Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Endocrinol ; 198(1): 29-39, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18417529

ABSTRACT

In this study, an insulinoma-associated antigen-1 (INSM1)-binding site in the proximal promoter sequence of the insulin gene was identified. The co-transfection of INSM1 with rat insulin I/II promoter-driven reporter genes exhibited a 40-50% inhibitory effect on the reporter activity. Mutational experiments were performed by introducing a substitution, GG to AT, into the INSM1 core binding site of the rat insulin I/II promoters. The mutated insulin promoter exhibited a three- to 20-fold increase in the promoter activity over the wild-type promoter in several insulinoma cell lines. Moreover, INSM1 overexpression exhibited no inhibitory effect on the mutated insulin promoter. Chromatin immunoprecipitation assays using beta TC-1, mouse fetal pancreas, and Ad-INSM1-transduced human islets demonstrated that INSM1 occupies the endogenous insulin promoter sequence containing the INSM1-binding site in vivo. The binding of the INSM1 to the insulin promoter could suppress approximately 50% of insulin message in human islets. The mechanism for transcriptional repression of the insulin gene by INSM1 is mediated through the recruitment of cyclin D1 and histone deacetylase-3 to the insulin promoter. Anti-INSM1 or anti-cyclin D1 morpholino treatment of fetal mouse pancreas enhances the insulin promoter activity. These data strongly support the view that INSM1 is a new zinc-finger transcription factor that modulates insulin gene transcription during early pancreas development.


Subject(s)
DNA-Binding Proteins/physiology , Insulin/genetics , Promoter Regions, Genetic , Repressor Proteins/physiology , Animals , Binding Sites , Cell Line, Tumor , Cyclin D1/physiology , Gene Expression Regulation , Histone Deacetylases/physiology , Humans , Mice , Pancreas/embryology , Rats , Transcription, Genetic
2.
FEBS Lett ; 581(5): 949-54, 2007 Mar 06.
Article in English | MEDLINE | ID: mdl-17300785

ABSTRACT

INSM1 is a downstream target gene of neurogenin 3 (ngn3). A promoter construct containing the -426/+40bp region transiently co-transfected into NIH-3T3 cells with a ngn3 expression plasmid resulted in a 12-fold increase in promoter activity. The ngn3/E47 heterodimer selectively binds and activates the E-box3 of the INSM1 promoter. The endogenous ngn3 and CREB-binding protein (CBP) co-activator occupy the INSM1 promoter, resulting in hyper-acetylation of histone H3/H4 chromatin in a human neuroblastoma cell line, IMR-32. Additionally, adenoviral ngn3 can induce endogenous INSM-1 expression in pancreatic ductal carcinoma-1 cells through the recruitment of CBP to the INSM1 promoter and increase the acetylation of the INSM1 promoter region.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carrier Proteins/metabolism , DNA-Binding Proteins/genetics , Histones/metabolism , Nerve Tissue Proteins/metabolism , Repressor Proteins/genetics , Transcription Factors/genetics , Acetylation , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Carrier Proteins/genetics , Cell Line , Corticosterone , DNA Primers/genetics , Gene Expression Regulation , Histones/chemistry , Humans , Mice , NIH 3T3 Cells , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
3.
J Cell Biochem ; 93(2): 215-23, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15368349

ABSTRACT

Heparanase (HPSE-1) is an endo-beta-D-glucuronidase involved in the degradation of cell-surface/extracellular matrix heparan sulfate (HS) in normal and neoplastic tissues. HPSE-1 represents the first example of purification and cloning of a mammalian HS-degradative enzyme. Elevated HPSE-1 levels are known to be associated with metastatic cancers, directly implicating HPSE-1 in metastatic events. The purpose of this study was to determine the role of cAMP response element-binding protein (CREB) in modulating HPSE-1-mediated effects on human melanoma cell invasion. Highly invasive, brain-metastatic melanoma cells (70W) were transfected with the dominant-negative CREB (KCREB) and subsequently analyzed for changes in their HPSE-1 content, functionality, and cell invasive properties. KCREB-transfected cells showed a decrease in HPSE-1 mRNA expression and activity. This correlated with a significantly decreased invasion of these cells through Matrigel-coated filters. Furthermore, adenoviral vectors containing the full-length human HPSE-1 cDNA in sense orientation (Ad-S/hep) were constructed to investigate CREB effects on HPSE-1. Restoration of HPSE-1 expression and functionality following Ad-S/hep infection of KCREB-transfected 70W cells recovered melanoma cell invasiveness. These results demonstrate that KCREB inhibits HPSE-1 and suggest that one of the roles CREB plays in the acquisition of melanoma cells metastatic phenotype is affecting HPSE-1 activity.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Glucuronidase/antagonists & inhibitors , Glucuronidase/metabolism , Melanoma/metabolism , Melanoma/pathology , Adenoviridae/genetics , Cell Line, Tumor , Collagen , Cyclic AMP/metabolism , DNA, Complementary/genetics , Drug Combinations , Gene Expression Regulation, Neoplastic , Genes, Dominant/genetics , Glucuronidase/genetics , Humans , Laminin , Melanoma/enzymology , Melanoma/genetics , Mutation/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Proteoglycans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
4.
Neuro Oncol ; 6(2): 154-65, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15134630

ABSTRACT

Brain metastasis, which occurs in 20% to 40% of all cancer patients, is an important cause of neoplastic morbidity and mortality. Successful invasion into the brain by tumor cells must include attachment to microvessel endothelial cells, penetration through the blood-brain barrier, and, of relevance, a response to brain survival and growth factors. Neurotrophins (NTs) are important in brain-invasive steps. Human melanoma cell lines express low-affinity NT receptor p75NTR in relation to their brain-metastatic propensity with their invasive properties being regulated by NGF, or nerve growth factor, the prototypic NT. They also express functional TrkC, the putative receptor for the invasion-promoting NT-3. In brain-metastatic melanoma cells, NTs promote invasion by enhancing the production of extracellular matrix (ECM)-degradative enzymes such as heparanase, an enzyme capable of locally destroying both ECM and the basement membrane of the blood-brain barrier. Heparanase is an endo-beta-d-glucuronidase that cleaves heparan sulfate (HS) chains of ECM HS proteoglycans, and it is a unique metastatic determinant because it is the dominant mammalian HS degradative enzyme. Brain-metastatic melanoma cells also produce autocrine/paracrine factors that influence their growth, invasion, and survival in the brain. Synthesis of these factors may serve to regulate NT production by brain cells adjacent to the neoplastic invasion front, such as astrocytes. Increased NT levels have been observed in tumor-adjacent tissues at the invasion front of human brain melanoma. Additionally, astrocytes may contribute to the brain-metastatic specificity of melanoma cells by producing NT-regulated heparanase. Trophic, autocrine, and paracrine growth factors may therefore determine whether metastatic cells can successfully invade, colonize, and grow in the CNS.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Melanoma/metabolism , Melanoma/secondary , Nerve Growth Factors/physiology , Animals , Humans , Receptors, Nerve Growth Factor/physiology
5.
J Cell Biochem ; 91(1): 206-15, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14689592

ABSTRACT

The p75 neurotrophin receptor (p75(NTR)), a common receptor for members of the neurotrophins (NT) family, was previously identified as a molecular determinant of brain metastasis. We have also reported that NT treatment of murine and human brain-metastatic melanoma cells affects their invasive capacities and increases the production of heparanase, an important and unique extracellular matrix (ECM) degradative enzyme. Neurotrophism can be a survival-support mechanism for brain-metastatic cells and a survival assay was devised to mimic the growth limiting conditions of rapidly expanding metastatic tumors prior to neoangiogenesis. We report that p75(NTR) promoted the survival of brain-metastatic melanoma cells but not melanocytes in stress cultures conditions. Secondly, melanoma cells fluorescently sorted for high p75(NTR) expression (p75(NTR-H) cells) had an up to a 15-fold greater survival than those sorted for low p75(NTR) expression (p75(NTR-L) cells). Thirdly, cells overexpressing p75(NTR) associated with the growth fraction and provided these cells with an inherent growth advantage. Finally, we observed an increased survival of sorted p75(NTR-L) cells, dependent upon treatment of NT members whose functional receptors are present on these cells. Together, these results delineate that p75(NTR)-mediated trophic support profoundly affects competitive melanoma-cell survival when the tumor cell microenvironment becomes growth limiting.


Subject(s)
Brain Neoplasms/metabolism , Melanoma/metabolism , Nerve Growth Factors/pharmacology , Receptors, Nerve Growth Factor/metabolism , Skin Neoplasms/metabolism , Animals , Brain Neoplasms/secondary , Cell Survival/drug effects , Cell Survival/physiology , Flow Cytometry , Glucuronidase/metabolism , Humans , Melanocytes/metabolism , Mice , Receptor, Nerve Growth Factor , Signal Transduction/physiology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...