Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 19(1): e0296427, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38165864

RESUMO

Breeding for higher fertility has resulted in a higher number of low birthweight (LBW) piglets. It has been shown that LBW piglets grow slower than normal birthweight (NBW) littermates. Differences in growth performance have been associated with impaired small intestinal development. In suckling and weaning piglets, glutamine (Gln) supplementation has been associated with improved growth and intestinal development. This study was designed to examine the effects of oral Gln supplementation on growth and small intestinal parameters in LBW and NBW suckling piglets. At birth (day 0), a total of 72 LBW (1.10 ± 0.06 kg) and 72 NBW (1.51 ± 0.06) male piglets were selected. At day 1, litters were standardized to 12 piglets, and experimental piglets supplemented daily with either Gln (1 g/kg BW) or isonitrogenous amounts of Alanine (Ala) as control (1.22 g/kg BW) until day 12. Creep feed was offered from day 14 onward. Subgroups of piglets were euthanized at days 5, 12, and 26 for the analyses of jejunal morphometry, cellular proliferation, glutathione concentration and transcript abundance of tight junction proteins. From age day 11 to 21, Gln supplemented LBW (LBW-Gln) piglets were heavier than Ala supplemented LBW (LBW-Ala) littermates (P = 0.034), while NBW piglets were heavier until age day 26 compared to LBW littermates. Villus height was higher in LBW-Gln compared to LBW-Ala on age day 12 (P = 0.031). Sporadic differences among supplementation and birthweight groups were detected for jejunal cellular proliferation, cellular population and glutathione concentration, whereas age was the most dominant factor. These results show that Gln supplementation improved the growth of LBW piglets compared to LBW-Ala beyond the termination of Gln supplementation, but this was not associated with consistent effects on selected parameters of jejunal development.


Assuntos
Suplementos Nutricionais , Glutamina , Animais , Masculino , Suínos , Glutamina/farmacologia , Peso ao Nascer , Desmame , Suplementos Nutricionais/análise , Alanina , Proliferação de Células , Hiperplasia , Glutationa
2.
Genes (Basel) ; 11(7)2020 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-32668786

RESUMO

The premelanosome protein (PMEL) is important for fibril formation within melanosomes during vertebrate melanogenesis. Fibrils form a matrix for pigment deposition within pigmented tissues such as skin and hair. PMEL mutations are known to modulate eumelanic pigmentation in vertebrates. However, in bovines, PMEL mutations were also found to alter pheomelanic pigmentation resulting in coat color dilution. Furthermore, epistatic effects of a mutated PMEL allele were detected in the phenotypic expression of the bovine hair defect "rat-tail syndrome" (RTS) characterized by charcoal coat color and hair deformation. Reports about PMEL gene expression in non-pigmented tissues raised the hypothesis that there may be unknown functions of the PMEL protein beyond eumelanin deposition to PMEL fibrils. In our study, we analysed the PMEL protein expression in pigmented skin and non-pigmented bovine tissues (non-pigmented skin, thyroid gland, rumen, liver, kidney, and adrenal gland cortex). We found that a processed form of the bovine PMEL protein is expressed in pigmented as well as in non-pigmented tissues, which is in line with gene expression data from targeted RT-PCR and whole transcriptome RNAseq analysis. The PMEL protein is located in membranes and within the cytosol of epithelial cells. Based on our data from bovine tissues, we concluded that at least in cattle PMEL potentially has additional, yet unexplored functions, which might contribute to effects of PMEL mutations on pheomelanin coat color dilution and charcoal coat color in RTS animals. However, indication of PMEL protein in unpigmented cells and tissues will require further confirmation in the future, because there have been no confirmed reports before, which had detected bovine PMEL protein with specific antibodies either in pigmented or unpigmented tissue.


Assuntos
Melaninas/genética , Melanossomas/genética , Pigmentação da Pele/genética , Antígeno gp100 de Melanoma/genética , Alelos , Animais , Bovinos , Regulação da Expressão Gênica/genética , Humanos , Melaninas/biossíntese , Melanócitos/metabolismo , Mutação/genética , Fenótipo , Sequenciamento do Exoma
3.
PLoS One ; 10(4): e0124574, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25875852

RESUMO

Feed efficiency is a paramount factor for livestock economy. Previous studies had indicated a substantial heritability of several feed efficiency traits. In our study, we investigated the genetic background of residual feed intake, a commonly used parameter of feed efficiency, in a cattle resource population generated from crossing dairy and beef cattle. Starting from a whole genome association analysis, we subsequently performed combined phenotype-metabolome-genome analysis taking a systems biology approach by inferring gene networks based on partial correlation and information theory approaches. Our data about biological processes enriched with genes from the feed efficiency network suggest that genetic variation in feed efficiency is driven by genetic modulation of basic processes relevant to general cellular functions. When looking at the predicted upstream regulators from the feed efficiency network, the Tumor Protein P53 (TP53) and Transforming Growth Factor beta 1 (TGFB1) genes stood out regarding significance of overlap and number of target molecules in the data set. These results further support the hypothesis that TP53 is a major upstream regulator for genetic variation of feed efficiency. Furthermore, our data revealed a significant effect of both, the Non-SMC Condensin I Complex, Subunit G (NCAPG) I442M (rs109570900) and the Growth /differentiation factor 8 (GDF8) Q204X (rs110344317) loci, on residual feed intake and feed conversion. For both loci, the growth promoting allele at the onset of puberty was associated with a negative, but favorable effect on residual feed intake. The elevated energy demand for increased growth triggered by the NCAPG 442M allele is obviously not fully compensated for by an increased efficiency in converting feed into body tissue. As a consequence, the individuals carrying the NCAPG 442M allele had an additional demand for energy uptake that is reflected by the association of the allele with increased daily energy intake as observed in our study.


Assuntos
Proteínas de Ciclo Celular/genética , Ingestão de Alimentos/genética , Redes Reguladoras de Genes , Genoma , Genótipo , Fenótipo , Alelos , Ração Animal , Animais , Peso Corporal/genética , Bovinos , Cruzamentos Genéticos , Comportamento Alimentar/fisiologia , Expressão Gênica , Perfilação da Expressão Gênica , Loci Gênicos , Variação Genética , Estudo de Associação Genômica Ampla , Miostatina/genética , Característica Quantitativa Herdável , Fator de Crescimento Transformador beta1/genética , Proteína Supressora de Tumor p53/genética
4.
J Pineal Res ; 56(4): 439-49, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24650091

RESUMO

The pineal secretory product melatonin exerts its influence on the insulin secretion of pancreatic islets by different signaling pathways. The purpose of this study was to analyze the impact of melatonin on calcium-signaling components under different conditions. In a transfected INS-1 cell line overexpressing the human MT2 receptor (hMT2-INS-1), melatonin treatment induced even stronger depressive effects on calcium/calmodulin-dependent kinase 2d and IV (Camk2d, CamkIV) transcripts during 3-isobutyl-1-methylxanthine (IBMX) treatment than in normal INS-1 cells, indicating a crucial influence of melatonin receptor density on transcript-level regulation. In addition, melatonin induced a significant downregulation of calmodulin (Calm1) in IBMX-treated hMT2-INS-1 cells. Long-term administration of melatonin alone reduced CamkIV transcript levels in INS-1 cells; however, transcript levels of Camk2d remained unchanged. The release of insulin was diminished under long-term melatonin treatment. The impact of melatonin also involved reductions in CAMK2D protein during IBMX or forskolin treatments in INS-1 cells, as measured by an enzyme-linked immunosorbent assay, indicating a functional significance of transcriptional changes in pancreatic islets. Furthermore, analysis of melatonin receptor knockout mice showed that the transcript levels of Camk2d, CamkIV, and Calm1 were differentially influenced according to the melatonin receptor subtype deleted. In conclusion, this study provides evidence that melatonin has different impacts on the regulation of Calm1 and Camk. These calcium-signaling components are known as participants in the calcium/calmodulin pathway, which plays an important functional role in the modulation of the ß-cell signaling pathways leading to insulin secretion.


Assuntos
Antioxidantes/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Insulinoma/metabolismo , Melatonina/farmacologia , Neoplasias Pancreáticas/metabolismo , Animais , Sequência de Bases , Linhagem Celular Tumoral , Humanos , Insulinoma/genética , Insulinoma/patologia , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Ratos
5.
BMC Genomics ; 14: 798, 2013 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-24246134

RESUMO

BACKGROUND: Systems biology enables the identification of gene networks that modulate complex traits. Comprehensive metabolomic analyses provide innovative phenotypes that are intermediate between the initiator of genetic variability, the genome, and raw phenotypes that are influenced by a large number of environmental effects. The present study combines two concepts, systems biology and metabolic analyses, in an approach without prior functional hypothesis in order to dissect genes and molecular pathways that modulate differential growth at the onset of puberty in male cattle. Furthermore, this integrative strategy was applied to specifically explore distinctive gene interactions of non-SMC condensin I complex, subunit G (NCAPG) and myostatin (GDF8), known modulators of pre- and postnatal growth that are only partially understood for their molecular pathways affecting differential body weight. RESULTS: Our study successfully established gene networks and interacting partners affecting growth at the onset of puberty in cattle. We demonstrated the biological relevance of the created networks by comparison to randomly created networks. Our data showed that GnRH (Gonadotropin-releasing hormone) signaling is associated with divergent growth at the onset of puberty and revealed two highly connected hubs, BTC and DGKH, within the network. Both genes are known to directly interact with the GnRH signaling pathway. Furthermore, a gene interaction network for NCAPG containing 14 densely connected genes revealed novel information concerning the functional role of NCAPG in divergent growth. CONCLUSIONS: Merging both concepts, systems biology and metabolomic analyses, successfully yielded new insights into gene networks and interacting partners affecting growth at the onset of puberty in cattle. Genetic modulation in GnRH signaling was identified as key modifier of differential cattle growth at the onset of puberty. In addition, the benefit of our innovative concept without prior functional hypothesis was demonstrated by data suggesting that NCAPG might contribute to vascular smooth muscle contraction by indirect effects on the NO pathway via modulation of arginine metabolism. Our study shows for the first time in cattle that integration of genetic, physiological and metabolomics data in a systems biology approach will enable (or contribute to) an improved understanding of metabolic and gene networks and genotype-phenotype relationships.


Assuntos
Adenosina Trifosfatases/genética , Proteínas de Ligação a DNA/genética , Hormônio Liberador de Gonadotropina/genética , Complexos Multiproteicos/genética , Miostatina/genética , Maturidade Sexual/genética , Biologia de Sistemas , Animais , Peso Corporal/genética , Bovinos , Epistasia Genética , Perfilação da Expressão Gênica , Variação Genética , Masculino , Redes e Vias Metabólicas/genética , Metabolômica , Miostatina/biossíntese , Fenótipo , Polimorfismo de Nucleotídeo Único/genética
6.
Gen Comp Endocrinol ; 193: 167-77, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23954363

RESUMO

Agouti-related protein (AgRP), produced by neurons located in the arcuate nucleus of the hypothalamus stimulates feed intake. During early lactation dairy cows increase their feed intake and additionally mobilize their fat reserves leading to increased plasma non-esterified fatty acid (NEFA) concentrations. Since cows with a higher extent of fat mobilization exhibit the lower feed intake, it seems that high NEFA concentrations confine hyperphagia. To test the involvement of AgRP neurons, we investigated 18 cows from parturition until day 40 postpartum (pp) and assigned the cows according to their NEFA concentration on day 40pp to either group H (high NEFA) or L (low NEFA). Both groups had comparable feed intake, body weight, milk yield, energy balance, plasma amino acids and leptin concentrations. Studies in respiratory chambers revealed the higher oxygen consumption and the lower respiratory quotient (RQ) in H compared to L cows. mRNA abundance of neuropeptide Y, peroxisome proliferator-activated receptor-gamma, AMP-activated protein kinase, and leptin receptor in the arcuate nucleus were comparable between groups. Immunohistochemical studies revealed the same number of AgRP neurons in H and L cows. AgRP neurons were co-localized with phosphorylated adenosine monophosphate-activated kinase without any differences between groups. The percentage of cFOS-activated AgRP neurons per total AgRP cells was lower in H cows and correlated negatively with oxygen consumption and NEFA, positively with RQ, but not with feed intake. We conclude that AgRP activation plays a pivotal role in the regulation of substrate utilization and metabolic rate in high NEFA dairy cows during early lactation.


Assuntos
Proteína Relacionada com Agouti/metabolismo , Hipotálamo/metabolismo , Parto/metabolismo , Animais , Bovinos , Ingestão de Alimentos/fisiologia , Ácidos Graxos não Esterificados/sangue , Ácidos Graxos não Esterificados/metabolismo , Feminino , Lactação/fisiologia , Metabolismo dos Lipídeos/fisiologia , Consumo de Oxigênio/fisiologia , Parto/fisiologia , Respiração
7.
Am J Physiol Regul Integr Comp Physiol ; 303(9): R909-20, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22972839

RESUMO

Ruminal vacuolar H(+)-ATPase (vH(+)-ATPase) activity is regulated by metabolic signals. Thus, we tested whether its localization, expression, and activity were changed by different feeding. Young male sheep (n = 12) were either fed hay ad libitum (h) or hay ad libitum plus additional concentrate (h/c) for 2 wk. The vH(+)-ATPase B subunit signal was predominantly found in the cell membrane and cytosol of rumen epithelial cells (REC) with basal/parabasal phenotype. The elevated number (threefold) of these cells in rumen mucosa of h/c-fed sheep reflects a high proliferative capacity and, explains the 2.3-fold increase of the total number of vH(+)-ATPase-expressing REC. However, in accordance with a 58% reduction of the vH(+)-ATPase B subunit mRNA expression in h/c-fed sheep, its protein amount per single REC was decreased. Using the fluorescent probe BCECF and selective inhibitors (foliomycin, amiloride), the contribution of vH(+)-ATPase and Na(+)/H(+) exchanger to intracellular pH (pH(i)) regulation was investigated. REC isolated from h/c-fed sheep keep their pH(i) at a significantly higher level (6.91 ± 0.03 vs. 6.74 ± 0.05 in h-fed sheep). Foliomycin or amiloride decreased pH(i) by 0.16 ± 0.02 and 0.57 ± 0.04 pH units when applied to REC from h-fed sheep, but the effects were markedly reduced (-88 and -33%) after concentrate feeding. Nevertheless, we found that REC proliferation rate and [cAMP](i) were reduced after foliomycin-induced vH(+)-ATPase inhibition. Our results provide the first evidence for a role of vH(+)-ATPase in regulation of REC proliferation, most probably by linking metabolically induced pH(i) changes to signaling pathways regulating this process.


Assuntos
Adaptação Fisiológica/fisiologia , Ração Animal , Dieta , Metabolismo Energético/fisiologia , ATPases Translocadoras de Prótons/fisiologia , Rúmen/fisiologia , Ovinos/fisiologia , Amilorida/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/fisiologia , Epitélio/fisiologia , Concentração de Íons de Hidrogênio , Macrolídeos/farmacologia , Masculino , Rúmen/citologia , Rúmen/efeitos dos fármacos , Transdução de Sinais/fisiologia , Trocadores de Sódio-Hidrogênio/fisiologia
8.
J Pineal Res ; 53(4): 390-8, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22672634

RESUMO

Melatonin has been shown to modulate glucose metabolism by influencing insulin secretion. Recent investigations have also indicated a regulatory function of melatonin on the pancreatic α-cells. The present in vitro and in vivo studies evaluated whether melatonin mediates its effects via melatonin receptors and which signaling cascade is involved. Incubation experiments using the glucagon-producing mouse pancreatic α-cell line αTC1 clone 9 (αTC1.9) as well as isolated pancreatic islets of rats and mice revealed that melatonin increases glucagon secretion. Preincubation of αTC1.9 cells with the melatonin receptor antagonists luzindole and 4P-PDOT abolished the glucagon-stimulatory effect of melatonin. In addition, glucagon secretion was lower in the pancreatic islets of melatonin receptor knockout mice than in the islets of the wild-type (WT) control animals. Investigations of melatonin receptor knockout mice revealed decreased plasma glucagon concentrations and elevated mRNA expression levels of the hepatic glucagon receptor when compared to WT mice. Furthermore, studies using pertussis toxin, as well as measurements of cAMP concentrations, ruled out the involvement of Gαi- and Gαs-coupled signaling cascades in mediating the glucagon increase induced by melatonin. In contrast, inhibition of phospholipase C in αTC1.9 cells prevented the melatonin-induced effect, indicating the physiological relevance of the Gαq-coupled pathway. Our data point to the involvement of the phosphatidylinositol 3-kinase signaling cascade in mediating melatonin effects in pancreatic α-cells. In conclusion, these findings provide evidence that the glucagon-stimulatory effect of melatonin in pancreatic α-cells is melatonin receptor mediated, thus supporting the concept of melatonin-modulated and diurnal glucagon release.


Assuntos
Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Células Secretoras de Glucagon/efeitos dos fármacos , Glucagon/metabolismo , Melaninas/farmacologia , Fosfatidilinositol 3-Quinase/metabolismo , Receptor MT1 de Melatonina/efeitos dos fármacos , Receptor MT2 de Melatonina/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular , AMP Cíclico/metabolismo , Diabetes Mellitus Tipo 2/enzimologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Glucagon/sangue , Células Secretoras de Glucagon/enzimologia , Células Secretoras de Glucagon/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Toxina Pertussis/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptor MT1 de Melatonina/deficiência , Receptor MT1 de Melatonina/genética , Receptor MT2 de Melatonina/deficiência , Receptor MT2 de Melatonina/genética , Receptores de Glucagon/efeitos dos fármacos , Receptores de Glucagon/genética , Receptores de Glucagon/metabolismo , Tetra-Hidronaftalenos/farmacologia , Técnicas de Cultura de Tecidos , Triptaminas/farmacologia , Fosfolipases Tipo C/metabolismo
9.
J Pineal Res ; 53(4): 344-57, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22616931

RESUMO

The pineal hormone melatonin exerts its influence on the insulin secretion of pancreatic islets by a variety of signalling pathways. The purpose of the present study was to analyse the impact of melatonin on the phosphorylated transcription factor cAMP-response element-binding protein (pCREB). In pancreatic rat insulinoma ß-cells (INS-1), pCREB immunofluorescence intensities in cell nuclei using digitised confocal image analysis were measured to semi-quantify differences in the pCREB immunoreactivity (pCREB-ir) caused by different treatments. Increasing concentrations of forskolin or 3-isobutyl-1-methylxanthine (IBMX) resulted in a dose-dependent rise of the mean fluorescence intensity in pCREB-ir nuclear staining. Concomitant melatonin application significantly decreased pCREB-ir in INS-1 cells after 30-min, 1-hr and 3-hr treatment. The melatonin receptor antagonists luzindole and 4-phenyl-2-propionamidotetraline (4P-PDOT) completely abolished the pCREB phosphorylation-decreasing effect of melatonin, indicating that both melatonin receptor isoforms (MT(1) and MT(2)) are involved. In a transfected INS-1 cell line expressing the human MT(2) receptor, melatonin caused the greatest reduction in pCREB after IBMX treatment compared with nontransfected INS-1 cells, indicating a crucial influence of melatonin receptor density on pCREB regulation. Furthermore, the downregulation of pCREB by melatonin is concomitantly associated with a statistically significant downregulation of Camk2d transcript levels, as measured after 3 hr. In conclusion, the present study provides evidence that the phosphorylation level of CREB is modulated in pancreatic ß-cells by melatonin. Mediated via CREB, melatonin regulates the expression of genes that play an important functional role in the regulation of ß-cell signalling pathways.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Insulinoma/metabolismo , Melatonina/farmacologia , Neoplasias Pancreáticas/metabolismo , 1-Metil-3-Isobutilxantina/farmacologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Linhagem Celular Tumoral , Colforsina/farmacologia , Relação Dose-Resposta a Droga , Imunofluorescência , Humanos , Células Secretoras de Insulina/metabolismo , Insulinoma/genética , Microscopia Confocal , Neoplasias Pancreáticas/genética , Fosforilação , Ratos , Receptor MT1 de Melatonina/efeitos dos fármacos , Receptor MT1 de Melatonina/metabolismo , Receptor MT2 de Melatonina/efeitos dos fármacos , Receptor MT2 de Melatonina/genética , Receptor MT2 de Melatonina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tetra-Hidronaftalenos/farmacologia , Fatores de Tempo , Transfecção , Triptaminas/farmacologia
10.
J Pineal Res ; 52(4): 446-59, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22288848

RESUMO

Several studies have revealed that melatonin affects the insulin secretion via MT(1) and MT(2) receptor isoforms. Owing to the lack of selective MT(1) receptor antagonists, we used RNA interference technology to generate an MT(1) knockdown in a clonal ß-cell line to evaluate whether melatonin modulates insulin secretion specifically via the MT(1) receptor. Incubation experiments were carried out, and the insulin concentration in supernatants was measured using a radioimmunoassay. Furthermore, the intracellular cAMP was determined using an enzyme-linked immunosorbent assay. Real-time RT-PCR indicated that MT(1) knockdown resulted in a significant increase in the rIns1 mRNA and a significantly elevated basal insulin secretion of INS-1 cells. Incubation with melatonin decreased the amount of glucagon-like peptide 1 or inhibited the glucagon-stimulated insulin release of INS-1 cells, while, in MT(1) -knockdown cells, no melatonin-induced reduction in insulin secretion could be found. No decrease in 3-isobutyl-1-methylxanthine-stimulated intracellular cAMP in rMT(1) -knockdown cells was detectable after treatment with melatonin either, and immunocytochemistry proved that MT(1) knockdown abolished phosphorylation of cAMP-response-element-binding protein. In contrast to the INS-1 cells, preincubation with melatonin did not sensitize the insulin secretion of rMT(1) -knockdown cells. We also monitored insulin secretion from isolated islets of wild-type and melatonin-receptor knockout mice ex vivo. In islets of wild-type mice, melatonin treatment resulted in a decrease in insulin release, whereas melatonin treatment of islets from MT(1) knockout and MT(1/2) double-knockout mice did not show a significant effect. The data indicate that melatonin inhibits insulin secretion, primarily via the MT(1) receptor in rat INS-1 cells and isolated mouse islets.


Assuntos
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Melatonina/metabolismo , Receptor MT1 de Melatonina/metabolismo , 1-Metil-3-Isobutilxantina/farmacologia , Animais , Linhagem Celular Tumoral , AMP Cíclico/metabolismo , Histocitoquímica , Insulina/genética , Secreção de Insulina , Insulinoma/metabolismo , Camundongos , Camundongos Knockout , Ratos , Receptor MT1 de Melatonina/genética , Estatísticas não Paramétricas
11.
J Pineal Res ; 51(3): 361-72, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21585522

RESUMO

Melatonin exerts some of its effects via G-protein-coupled membrane receptors. Two membrane receptor isoforms, MT1 and MT2, have been described. The MT1 receptor is known to inhibit second messenger cyclic adenosine monophosphate (cAMP) signaling through receptor-coupling to inhibitory G-proteins (G(i) ). Much less is known about the MT2 receptor, but it has also been implicated in signaling via G(i) -proteins. In rat pancreatic ß-cells, it has recently been reported that the MT2 receptor plays an inhibitory role in the cyclic guanosine monophosphate (cGMP) pathway. This study addresses the signaling features of the constitutively expressed human recombinant MT2 receptor (hMT2) and its impact on insulin secretion, using a rat insulinoma ß-cell line (INS-1). On the basis of a specific radioimmunoassay, insulin secretion was found to be more strongly reduced in the clones expressing hMT2 than in INS-1 controls, when incubated with 1 or 100 nm melatonin. Similarly, cAMP and cGMP levels, measured by specific enzyme-linked immunosorbent assays (ELISAs), were reduced to a greater extent in hMT2 clones after melatonin treatment. In hMT2-expressing cells, the inhibitory effect of melatonin on insulin secretion was blocked by pretreatment with pertussis toxin, demonstrating the coupling of the hMT2 to G(i) -proteins. These results indicate that functional hMT2 expression leads to the inhibition of cyclic nucleotide signaling and a reduction in insulin release. Because genetic variants of the hMT2 receptor are considered to be risk factors in the development of type 2 diabetes, our results are potentially significant in explaining and preventing the pathogenesis of this disease.


Assuntos
Insulina/metabolismo , Insulinoma/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptor MT2 de Melatonina/genética , Animais , Sequência de Bases , Northern Blotting , Linhagem Celular Tumoral , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Secreção de Insulina , Radioimunoensaio , Ratos , Reação em Cadeia da Polimerase em Tempo Real
12.
Cell Mol Life Sci ; 66(15): 2525-37, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19488676

RESUMO

The regulated secretion of pancreatic zymogens depends on a functional cytoskeleton and intracellular vesicle transport. To study the dynamics of tubulin and its motor proteins dynein and kinesin during secretion in pancreatic acinar cells, we infused rats with 0.1 mug/kg/h caerulein. Electron and fluorescence microscopy detected neither dynein nor kinesin at the apical secretory pole, nor on the surface of mature zymogen granules. After 30 min of secretagogue stimulation, kinesin and the Golgi marker protein 58 K were reallocated towards the apical plasma membrane and association of kinesin with tubulin was enhanced. Disruption of acinar cell microtubules had no effect on initial caerulein-induced amylase release but completely blocked secretion during a second stimulus. Our results suggest that mature zymogen granule exocytosis is independent of intact microtubules, kinesin and dynein. However, microtubule-dependent mechanisms seem to be important for the replenishment of secretory vesicles by redistribution of Golgi elements towards the apical cell pole.


Assuntos
Dineínas/metabolismo , Cinesinas/metabolismo , Microtúbulos/metabolismo , Pâncreas/metabolismo , Suco Pancreático , Amilases/metabolismo , Animais , Ceruletídeo/metabolismo , Colchicina/farmacologia , Grânulos Citoplasmáticos/química , Grânulos Citoplasmáticos/metabolismo , Precursores Enzimáticos/metabolismo , Exocitose/fisiologia , Complexo de Golgi/metabolismo , Masculino , Microscopia Imunoeletrônica , Proteínas Motores Moleculares/metabolismo , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Suco Pancreático/química , Suco Pancreático/metabolismo , Ratos , Ratos Wistar , Tubulina (Proteína)/isolamento & purificação , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/farmacologia
13.
J Comp Physiol B ; 178(3): 285-95, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17989983

RESUMO

In this study, we have studied the expression, localization, and functionality of vacuolar-type H(+)-ATPase (vH(+)-ATPase) and Na(+)/K(+)-ATPase in the bovine rumen epithelium. Compared with the intracellular pH (pH(i)) of control rumen epithelial cells (REC; 7.06 +/- 0.07), application of inhibitors selective for vH(+)-ATPase (foliomycin) and Na(+)/K(+)-ATPase (ouabain) reduced pH(i) by 0.10 +/- 0.03 and 0.18 +/- 0.03 pH-units, respectively, thereby verifying the existence of both functional proteins. Results from qRT-PCR and immunoblotting clearly confirm the expression of vH(+)-ATPase B subunit in REC. However, the amount of Na(+)/K(+)-ATPase mRNA and protein is tenfold and 11-fold of those of vH(+)-ATPase subunit B, respectively, reflecting a lower overall abundance of the latter in REC. Na(+)/K(+)-ATPase immunostaining has revealed the protein in the plasma membrane of all REC from the stratum basale to stratum granulosum, with the highest abundance in basal cells. In contrast, the vH(+)-ATPase B subunit has been detected in groups of cells only, mainly localized in the stratum spinosum and stratum granulosum of the epithelium. Furthermore, vH(+)-ATPase has been detected in the cell membrane and in intracellular pools. Thus, functional vacuolar-type H(+) pumps are expressed in REC and probably play a role in the adaptation of epithelial transport processes.


Assuntos
Células Epiteliais/enzimologia , Rúmen/citologia , Rúmen/enzimologia , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Bovinos , Membrana Celular/enzimologia , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Concentração de Íons de Hidrogênio , Macrolídeos/farmacologia , Ouabaína/farmacologia , Subunidades Proteicas/metabolismo , RNA Mensageiro/metabolismo , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo , ATPases Vacuolares Próton-Translocadoras/efeitos dos fármacos , ATPases Vacuolares Próton-Translocadoras/genética
14.
Am J Physiol Gastrointest Liver Physiol ; 282(2): G367-74, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11804859

RESUMO

A premature and intracellular activation of digestive zymogens is thought to be responsible for the onset of pancreatitis. Because trypsin has a critical role in initiating the activation cascade of digestive enzymes in the gut, it has been assumed that trypsin also initiates intracellular zymogen activation in the pancreas. We have tested this hypothesis in isolated acini and lobules from rat pancreas. Intracellular trypsinogen activation was induced by supramaximal secretagogue stimulation and measured using either specific trypsin substrates or immunoreactivity of the trypsinogen activation peptide (TAP). To prevent a trypsin-induced trypsinogen activation, we used the cell-permeant, highly specific, and reversible inhibitor Nalpha-(2-naphthylsulfonyl)-3-amidinophenylalanine-carboxymethylpiperazide (S124), and to prevent cathepsin-induced trypsinogen activation, we used the cysteine protease inhibitor E-64d. Incubation of acini or lobules in the presence of S124 completely prevented the generation of trypsin activity in response to supramaximal caerulein but had no effect whatsoever on the generation of TAP. Conversely, when trypsin activity was recovered at the end of the experiment by either washout of S124 from acini or extensive dilution of lobule homogenates, it was up to 400% higher than after caerulein alone and corresponded, in molar terms, to the generation of TAP. Both trypsin activity and TAP release were inhibited in parallel by E-64d. We conclude that caerulein-induced trypsinogen activation in the pancreas is caused by an E-64d-inhibitable mechanism such as cathepsin-induced trypsinogen activation, and neither involves nor requires intracellular trypsin activity. Specific trypsin inhibition, on the other hand, prevents 80% of trypsin inactivation or autodegradation in the pancreas.


Assuntos
Pâncreas/enzimologia , Pancreatite/metabolismo , Tripsina/metabolismo , Tripsinogênio/metabolismo , Doença Aguda , Animais , Catepsina B/metabolismo , Ceruletídeo/farmacologia , Ativação Enzimática/fisiologia , Masculino , Naftalenos/farmacologia , Piperazinas/farmacologia , Ratos , Ratos Wistar , Inibidores da Tripsina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA