Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Oncogene ; 34(8): 1019-34, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-24632619

ABSTRACT

High cellular heterogeneity within neuroblastomas (NBs) may account for the non-uniform response to treatment. c-KIT(+) cells are frequently detected in NB, but how they influence NB behavior still remains elusive. Here, we used NB tumor-initiating cells to reconstitute NB development and demonstrated that c-KIT(+) cells are de novo generated and dynamically maintained within the tumors to sustain tumor progression. c-KIT(+) NB cells express higher levels of neural crest and stem cell markers (SLUG, SOX2 and NANOG) and are endowed with high clonogenic capacity, differentiation plasticity and are refractory to drugs. With serial transplantation assays, we found that c-KIT expression is not required for tumor formation, but c-KIT(+) cells are more aggressive and can induce tumors ninefold more efficiently than c-KIT(-/low) cells. Intriguingly, c-KIT(+) cells exhibited a long-term in vivo self-renewal capacity to sustain the formation of secondary and tertiary tumors in mice. In addition, we showed that Prokineticin signaling and mitogen-activated protein kinase pathways are crucial for the maintenance of c-KIT(+) cells in tumor to promote NB progression. Our results highlight the importance of this de novo population of NB cells in sustainable growth of NB and reveal specific signaling pathways that may provide targets leading to more effective NB therapies.


Subject(s)
Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neuroblastoma/pathology , Proto-Oncogene Proteins c-kit/metabolism , Vascular Endothelial Growth Factor, Endocrine-Gland-Derived/physiology , Animals , Cell Proliferation , Disease Progression , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Neuroblastoma/genetics , Neuroblastoma/metabolism , Proto-Oncogene Proteins c-kit/genetics , Signal Transduction , Tumor Cells, Cultured
2.
Mol Syndromol ; 4(1-2): 32-45, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23653575

ABSTRACT

Hedgehog (Hh) signaling is vital for the patterning and organogenesis of almost every system. The specificity of these developmental processes is achieved through a tight spatio-temporal regulation of Hh signaling. Mice with defective Hh signal exhibit a wide spectrum of anomalies, including Vertebral defects, Anal atresia, Cardiovascular anomalies, Tracheoesophageal fistula, Renal dysplasia, and Limb defects, that resemble strikingly the phenotypes observed in VACTERL association in humans. In this review, we summarize our current understanding of mammalian Hh signaling and highlight the relevance of various mouse models for studying the etiology and pathogenesis of VACTERL association. In addition, recent advances in genetic study for unraveling the complexity of genetic inheritance of VACTERL and the implication of the Sonic hedgehog pathway in disease pathogenesis are also discussed.

3.
Oncogene ; 32(35): 4086-99, 2013 Aug 29.
Article in English | MEDLINE | ID: mdl-23045286

ABSTRACT

Neuroblastoma (NB) is an embryonal tumor and possesses a unique propensity to exhibit either a spontaneous regression or an unrestrained growth. However, the underlying mechanism for this paradoxical clinical outcome remains largely unclear. Quantitative RT-PCR analysis on 102 primary NB tumors revealed that lower Krüppel-like factor 4 (KLF4) expression is frequently found in the unfavorable NB (Mann-Whitney test, P=0.027). In particular with the high-risk factors such as age of patient >1 year, MYCN amplification and low TRKA expression, the decreased expression of KLF4 was significantly associated with an unfavorable NB outcome. Despite knockdown of KLF4 alone is not sufficient to increase tumorigenicity of NB cells in vivo, stable expression of KLF4 short hairpin RNA in Be(2)-C cells significantly promoted growth of NB cells and inhibited cell differentiation toward fibromuscular lineage. In concordant with these observations, overexpression of KLF4 in SH-SY-5Y cells profoundly suppressed cell proliferation by direct upregulation of cell-cycle inhibitor protein p21(WAF1/CIP1), and knocking down p21(WAF1/CIP1) could partially rescue the suppressive effect of KLF4. Importantly, KLF4 overexpressing cells have lost their neuroblastic phenotypes, they were epithelial-like, strongly substrate-adherent, expressing smooth muscle marker and became non-tumorigenic, suggesting that KLF4 expression is crucial for lineage determination of NB cells, probably, favoring spontaneous tumor regression. Subsequent global gene expression profiling further revealed that transforming growth factor beta (TGFß) and cell-cycle pathways are highly dysregulated upon KLF4 overexpression, and myogenic modulators, MEF2A and MYOD1 were found significantly upregulated. Taken together, we have demonstrated that KLF4 contributes to the favorable disease outcome by directly mediating the growth and lineage determination of NB cells.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Proliferation , Kruppel-Like Transcription Factors/physiology , Neuroblastoma/pathology , Animals , Cell Cycle , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/analysis , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/analysis , Mice , N-Myc Proto-Oncogene Protein , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Transforming Growth Factor beta/genetics , Tumor Suppressor Protein p53/analysis
4.
J Med Genet ; 46(1): 32-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18782836

ABSTRACT

BACKGROUND: The BRCA1 gene is an important breast-cancer susceptibility gene. Promoter polymorphisms can alter the binding affinity of transcription factors, changing transcriptional activity and may affect susceptibility to disease. METHODS AND RESULTS: Using direct sequencing of the BRCA1 promoter region, we identified four polymorphisms c.-2804T-->C (rs799908:T-->C), c.-2265C-->T (rs11655505:C-->T), c.-2004A-->G (rs799906:A-->G) and c.-1896(ACA)(1)-->(ACA)(2) (rs8176071:(ACA)(1)-->(ACA)(2)) present in Hong Kong Chinese. Each polymorphism was studied independently and in combination by functional assays. Although all four variants significantly altered promoter activity, the c.-2265T allele had stronger binding than the C allele, and the most common mutant haplotype, which contains the c.-2265T allele, increased promoter activity by 70%. Risk association first tested in Hong Kong Chinese women with breast cancer and age-matched controls and replicated in a large population-based study of Shanghai Chinese, together totalling >3000 participants, showed that carriers of the c.-2265T allele had a reduced risk for breast cancer (combined odd ratio (OR) = 0.80, 95% CI 0.69 to 0.93; p = 0.003) which was more evident among women aged >or=45 years at first diagnosis of breast cancer and without a family history of breast cancer (combined OR = 0.75, 95% CI 0.61 to 0.91; p = 0.004). The most common haplotype containing the c.-2265T allele also showed significant risk association for women aged >or=45 years without a family history of breast cancer (OR = 0.64, 95% CI 0.46 to 0.89; p = 0.008). CONCLUSION: This comprehensive study of BRCA1 promoter polymorphisms found four variants that altered promoter activity and with the most significant contribution from c.-2265C-->T, which could affect susceptibility to breast cancer in the Chinese population. Its significance in other populations remains to be investigated.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Polymorphism, Genetic/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic , Asian People/genetics , Binding Sites , Breast Neoplasms/epidemiology , Case-Control Studies , China/epidemiology , Cohort Studies , Electrophoretic Mobility Shift Assay , Female , Genetic Predisposition to Disease , Genotype , Hong Kong/epidemiology , Humans , Risk Factors , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Ann Hum Genet ; 72(Pt 2): 170-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18081917

ABSTRACT

Hirschsprung's disease (HSCR, colonic aganglionosis) is an oligogenic entity that usually requires mutations in RET and other interacting loci. Decreased levels of RET expression may lead to the manifestation of HSCR. We previously showed that RET transcription was decreased due to alteration of the NKX2-1 binding site by two HSCR-associated RET promoter single nucleotide polymorphisms (SNPs). This prompted us to investigate whether DNA alterations in NKX2-1 could play a role in HSCR by affecting the RET-regulatory properties of the NKX2-1 protein. Our initial study on 86 Chinese HSCR patients revealed a Gly322Ser amino acid substitution in the NKX2-1 protein. In this study, we have examined 102 additional Chinese and 70 Caucasian patients and 194 Chinese and 60 Caucasian unselected, unrelated, subjects as controls. The relevance of the DNA changes detected in NKX2-1 by direct sequencing were evaluated using bioinformatics, reporter and binding-assays, mouse neurosphere culture, immunohistochemistry and immunofluorescence techniques. Met3Leu and Pro48Pro were identified in 2 Caucasian and 1 Chinese patients respectively. In vitro analysis showed that Met3Leu reduced the activity of the RET promoter by 100% in the presence of the wild-type or HSCR-associated RET promoter SNP alleles. The apparent binding affinity of the NKX2-1 mutated protein was not decreased. The Met3Leu mutation may affect the interaction of NKX2-1 with its protein partners. The absence of NKX2-1 expression in mouse but not in human gut suggests that the role of NKX2-1 in gut development differs between the two species. NKX2-1 mutations could contribute to HSCR by affecting RET expression through defective interactions with other transcription factors.


Subject(s)
Genetic Predisposition to Disease/genetics , Hirschsprung Disease/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-ret/metabolism , Transcription Factors/genetics , Animals , Asian People/genetics , Australia , Base Sequence , Cell Line, Tumor , China , Computational Biology , Digestive System/embryology , Digestive System/metabolism , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique , Gene Components , Genotype , Humans , Immunohistochemistry , Mice , Molecular Sequence Data , Mutation/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-ret/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Thyroid Nuclear Factor 1 , Transcription Factors/metabolism , White People/genetics
6.
Ann Hum Genet ; 71(Pt 4): 526-36, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17274802

ABSTRACT

Interactions between migrating neural crest cells and the environment of the gut are crucial for the development of the enteric nervous system (ENS). A key signalling mediator is the RET-receptor-tyrosine-kinase which, when defective, causes Hirschprung's disease (HSCR, colon aganglionosis). RET mutations alone cannot account for the variable HSCR phenotype, invoking interactions with as yet unknown, and probably inter-related, loci involved in ENS development. Homeobox (HOX) genes have a major role in gut development as depicted by the enteric Hox code. We investigated whether DNA alterations in HOX genes, either alone or in combination with RET, are implicated in HSCR. Genotyping effort was minimized by applying the HapMap data on Han Chinese from Beijing (CHB). 194 HSCR patients and 168 controls were genotyped using Sequenom technology for 72 tag, single nucleotide polymorphisms (SNPs) distributed along the HOX clusters. The HapMap frequencies were compared to those in our population and standard statistics were used for frequency comparisons. The multifactor-dimensionality-reduction method was used for multilocus analysis, in which RET promoter SNP genotypes were included. Genetic interactions were found between two HOX loci (5'-HOXA13 and 3'UTR-HOXB7) and the RET loci tested. Minor allele frequencies (MAF) of the SNPs tested in our sample were not significantly different from those reported by HapMap when the sample sizes of the populations compared were considered. This is the first evaluation of the HOX genes in HSCR and the first application of HapMap data in a Chinese population. The interacting HOX loci may affect the penetrance of the RET risk allele. HapMap data for the CHB population correlated well with the general Chinese population.


Subject(s)
Genetic Variation , Hirschsprung Disease/genetics , Homeodomain Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Alleles , Gene Frequency , Hirschsprung Disease/metabolism , Humans
7.
Mol Cell Biol ; 21(23): 8056-67, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11689696

ABSTRACT

We have previously demonstrated that overexpression of Cdc25B in transgenic mice resulted in mammary gland hyperplasia and increased steroid hormone responsiveness. To address how Cdc25B enhances the hormone responsiveness in mammary glands, we showed that Cdc25B stimulates steroid receptor-dependent transcription in transient transfection assays and in a cell-free assay with chromatin templates. Surprisingly, the effect of Cdc25B on steroid receptors is independent of its protein phosphatase activity in vitro. The direct interactions of Cdc25B with steroid receptors, on the other hand, were evidenced in in vivo and in vitro assays, suggesting the potential direct contribution of Cdc25B on the steroid receptor-mediated transcription. In addition, p300/CBP-associated factor and CREB binding protein were shown to interact and synergize with Cdc25B and further enhance its coactivation activity. Thus, we have uncovered a novel function of Cdc25B that serves as a steroid receptor coactivator in addition to its role as a regulator for cell cycle progression. This dual function might likely contribute to its oncogenic action in breast cancer.


Subject(s)
Breast/metabolism , Cell Cycle Proteins/metabolism , Receptors, Steroid/metabolism , Saccharomyces cerevisiae Proteins , cdc25 Phosphatases/metabolism , Acetyltransferases/metabolism , Animals , CREB-Binding Protein , Cell Cycle/physiology , Cell Cycle Proteins/pharmacology , Cell Line , Cell-Free System/metabolism , Cyclin D1/metabolism , Enzyme Activation/physiology , Female , Gene Expression Regulation/physiology , Histone Acetyltransferases , Humans , Nuclear Proteins/metabolism , Rats , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism , Trans-Activators/metabolism , Transcription Factors , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , cdc25 Phosphatases/pharmacology , p300-CBP Transcription Factors
8.
Mol Cell Endocrinol ; 176(1-2): 135-44, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11369453

ABSTRACT

Multiple transcription start sites were identified in the human gonadotropin releasing hormone receptor (hGnRHR) gene. Recently, an upstream promoter residing at -1727/-1674, in vicinity of a CAP site at -1673, was characterized. In this report, we elucidated the underlying mechanisms for the regulation of this promoter. Functionally, this promoter was constitutively suppressed by a silencer element (-1673/-1351) situated immediately downstream to it. On the other hand, pituitary adenylate cyclase-activating polypeptide (PACAP), via the cAMP pathway, was found to be the extracellular cue to control the upstream promoter. Following PACAP-27, PACAP-38 (30 nM) and forskolin (25 microM) treatment, there were significant increases in the reporter gene activities. By deletion analysis, the region residing at -1727 to -1577, containing the distal promoter and 97 bp of the silencer was subsequently found to be responsible for PACAP/cAMP induction. To localize the PACAP-dependent cis-acting element(s) within the silencer, block replacement scanning mutation was performed and a hGnRHR gene PACAP-responsive element (GPRE) was identified at -1676/-1648. The actions of PACAPs and forskolin on the GPRE were further evidenced by gel mobility shift assays. There was an increase in protein binding onto this element only after peptide treatment. As GnRH receptor number on gonadotrope cell surface is a key factor in regulating gonadotropin release, the present study provides an insight into the interplay between PACAP and GnRH receptors on pituitary gonadotropes to control human reproductive functions.


Subject(s)
Gene Silencing/drug effects , Neuropeptides/pharmacology , Promoter Regions, Genetic/genetics , Receptors, LHRH/genetics , Response Elements/genetics , Base Sequence , Binding, Competitive , Cell Line , Colforsin/pharmacology , Cyclic AMP/metabolism , DNA/genetics , DNA/metabolism , Down-Regulation/drug effects , Electrophoresis, Polyacrylamide Gel , Female , Genes, Reporter/genetics , Humans , Molecular Sequence Data , Mutation/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide , Protein Binding/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Tumor Cells, Cultured
9.
Endocrinology ; 141(10): 3611-22, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11014215

ABSTRACT

Clinical applications of GnRH agonists (GnRHa) are based primarily on the decrease in gonadotropin release after down-regulation of the GnRH receptor (GnRHR) by continuous GnRHa administration. However, the molecular mechanisms underlying the transcriptional regulation of the human GnRHR gene after prolonged GnRH treatment remain poorly understood. In the present study GnRHa-mediated regulation of human GnRHR gene transcription was studied by transiently transfecting the mouse gonadotrope-derived (alphaT3-1) cells with a 2297-bp human GnRHR promoter-luciferase construct (p2300-LucF). A dose- and time-dependent decrease in human GnRHR promoter activity was observed after GnRHa treatment. An average 71% decrease in promoter activity was observed after 24-h treatment with 0.1 microM GnRHa, which was blocked by cotreatment of the GnRH antagonist, antide. This effect was mimicked by phorbol 12-myristate 13-acetate (TPA) administration. In addition, the GnRHa- and TPA-mediated decrease in the human GnRHR promoter activity was reversed by a specific protein kinase C (PKC) inhibitor, GF109203X, or depletion of PKC by TPA pretreatment. These findings indicate that the activation of the PKC pathway is important in regulating the human GnRHR gene expression. By progressive 5'-deletion studies, we have identified a 248-bp DNA fragment (-1018 to -771, relative to the translation start site) at the 5'-flanking region of the human GnRHR gene that is responsible for the GnRHa-mediated down-regulation of human GnRHR promoter activity. Analysis of this sequence reveals the existence of two putative activating protein-1 (AP-1) sites with 87% homology to the consensus sequence (5'-TGA(G/C)T(C/A)A-3'), located at -1000 to -994 (5'-TTAGACA-3', in complementary orientation) and -943 to 937 (5'-TGAATAA-3'). Using competitive gel mobility shift assays, AP-1 binding was observed within this 248-bp region. Site-directed mutation of the putative AP-1-binding site located at -1000 to -994 abolished the GnRHa-induced inhibition. Further competitive GMSA and supershift experiments confirmed the identity of AP-1 binding in this region. By the use of Western blot analysis, a significant increase in c-Jun (100%; P < 0.05) and c-Fos (50%; P < 0.05) protein levels was observed after GnRHa treatment in alphaT3-1 cells. In addition, our data suggested that a change in AP-1 composition, particularly c-Fos, was important in mediating GnRHa-induced inhibition of human GnRHR gene expression. We conclude that activation of the PKC pathway by GnRH is important in controlling human GnRHR gene expression. In addition, the putative AP-1-binding site located at -1000 to -994 of the human GnRHR5'-flanking region has been functionally identified to be involved in mediating this down-regulatory effect.


Subject(s)
Down-Regulation , Gonadotropin-Releasing Hormone/physiology , Protein Kinase C/physiology , Receptors, LHRH/genetics , Transcription Factor AP-1/physiology , Transcription, Genetic/physiology , Animals , Cell Line , Gonadotropin-Releasing Hormone/agonists , Hormone Antagonists/pharmacology , Humans , Luciferases/metabolism , Mice , Oligopeptides/pharmacology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/physiology , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Response Elements/physiology , Tetradecanoylphorbol Acetate/pharmacology
10.
Mol Cell Endocrinol ; 162(1-2): 157-66, 2000 Apr 25.
Article in English | MEDLINE | ID: mdl-10854709

ABSTRACT

In terms of regulation of gene expression, gonadotropin-releasing hormone receptor (GnRHR) found in extrapituitary tissues has been suggested to be different from that in the pituitary. In the present study, we examined the molecular basis of this difference using the pituitary alphaT3-1 and ovarian carcinoma OVCAR-3 cells. As a first step, the different expression levels of GnRHR mRNA in the pituitary and ovarian cells were investigated using semi-quantitative RT-PCR. Quantitative analysis showed that the expression level of hGnRHR is a nine-fold higher in primary pituitary tissues than the primary culture of ovarian carcinomas (PCO). In pituitary alphaT3-1 cells, the expression level of hGnRHR was ten-fold higher than ovarian carcinoma OVCAR-3 cells. The possibility of the differential use of various cell-specific promoters in different cells was addressed by transiently transfecting cells with 3'-deletion clones of human GnRHR promoter. Sequential deletion of the 5'-flanking region of the gene revealed the use of two putative promoters, designated PR1 (-771 to -557) and PR2 (-1351 to -1022), and a negative control region (-1022 to -771), in the pituitary and ovarian cells. The same promoters appeared to be utilized for driving the basal promoter activities in both alphaT3-1 and OVCAR-3 cells, suggesting that there is no cell-specific promoter usage for the human GnRHR gene. Alternatively, the involvement of different regulatory protein factors was investigated using electrophoretic gel mobility shift assays. When end-labeled PR1 was used as a probe, two unique shifted complexes were identified in OVCAR-3 cells when compared to alphaT3-1 cells. One unique protein-DNA complex was observed in alphaT3-1 cells compared to OVCAR-3 cells when incubated with end-labeled PR2 as a probe. These DNA-protein complexes appeared to be specific, as they competed with excess amount of unlabelled competitor PR1 and PR2, respectively. In summary, it is unlikely that the use of a cell-specific promoter contributes to the different characteristics of ovarian GnRHR. Our study demonstrates that one mechanism by which cell-specific expression of human GnRHR is achieved is through the binding of distinct and/or combinations of cell-specific regulatory factors to various promoter elements in the 5'-flanking region of the gene.


Subject(s)
Ovary/metabolism , Pituitary Gland/metabolism , Receptors, LHRH/genetics , Animals , Base Sequence , Cell Line , DNA Primers/genetics , Female , Gene Expression , Humans , Mice , Ovarian Neoplasms/genetics , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Tissue Distribution , Transfection , Tumor Cells, Cultured
11.
Biochem Biophys Res Commun ; 270(3): 766-72, 2000 Apr 21.
Article in English | MEDLINE | ID: mdl-10772899

ABSTRACT

Analysis of the human gonadotropin-releasing hormone receptor (hGnRHR) gene 5' flanking region revealed the presence of multiple TATA, CCAAT, and transcription start sites. In addition, at least three different transcripts (5.0, 2.5, and 1.5 kb) were detected by Northern blot analysis. Taken together, these data indicated the existence of multiple promoter elements in the hGnRHR gene, and these promoters are responsible for the multiplicity of regulation of human reproductive functions. In this report, by progressive 5' and 3' deletion (-2197 to -1351, relative to the ATG) and NotI linker scanning mutagenesis coupled to transient transfection into the mouse gonadotrope-derived alphaT3-1 cell, a distal promoter element was identified at -1705/-1674. The promoter was located immediately 5' to a previously identified CAP site at -1673 in human pituitary and it drove a 17.6- +/- 1.0-fold increase in reporter gene activity. Within the promoter, a pyrimidine-rich initiator element (Inr) (-1682) and a CCAAT box (-1702) were found and mutation of these elements abrogated both protein bindings and promoter activities. By 1- and 2-D SouthWestern blot assays, multiple nuclear factors (40 to 54 kDa) were found to interact specifically with this promoter element. These nuclear factors were also present in other cells, including COS-7, JEG-3, and SKOV-3 cells, and these findings were consistent with functional studies which showed that the promoter is also active in these cells.


Subject(s)
Promoter Regions, Genetic , Receptors, LHRH/genetics , Animals , Base Sequence , COS Cells , Humans , Mice , Molecular Sequence Data , Mutagenesis, Insertional , Mutagenesis, Site-Directed , Nuclear Proteins/metabolism , Oligodeoxyribonucleotides , Pituitary Gland/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Sequence Deletion , Transfection , Tumor Cells, Cultured
12.
FEBS Lett ; 457(3): 499-504, 1999 Sep 03.
Article in English | MEDLINE | ID: mdl-10471837

ABSTRACT

In this report, the first amphibian glucagon receptor (GluR) cDNA was characterized from the liver of the frog Rana tigrina rugulosa. Functional expression of the frog GluR in CHO and COS-7 cells showed a high specificity of the receptor towards human glucagon with an EC(50) value of 0.8+/-0.5 nM. The binding of radioiodinated human glucagon to GluR was displaced in a dose-dependent manner only with human glucagon and its antagonist (des-His(1)-[Nle(9)-Ala(11)-Ala(16)]) with IC(50) values of 12.0+/-3. 0 and 7.8+/-1.0 nM, respectively. The frog GluR did not display any affinity towards fish and human GLP-1s, and towards glucagon peptides derived from two species of teleost fishes (goldfish, zebrafish). These fish glucagons contain substitutions in several key residues that were previously shown to be critical for the binding of human glucagon to its receptor. By RT-PCR, mRNA transcripts of frog GluR were located in the liver, brain, small intestine and colon. These results demonstrate a conservation of the functional characteristics of the GluRs in frog and mammalian species and provide a framework for a better understanding of the molecular evolution of the GluR and its physiological function in vertebrates.


Subject(s)
Evolution, Molecular , Ranidae/genetics , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Venoms , Vertebrates/physiology , Amino Acid Sequence , Animals , Binding, Competitive , CHO Cells/metabolism , COS Cells , Cricetinae , Cyclic AMP/metabolism , Exenatide , Glucagon/analogs & derivatives , Glucagon/antagonists & inhibitors , Glucagon/metabolism , Glucagon/pharmacology , Glucagon-Like Peptide 1 , Humans , Inhibitory Concentration 50 , Molecular Sequence Data , Neuropeptides/metabolism , Neuropeptides/pharmacology , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Peptides/metabolism , Peptides/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide , Protein Precursors/metabolism , Protein Precursors/pharmacology , Receptors, Glucagon/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Secretin/metabolism , Secretin/pharmacology , Sequence Homology, Amino Acid , Species Specificity , Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/pharmacology , Zebrafish
13.
FEBS Lett ; 455(3): 209-14, 1999 Jul 23.
Article in English | MEDLINE | ID: mdl-10437774

ABSTRACT

Secretin is the most potent regulator of pancreatic bicarbonate, electrolyte and volume secretion. In this report, the organization of the human secretin receptor (hSR) gene was characterized by overlapping genomic phage clones. The hSR gene consists of 13 exons and 12 introns with all the splice donor and acceptor sites conforming to the canonical GT/AG rule. By transient reporter gene assays, the wild-type promoter, containing 3.0 kb of the hSR gene 5' flanking region, was able to drive 5.8 +/- 0.6 and 6.6 +/- 0.2-fold (P < 0.01) increases in luciferase activities in pancreatic ductule-derived PANC-1 and BPD-1 cells, respectively. By subsequent 5' and 3' deletion analysis, a promoter element was identified within -408 to -158, relative to the ATG codon. This promoter element was found to be cell-specific since it could drive reporter gene expression in PANC-1 and BPD-1 cells but not in Hs 262.St, Hs 746T and alphaT3-1 cells. The study of the transcriptional control of human secretin and its receptor should shed light on the pathological developments of pancreatic cancer and autism in the future.


Subject(s)
Promoter Regions, Genetic , Receptors, Gastrointestinal Hormone/genetics , Secretin/metabolism , Animals , Autistic Disorder/genetics , Base Sequence , Cell Line , Chromosome Mapping , DNA/genetics , DNA Probes/genetics , Exons , Genes, Reporter , Genome, Human , Humans , Introns , Mice , Molecular Sequence Data , Receptors, G-Protein-Coupled
14.
Endocrinology ; 140(6): 2452-62, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10342829

ABSTRACT

GnRH plays a pivotal role in regulating human reproductive functions. This hypothalamic peptide interacts with its receptor (GnRHR) on the pituitary gonadotropes to trigger the secretion of gonadotropins, which, in turn, regulates the release of sex steroids from the gonads. In light of the importance of GnRHR, the molecular mechanisms underlying the transcriptional regulation of the human GnRHR (hGnRHR) gene become a key issue in understanding human reproduction. In this report, the possible involvement of steriodogenic factor-1 (SF-1) as a key cell-specific regulator for hGnRHR gene expression was examined. By the transient luciferase reporter gene assays, the wild-type promoter, containing 2.3 kb ofthe hGnRHR gene 5'-flanking region relative to the ATG codon, was able to drive a 3.6 +/- 0.2-fold (P < 0.05) increase in luciferase activity in the mouse alphaT3-1 gonadotropes. Subsequent deletion analysis indicated that the most proximal 173 bp within the first exon of the gene, although not a promoter itself, contains a critical regulatory element(s) essential for the basal expression of the hGnRHR gene. The functional roles of the putative gonadotrope-specific elements (GSE; consensus 5'-CTG(A)/(T)CCTTG-3') residing at positions -5, -134, and -396 were studied by site-directed mutagenesis, and it was found that only the mutation at position -134 significantly reduced the promoter activity (80% reduction; P < 0.05). The attenuation effect of this GSE mutant was cell specific, as it was restricted to alphaT3-1 cells, but not to COS-7 and human ovarian adenocarcinoma (SKOV-3) cells. Competitive mobility shift assays using either alphaT3-1 nuclear extract or recombinant SF-1 protein clearly indicated that SF-1 is able to interact specifically with this GSE element positioned at -134. Using a SF-1 antibody that completely abrogated complex formation in the gel shift assays, the involvement of endogenous nuclear SF-1 was further evidenced. By competitive gel shift assays using oligoprimers with 2-bp scanning mutations, the sequences essential for the interaction with SF-1 were identified (5'-TTG(A)/(T)CCCTG-3', underlined sequences were important). To study the in vivo function of SF-1, vector directing expression of sense or antisense SF-1 messenger RNA (mRNA) was cotransfected with the hGnRHR promoter-luciferase construct into alphaT3-1, SKOV-3, and COS-7 cells. Overexpression of the SF-1 mRNA was able to enhance promoter activities in all of the cells tested. On the contrary, expression of the antisense SF-1 mRNA reduced the hGnRHR promoter activity only in alphaT3-1 cells, not in COS-7 or SKOV-3 cells. In summary, the data reported here provide conclusive evidence that SF-1 interacts with the GSE motif at position -134 within the first exon of the hGnRHR gene to mediate its cell-specific expression.


Subject(s)
DNA-Binding Proteins/physiology , Exons , Gene Expression Regulation , Receptors, LHRH/genetics , Transcription Factors/physiology , 5' Untranslated Regions , Animals , Base Sequence , Cell Line , Fushi Tarazu Transcription Factors , Homeodomain Proteins , Humans , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear , Steroidogenic Factor 1 , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL