Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Diabetol Metab Syndr ; 7: 84, 2015.
Article in English | MEDLINE | ID: mdl-26435753

ABSTRACT

BACKGROUND: Insulinomas are the most common functional pancreatic neuroendocrine tumors, whereas histopathological features do not predict their biological behaviour. In an attempt to better understand the molecular processes involved in the tumorigenesis of islet beta cells, the present study evaluated the expression of genes belonging to the hepatocyte growth factor and its receptor (HGF/MET) system, namely, MET, HGF; HGFAC and ST14 (encode HGF activator and matriptase, respectively, two serine proteases that catalyze conversion of pro-HGF to active HGF); and SPINT1 and SPINT2 (encode serine peptidase inhibitors Kunitz type 1 and type 2, respectively, two inhibitors of HGF activator and of matriptase). METHODS: Quantitative real-time reverse transcriptase polymerase chain reaction was employed to assess RNA expression of the target genes in 24 sporadic insulinomas: 15 grade 1 (G1), six grade 2 (G2) and three hepatic metastases. Somatic mutations of MET gene were searched by direct sequencing of exons 2, 10, 14, 16, 17 and 19. RESULTS: Overexpression of MET was observed in the three hepatic metastases concomitantly with upregulation of the genes encoding HGF and matriptase and downregulation of SPINT1. A positive correlation was observed between MET RNA expression and Ki-67 proliferation index while a negative correlation was detected between SPINT1 expression and the mitotic index. No somatic mutations were found in MET gene. CONCLUSION: The final effect of the increased expression of HGF, its activator (matriptase) and its specific receptor (MET) together with a decreased expression of one potent inhibitor of matriptase (SPINT1) is probably a contribution to tumoral progression and metastatization in insulinomas.

2.
BMC Cancer ; 13: 451, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-24088503

ABSTRACT

BACKGROUND: Human homeobox genes encode nuclear proteins that act as transcription factors involved in the control of differentiation and proliferation. Currently, the role of these genes in development and tumor progression has been extensively studied. Recently, increased expression of HOXB7 homeobox gene (HOXB7) in pancreatic ductal adenocarcinomas (PDAC) was shown to correlate with an invasive phenotype, lymph node metastasis and worse survival outcomes, but no influence on cell proliferation or viability was detected. In the present study, the effects arising from the knockdown of HOXB7 in PDAC cell lines was investigated. METHODS: Real time quantitative PCR (qRT-PCR) (Taqman) was employed to assess HOXB7 mRNA expression in 29 PDAC, 6 metastatic tissues, 24 peritumoral tissues and two PDAC cell lines. siRNA was used to knockdown HOXB7 mRNA in the cell lines and its consequences on apoptosis rate and cell proliferation were measured by flow cytometry and MTT assay respectively. RESULTS: Overexpression of HOXB7 mRNA was observed in the tumoral tissues and in the cell lines MIA PaCa-2 and Capan-1. HOXB7 knockdown elicited (1) an increase in the expression of the pro-apoptotic proteins BAX and BAD in both cell lines; (2) a decrease in the expression of the anti-apoptotic protein BCL-2 and in cyclin D1 and an increase in the number of apoptotic cells in the MIA PaCa-2 cell line; (3) accumulation of cell in sub-G1 phase in both cell lines; (4) the modulation of several biological processes, especially in MIA PaCa-2, such as proteasomal ubiquitin-dependent catabolic process and cell cycle. CONCLUSION: The present study confirms the overexpression of HOXB7 mRNA expression in PDAC and demonstrates that decreasing its protein level by siRNA could significantly increase apoptosis and modulate several biological processes. HOXB7 might be a promising target for future therapies.


Subject(s)
Apoptosis/genetics , Carcinoma, Pancreatic Ductal/genetics , Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Pancreatic Neoplasms/genetics , RNA, Messenger , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Proliferation , E2F Transcription Factors/genetics , Gene Dosage , Gene Expression Profiling , Gene Knockdown Techniques , Gene Silencing , Humans , Pancreatic Neoplasms/metabolism , RNA, Small Interfering/genetics , Reproducibility of Results , Retinoblastoma Protein/genetics
3.
Arq. bras. endocrinol. metab ; 56(8): 485-489, Nov. 2012. ilus
Article in English | LILACS | ID: lil-660254

ABSTRACT

The hyperinsulinism/hyperammonemia (HI/HA) syndrome is a rare autosomal dominant disease manifested by hypoglycemic symptoms triggered by fasting or high-protein meals, and by elevated serum ammonia. HI/HA is the second most common cause of hyperinsulinemic hypoglycemia of infancy, and it is caused by activating mutations in GLUD1, the gene that encodes mitochondrial enzyme glutamate dehydrogenase (GDH). Biochemical evaluation, as well as direct sequencing of exons and exon-intron boundary regions of the GLUD1 gene, were performed in a 6-year old female patient presenting fasting hypoglycemia and hyperammonemia. The patient was found to be heterozygous for one de novo missense mutation (c.1491A>G; p.Il497Met) previously reported in a Japanese patient. Treatment with diazoxide 100 mg/day promoted complete resolution of the hypoglycemic episodes. Arq Bras Endocrinol Metab. 2012;56(8):485-9.


A síndrome de hiperinsulinemia/hiperamonemia (HI/HA) é uma condição rara, de herança autossômica dominante, que se manifesta por sintomas de hipoglicemia desencadeada por jejum ou refeições de alto conteúdo proteico, juntamente com elevação da concentração de amônia sérica. HI/HA é a segunda causa de hipoglicemia hiperinsulinêmica da infância e é causada por mutações ativadoras no GLUD1, o gene que codifica a enzima mitocondrial glutamato desidrogenase (GDH). A avaliação bioquímica, bem como o sequenciamento direto dos éxons e junções éxon-íntron do gene GLUD1, foi realizada em uma paciente de 6 anos de idade com hipoglicemia de jejum e hiperamonemia. A paciente apresentava uma mutação de novo missense (c.1491A>G; p.Il497Met) em heterozigose, que havia sido previamente relatada em um paciente japonês. O tratamento com diazóxido 100 mg/dia promoveu resolução completa dos episódios hipoglicêmicos. Arq Bras Endocrinol Metab. 2012;56(8):485-9.


Subject(s)
Child , Female , Humans , Glutamate Dehydrogenase/genetics , Hyperinsulinism/genetics , Hypoglycemia/genetics , Mutation, Missense/genetics
4.
Arq Bras Endocrinol Metabol ; 56(8): 485-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23295286

ABSTRACT

The hyperinsulinism/hyperammonemia (HI/HA) syndrome is a rare autosomal dominant disease manifested by hypoglycemic symptoms triggered by fasting or high-protein meals, and by elevated serum ammonia. HI/HA is the second most common cause of hyperinsulinemic hypoglycemia of infancy, and it is caused by activating mutations in GLUD1, the gene that encodes mitochondrial enzyme glutamate dehydrogenase (GDH). Biochemical evaluation, as well as direct sequencing of exons and exon-intron boundary regions of the GLUD1 gene, were performed in a 6-year old female patient presenting fasting hypoglycemia and hyperammonemia. The patient was found to be heterozygous for one de novo missense mutation (c.1491A>G; p.Il497Met) previously reported in a Japanese patient. Treatment with diazoxide 100 mg/day promoted complete resolution of the hypoglycemic episodes.


Subject(s)
Glutamate Dehydrogenase/genetics , Hyperinsulinism/genetics , Hypoglycemia/genetics , Mutation, Missense/genetics , Child , Female , Humans
5.
BMC Med Genet ; 11: 3, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-20051115

ABSTRACT

BACKGROUND: Glycogen storage disease type 0 is an autosomal recessive disease presenting in infancy or early childhood and characterized by ketotic hypoglycemia after prolonged fasting and postprandial hyperglycemia and hyperlactatemia. Sixteen different mutations have been identified to date in the gene which encodes hepatic glycogen synthase, resulting in reduction of glycogen storage in the liver. CASE PRESENTATION: Biochemical evaluation as well as direct sequencing of exons and exon-intron boundary regions of the GYS2 gene were performed in a patient presenting fasting hypoglycemia and postprandial hyperglycemia and her parents. The patient was found to be compound heterozygous for one previously reported nonsense mutation (c.736 C>T; R243X) and a novel frameshift mutation (966_967delGA/insC) which introduces a stop codon 21 aminoacids downstream from the site of the mutation that presumably leads to loss of 51% of the COOH-terminal part of the protein. The glycemia and lactatemia of the parents after an oral glucose tolerance test were evaluated to investigate a possible impact of the carrier status on the metabolic profile. The mother, who presented a positive family history of type 2 diabetes, was classified as glucose intolerant and the father, who did not exhibit metabolic changes after the glucose overload, had an antecedent history of hypoglycemia after moderate alcohol ingestion. CONCLUSION: The current results expand the spectrum of known mutations in GYS2 and suggest that haploinsufficiency could explain metabolic abnormalities in heterozygous carriers in presence of predisposing conditions.


Subject(s)
Codon, Nonsense , Frameshift Mutation , Glycogen Storage Disease/genetics , Glycogen Synthase/genetics , Base Sequence , Blood Glucose/analysis , Child , Exons , Fasting , Female , Glycogen Storage Disease/diagnosis , Heterozygote , Humans , Insulin/analysis , Introns , Lactic Acid/analysis , Phenotype , Sequence Analysis, DNA
6.
J Gastroenterol Hepatol ; 25(2): 357-61, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19817962

ABSTRACT

BACKGROUND AND AIMS: Although the metabolic risk factors for non-alcoholic fatty liver disease (NAFLD) progression have been recognized, the role of genetic susceptibility remains a field to be explored. The aim of this study was to examine the frequency of two polymorphisms in Brazilian patients with biopsy-proven simple steatosis or non-alcoholic steatohepatitis (NASH): -493 G/T in the MTP gene, which codes the protein responsible for transferring triglycerides to nascent apolipoprotein B, and -129 C/T in the GCLC gene, which codes the catalytic subunit of glutamate-cystein ligase in the formation of glutathione. METHODS: One hundred and thirty-one biopsy-proven NAFLD patients (n = 45, simple steatosis; n = 86, NASH) and 141 unrelated healthy volunteers were evaluated. Genomic DNA was extracted from peripheral blood cells, and the -129 C/T polymorphism of the GCLC gene was determined by restriction fragment length polymorphism (RFLP). The -493 G/T polymorphism of the MTP gene was determined by direct sequencing of the polymerase chain reaction products. RESULTS: The presence of at least one T allele in the -129 C/T polymorphism of the GCLC gene was independently associated with NASH (odds ratio 12.14, 95% confidence interval 2.01-73.35; P = 0.007), whereas, the presence of at least one G allele in the -493 G/T polymorphism of the MTP gene differed slightly between biopsy-proven NASH and simple steatosis. CONCLUSION: This difference clearly warrants further investigation in larger samples. These two polymorphisms could represent an additional factor for consideration in evaluating the risk of NAFLD progression. Further studies involving a larger population are necessary to confirm this notion.


Subject(s)
Carrier Proteins/genetics , Fatty Liver/genetics , Glutamate-Cysteine Ligase/genetics , Polymorphism, Genetic , Biopsy , Brazil , Case-Control Studies , Chi-Square Distribution , Fatty Liver/enzymology , Fatty Liver/pathology , Female , Gene Frequency , Genetic Predisposition to Disease , Humans , Liver/enzymology , Liver/pathology , Male , Middle Aged , Odds Ratio , Phenotype , Polymerase Chain Reaction , Promoter Regions, Genetic , Risk Assessment , Risk Factors
7.
Am J Physiol Renal Physiol ; 297(4): F916-22, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19656910

ABSTRACT

Sirolimus, an antiproliferative immunosuppressant, induces hypomagnesemia and hypokalemia. Rosiglitazone activates renal sodium- and water-reabsorptive pathways. We evaluated whether sirolimus induces renal wasting of magnesium and potassium, attempting to identify the tubule segments in which this occurs. We tested the hypothesis that reduced expression of the cotransporter NKCC2 forms the molecular basis of this effect and evaluated the possible association between increased urinary excretion of magnesium and renal expression of the epithelial Mg2+ channel TRPM6. We then analyzed whether rosiglitazone attenuates these sirolimus-induced tubular effects. Wistar rats were treated for 14 days with sirolimus (3 mg/kg body wt in drinking water), with or without rosiglitazone (92 mg/kg body wt in food). Protein abundance of NKCC2, aquaporin-2 (AQP2), and TRPM6 was assessed using immunoblotting. Sirolimus-treated animals presented no change in glomerular filtration rate, although there were marked decreases in plasma potassium and magnesium. Sirolimus treatment reduced expression of NKCC2, and this was accompanied by greater urinary excretion of sodium, potassium, and magnesium. In sirolimus-treated animals, AQP2 expression was reduced. Expression of TRPM6 was increased, which might represent a direct stimulatory effect of sirolimus or a compensatory response. The finding that rosiglitazone prevented or attenuated all sirolimus-induced renal tubular defects has potential clinical implications.


Subject(s)
Hypoglycemic Agents/therapeutic use , Hypokalemia/prevention & control , Immunosuppressive Agents/adverse effects , Sirolimus/adverse effects , Sodium-Potassium-Chloride Symporters/metabolism , Thiazolidinediones/therapeutic use , Animals , Aquaporin 2/metabolism , Drug Interactions , Hypokalemia/chemically induced , Immunosuppressive Agents/blood , Kidney/metabolism , Kidney Function Tests , Magnesium/blood , Magnesium/urine , Male , Polyuria/chemically induced , Rats , Rats, Wistar , Rosiglitazone , Sirolimus/blood , Sodium/metabolism , Solute Carrier Family 12, Member 1 , TRPM Cation Channels/metabolism , Water/metabolism
8.
Neuroendocrinology ; 89(2): 163-70, 2009.
Article in English | MEDLINE | ID: mdl-18791324

ABSTRACT

BACKGROUND/AIMS: Prolactin (PRL) secretion and its gene expression are inhibited by dopamine. Prolactinomas are the most common secreting pituitary adenomas, and dopamine agonists (DA) are the first choice for their treatment. However, a subset of patients is resistant to DA. As the mechanisms involved in DA resistance are not fully understood, the aim of this study was to obtain new insights regarding the molecular differences between the prolactinomas that are responsive to DA and those that are resistant. METHODS: Tumor tissue samples were collected from 17 patients who harbored prolactinomas, which were classified as responsive or resistant according to their clinical and laboratorial reaction to DA. The expression of 6 genes was evaluated by real-time polymerase chain reaction: dopamine receptor type 2 (DRD(2)), nerve growth factor-beta (NGFB) and its receptor (NGFR), estrogen receptor-alpha (ERA), estrogen receptor-beta (ERB) and the pituitary tumor transforming gene (PTTG). RESULTS: Median DRD(2) and NGFR expression in responsive patients was significantly higher than in resistant ones (p = 0.029 and p = 0.020, respectively). Moreover, the expressions of DRD(2) and NGFR were positively correlated with PRL decrease during treatment (r = 0.66, p = 0.005 and r = 0.57, p = 0.044, respectively). Furthermore, ERB expression was positively correlated to PTTG expression (r = 0.68, p = 0.032) and negatively correlated to NGFB expression (r = -0.75, p = 0.02). CONCLUSIONS: DRD(2) and NGFR expressions are related to the responsiveness of prolactinoma to DA. However, PTTG, ERB and ERA expressions are not. Also ERB, ERA and PTTG expressions did not present a clear correlation to tumor aggressiveness. Furthermore, the response of prolactinomas to DA should be viewed as a spectrum ranging from the most responsive to the most resistant ones.


Subject(s)
Dopamine Agonists/therapeutic use , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/metabolism , Prolactinoma/drug therapy , Prolactinoma/metabolism , Adult , Drug Resistance, Neoplasm/genetics , Female , Gene Expression , Humans , Male , RNA, Messenger/metabolism
9.
Cell Tissue Res ; 327(3): 529-37, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17149594

ABSTRACT

Hepatic oval cells (OC) are considered to be facultative liver stem cells and, because they may undergo differentiation into a variety of cell lineages, they might have the potential to be used in cellular therapy. Signals delivered by extracellular matrix (ECM) proteins take part in cellular differentiation in cooperation with signals from growth factors; indeed, some ECM proteins, such as laminin (LAM) and fibronectin (FN), have been shown to contribute to beta-cell differentiation and islet development, respectively. Since no previous studies have investigated the effect of ECM proteins on the expression of islet cell markers by cultured OC, the purpose of the present study was to evaluate whether FN and LAM modulate the expression of genes related to the endocrine pancreas in these liver cells. OC proliferation was induced in Wistar rats by prolonged treatment with 2-acetoaminofluorene/allyl alcohol and confirmed by reverse transcription/polymerase chain reaction and hepatic immunocytochemical and histopathological analyses. OC isolation was performed by Ficoll gradient and magnetic-activated cell sorting. OC were cultured for 1 and 2 months under several conditions with specific growth factors, over a FN or LAM substrate or in high glucose, nicotinamide and fetal calf serum. OC cultured on FN substrate expressed Pdx-1, Pax-6, insulin 2 and glucagon. LAM also induced the expression of Pdx-1, insulin 1 and insulin 2, glucagon, somatostatin and GLUT-2. Our results suggest that these ECM proteins can be used in protocols of OC transdifferentiation aimed at reducing the period necessary for complete transdifferentiation.


Subject(s)
Fibronectins/pharmacology , Insulin-Secreting Cells/metabolism , Laminin/pharmacology , Liver/drug effects , Pancreatic Hormones/metabolism , Stem Cells/drug effects , 2-Acetylaminofluorene/toxicity , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Separation , Cells, Cultured , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Expression/drug effects , Glucagon/genetics , Glucagon/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin/genetics , Insulin/metabolism , Liver/cytology , Liver/metabolism , Male , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Pancreatic Hormones/genetics , Propanols/toxicity , Rats , Rats, Wistar , Repressor Proteins/genetics , Repressor Proteins/metabolism , Specific Pathogen-Free Organisms , Stem Cells/cytology , Stem Cells/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
10.
Pituitary ; 9(2): 83-92, 2006.
Article in English | MEDLINE | ID: mdl-16832584

ABSTRACT

Pituitary tumors, adenomas in their vast majority, represent around 10-15% of the intracranial neoplasms. Pituitary carcinomas are exceedingly rare. Clinically, these neoplasms cause hormonal dysfunctions, and mass effect symptoms as headache and visual disorders in the case of macroadenomas. Pituitary tumorigenesis is still poorly understood. In order to investigate the expression of cancer-related genes in pituitary tumors, we employed a human cancer cDNA macroarray membrane with 1176 well-characterized human genes related to cancer and tumor biology. We were able to identify several differentially expressed genes, among them hepatocyte growth factor-regulated tyrosine kinase substrate (HGS) and guanylate kinase 1 (GUK1) which were over expressed in a pool of clinically nonfunctioning pituitary adenomas, compared with a spinal cord metastasis of a nonfunctioning pituitary carcinoma. HGS and GUK1 mRNA expression were chosen to be validated by quantitative RT-qPCR, however, only GUK1 had the differential expression confirmed between the adenomas and the metastasis of a pituitary carcinoma. We have also investigated HGS and GUK1 mRNA expressions in a series of 46 pituitary adenomas (18 nonfunctioning, 12 GH-secreting, nine PRL-secreting, and seven ACTH-secreting adenomas). HGS and GUK1 were significantly over expressed in GH-secreting adenomas, compared with ACTH-secreting adenomas and nonfunctioning tumors, and with PRL-secreting adenomas, respectively. We have shown that these genes, involved in tumorigenesis in other tissues, are as well over expressed in the pituitary tumors, however, their role in the oncogenesis of these tumors need to be further investigated.


Subject(s)
Adenoma/metabolism , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Guanylate Kinases/metabolism , Phosphoproteins/metabolism , ACTH-Secreting Pituitary Adenoma/genetics , ACTH-Secreting Pituitary Adenoma/metabolism , Adenoma/genetics , Adolescent , Adult , Aged , DNA, Neoplasm/genetics , Endosomal Sorting Complexes Required for Transport , Female , Gene Expression Regulation, Neoplastic , Growth Hormone-Secreting Pituitary Adenoma/genetics , Guanylate Kinases/genetics , Humans , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Phosphoproteins/genetics , Prolactinoma/genetics , Prolactinoma/metabolism , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...