examined, as were mRNA expression and protein localisation for MCT1 and MCT3, using confocal laser scanning immunofluorescence in freshly collected and cultured embryos. Blastocysts demonstrated significantly higher affinity for DL-lactate than zygotes (Km 20 +/- 10 vs 87 +/- 35 mmol lactate/l; P = 0.03 by linear regression) but was similar for all stages. For embryos derived in vivo and those cultured with glucose, MCT1 mRNA was present throughout preimplantation development, protein immunoreactivity appearing diffuse throughout the cytoplasm with brightest intensity in the outer cortical region of blastomeres. In expanding blastocysts, MCT1 became more prominent in the cytoplasmic cortex of blastomeres, with brightest intensity in the polar trophectoderm. Without glucose, MCT1 mRNA was not expressed, and immunoreactivity dramatically reduced in intensity as morulae died. MCT3 mRNA and immunoreactivity were not detected in early embryos. The differential expression of MCT1 in the presence or absence of glucose demonstrates that it is important in the critical regulation of pH and monocarboxylate transport during preimplantation development, and implies a role for glucose in the control of MCT1, but not MCT3, expression.


Assuntos
Blastocisto/metabolismo , Desenvolvimento Embrionário/efeitos dos fármacos , Glucose/farmacologia , Transportadores de Ácidos Monocarboxílicos/genética , Simportadores/genética , Animais , Transporte Biológico/efeitos dos fármacos , Blastocisto/efeitos dos fármacos , Western Blotting/métodos , Proteínas de Transporte/análise , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Ácido Láctico/metabolismo , Camundongos , Camundongos Endogâmicos , Microscopia Confocal , Transportadores de Ácidos Monocarboxílicos/análise , Transportadores de Ácidos Monocarboxílicos/metabolismo , Gravidez , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Simportadores/análise , Simportadores/metabolismo , Zigoto/efeitos dos fármacos , Zigoto/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa