Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
4.
Biol Open ; 3(5): 372-8, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24728959

ABSTRACT

The HMGA1 and HMGA2 genes code for proteins belonging to the High Mobility Group A family. Several genes are negatively or positively regulated by both these proteins, but a number of genes are specifically regulated by only one of them. Indeed, knock-out of the Hmga1 and Hmga2 genes leads to different phenotypes: cardiac hypertrophy and type 2 diabetes in the former case, and a large reduction in body size and amount of fat tissue in the latter case. Therefore, to better elucidate the functions of the Hmga genes, we crossed Hmga1-null mice with mice null for Hmga2. The Hmga1(-/-)/Hmga2(-/-) mice showed reduced vitality and a very small size (75% smaller than the wild-type mice); they were even smaller than pygmy Hmga2-null mice. The drastic reduction in E2F1 activity, and consequently in the expression of the E2F-dependent genes involved in cell cycle regulation, likely accounts for some phenotypic features of the Hmga1(-/-)/Hmga2(-/-) mice.

5.
Endocr Relat Cancer ; 19(2): 123-35, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22199144

ABSTRACT

We have previously demonstrated that HMGA1B and HMGA2 overexpression in mice induces the development of GH and prolactin (PRL) pituitary adenomas mainly by increasing E2F1 transcriptional activity. Interestingly, these adenomas showed very high expression levels of PIT1, a transcriptional factor that regulates the gene expression of Gh, Prl, Ghrhr and Pit1 itself, playing a key role in pituitary gland development and physiology. Therefore, the aim of our study was to identify the role of Pit1 overexpression in pituitary tumour development induced by HMGA1B and HMGA2. First, we demonstrated that HMGA1B and HMGA2 directly interact with both PIT1 and its gene promoter in vivo, and that these proteins positively regulate Pit1 promoter activity, also co-operating with PIT1 itself. Subsequently, we showed, by colony-forming assays on two different pituitary adenoma cell lines, GH3 and αT3, that Pit1 overexpression increases pituitary cell proliferation. Finally, the expression analysis of HMGA1, HMGA2 and PIT1 in human pituitary adenomas of different histological types revealed a direct correlation between PIT1 and HMGA expression levels. Taken together, our data indicate a role of Pit1 upregulation by HMGA proteins in pituitary tumours.


Subject(s)
HMGA1a Protein/metabolism , HMGA2 Protein/metabolism , Pituitary Neoplasms/metabolism , Transcription Factor Pit-1/metabolism , Up-Regulation/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , HMGA1a Protein/genetics , HMGA2 Protein/genetics , Humans , Linear Models , Pituitary Neoplasms/genetics , Promoter Regions, Genetic/physiology , RNA/chemistry , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor Pit-1/biosynthesis , Transcription Factor Pit-1/genetics , Transcriptional Activation/physiology
6.
Eur J Cancer ; 47(3): 470-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21044834

ABSTRACT

HMGA1 gene rearrangements have been frequently described in human lipomas. In vitro studies suggest that HMGA1 proteins have a negative role in the control of adipocyte cell growth, and that HMGA1 gene truncation acts in a dominant-negative fashion. Therefore, to define better the role of the HMGA1 alterations in the generation of human lipomas, we generated mice carrying an Hmga1b truncated (Hmga1b/T) gene. These mice develop a giant phenotype together with a drastic expansion of the retroperitoneal and subcutaneous white adipose tissue. We show that the activation of the E2F pathway likely accounts, at least in part, for this phenotype. Interestingly, the Hmga1b/T mice also develop B-cell lymphomas similar to that occurring in Hmga1-knockout mice, supporting a dominant-negative role of the Hmga1b/T mutant also in vivo.


Subject(s)
Gigantism/genetics , HMGA1b Protein/genetics , Lipomatosis/genetics , Lymphoma, B-Cell/genetics , Adipocytes/pathology , Animals , Cell Proliferation , E2F1 Transcription Factor/metabolism , Female , HMGA1b Protein/metabolism , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Male , Mice , Mice, Transgenic , Mutation/genetics , Phenotype , Reverse Transcriptase Polymerase Chain Reaction
7.
Cancer Res ; 70(13): 5379-88, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20530667

ABSTRACT

We have previously described a mechanism through which the high-mobility group A1 (HMGA1) proteins inhibit p53-mediated apoptosis by delocalizing the p53 proapoptotic activator homeodomain-interacting protein kinase 2 from the nucleus to the cytoplasm. By this mechanism, HMGA1 modulates the transcription of p53 target genes such as Mdm2, p21(waf1), and Bax, inhibiting apoptosis. Here, we report that HMGA1 antagonizes the p53-mediated transcriptional repression of another apoptosis-related gene, Bcl-2, suggesting a novel mechanism by which HMGA1 counteracts apoptosis. Moreover, HMGA1 overexpression promotes the reduction of Brn-3a binding to the Bcl-2 promoter, thereby blocking the Brn-3a corepressor function on Bcl-2 expression following p53 activation. Consistently, a significant direct correlation between HMGA1 and Bcl-2 overexpression has been observed in human breast carcinomas harboring wild-type p53. Therefore, this study suggests a novel mechanism, based on Bcl-2 induction, by which HMGA1 overexpression contributes to the escape from apoptosis leading to neoplastic transformation.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genes, bcl-2 , HMGA1a Protein/genetics , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis/physiology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Down-Regulation , Female , HMGA1a Protein/biosynthesis , Humans , Mice , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Transcription Factor Brn-3A/genetics , Transcription Factor Brn-3A/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism
8.
Endocr Relat Cancer ; 16(2): 483-90, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19261681

ABSTRACT

Impairment of the p27(kip1) function, caused by a drastic reduction of its expression or cytoplasmic mislocalization, has been frequently observed in thyroid carcinomas. To understand the role of p27(kip1) impairment in thyroid carcinogenesis, we investigated the consequences of the loss of p27(kip1) expression in the context of a mouse modeling of papillary thyroid cancer, expressing the TRK-T1 oncogene under the transcriptional control of thyroglobulin promoter. We found that double mutant mice homozygous for a p27(kip1) null allele (TRK-T1/p27(-/-)) display a higher incidence of papillary thyroid carcinomas, with a shorter latency period and increased proliferation index, compared with p27(kip1) wild-type compounds (TRK-T1/p27(+/+)). Consistently, double mutant mice heterozygous for a p27(kip1) null allele (TRK-T1/p27(+/-)) show an incidence of thyroid carcinomas that is intermediate between TRK-T1/p27(-/-) and TRK-T1/p27(+/+) mice. Therefore, our findings suggest a dose-dependent role of p27(kip1) function in papillary thyroid cancer development.


Subject(s)
Carcinoma, Papillary/pathology , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/physiology , Oncogene Proteins/physiology , Thyroid Neoplasms/pathology , Animals , Blotting, Western , Carcinoma, Papillary/genetics , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Survival Rate , Thyroid Neoplasms/genetics
9.
Cancer Res ; 69(5): 1844-50, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19223528

ABSTRACT

The high mobility group As (HMGAs) belong to a family of nonhistone nuclear proteins that orchestrate the assembly of nucleoprotein complexes. Through a complex network of protein-DNA and protein-protein interaction, they play important roles in gene transcription, recombination, and chromatin structure. This protein family is involved, through different mechanisms, in both benign and malignant neoplasias. We have recently reported that transgenic mice carrying the Hmga1 or Hmga2 genes under transcriptional control of the cytomegalovirus promoter develop pituitary adenomas secreting prolactin and growth hormone. We have shown that the mechanism of the HMGA2-induced pituitary adenoma is based on the increased E2F1 activity. The expression profile of mouse normal pituitary glands and adenomas induced in HMGA transgenic mice revealed an increased expression of the ccnb2 gene, coding for the cyclin B2 protein, in the neoplastic tissues compared with the normal pituitary gland. Here, we show, by electrophoretic mobility shift assay and chromatin immunoprecipitation, a direct binding of HMGA proteins to the promoter of ccnb2 gene, whereas luciferase assays showed that HMGAs are able to up-regulate ccnb2 promoter activity. Finally, we report an increased CCNB2 expression in human pituitary adenomas of different histotypes that is directly correlated with HMGA1 and HMGA2 expression. Because cyclin B2 is involved in the regulation of the cell cycle, these results taken together indicate that HMGA-induced cyclin B2 overexpression gives an important contribution to experimental and human pituitary tumorigenesis.


Subject(s)
Adenoma/genetics , Cyclin B/genetics , Gene Expression Regulation, Neoplastic , HMGA1a Protein/physiology , HMGA2 Protein/physiology , Pituitary Neoplasms/genetics , Animals , Cyclin B2 , Humans , Mice , Mice, Transgenic , NIH 3T3 Cells , Promoter Regions, Genetic , Rats , Up-Regulation
10.
Cancer Cell ; 13(3): 272-86, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18328430

ABSTRACT

Deregulation of E2F1 activity and resistance to TGFbeta are hallmarks of gastric cancer. MicroRNAs (miRNAs) are small noncoding RNAs frequently misregulated in human malignancies. Here we provide evidence that the miR-106b-25 cluster, upregulated in a subset of human gastric tumors, is activated by E2F1 in parallel with its host gene, Mcm7. In turn, miR-106b and miR-93 regulate E2F1 expression, establishing a miRNA-directed negative feedback loop. Furthermore, upregulation of these miRNAs impairs the TGFbeta tumor suppressor pathway, interfering with the expression of CDKN1A (p21(Waf1/Cip1)) and BCL2L11 (Bim). Together, these results suggest that the miR-106b-25 cluster is involved in E2F1 posttranscriptional regulation and may play a key role in the development of TGFbeta resistance in gastric cancer.


Subject(s)
Adenocarcinoma/genetics , Apoptosis/genetics , Cell Cycle/genetics , E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , Stomach Neoplasms/genetics , Transforming Growth Factor beta/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E2F1 Transcription Factor/genetics , Feedback, Physiological , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Minichromosome Maintenance Complex Component 7 , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Time Factors , Transfection , Up-Regulation
11.
Eur J Cancer ; 44(2): 318-25, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18155899

ABSTRACT

The HMGA1 proteins have a critical role in the process of carcinogenesis. They are overexpressed in most human malignant neoplasias, and the inhibition of their expression has been shown to prevent cell transformation and results in malignant cell death. To determine whether HMGA1 proteins are also required for in vivo carcinogenesis, we compared the tumour susceptibility of mice wild-type or knockout for the Hmga1-null allele using a two-stage chemical skin carcinogenesis protocol. Hmga1-/- mice exhibited a decreased number and a delayed onset of skin papillomas in comparison with wild-type mice. Moreover, the progression of skin papillomas to carcinomas was observed in only 5% of Hmga1-/- compared to 18% of wild-type mice. These results suggest a lower susceptibility of Hmga1-/- mice to skin carcinogenesis induced by chemical agents.


Subject(s)
HMGA Proteins/genetics , Papilloma/chemically induced , Skin Neoplasms/chemically induced , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Animals , Carcinogens/toxicity , Cell Transformation, Neoplastic/genetics , Genetic Predisposition to Disease/genetics , Keratinocytes/metabolism , Mice , Mice, Knockout , Papilloma/genetics , Proto-Oncogene Proteins c-fos/metabolism , Skin Neoplasms/genetics , Tetradecanoylphorbol Acetate/toxicity , rab GTP-Binding Proteins/metabolism
12.
Endocr Relat Cancer ; 14(3): 791-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17914108

ABSTRACT

We have recently reported that MicroRNAs (miR)-221 and miR-222 were up-regulated in human thyroid papillary carcinomas in comparison with the normal thyroid tissue. Bioinformatic analysis proposed the p27(Kip1) protein, a key regulator of cell cycle, as a candidate target for the miR-221/222 cluster. Here, we report that the enforced expression of miR-221 and miR-222 was able to reduce p27(Kip1) protein levels in thyroid carcinoma and HeLa cells in the absence of significant changes in specific p27(Kip1) mRNA levels. This effect is direct as miR-221 and miR-222 negatively regulate the expression of the 3'-untranslated region-based reporter construct from the p27(Kip1) gene, and is dependent on two target sites in this region. Consistent with these results, an enforced expression of the miR-221 and miR-222 induced the thyroid papillary carcinoma cell line (TPC-1) to progress to the S phase of the cell cycle. It is likely that the negative regulation of p27(Kip1) by miR-221 and miR-222 might also have a role in vivo since we report an inverse correlation between miR-221 and miR-222 up-regulation and down-regulation of the p27(Kip1) protein levels in human thyroid papillary carcinomas. Therefore, the data reported here demonstrate that miR-221 and miR-222 are endogenous regulators of p27(Kip1) protein expression, and thereby, the cell cycle.


Subject(s)
Carcinoma, Papillary/genetics , Cell Cycle/genetics , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , MicroRNAs/genetics , MicroRNAs/physiology , Thyroid Neoplasms/genetics , Base Sequence , Cyclin-Dependent Kinase Inhibitor p27 , Down-Regulation , HeLa Cells , Humans , Tumor Cells, Cultured
13.
Endocr Relat Cancer ; 14(3): 875-86, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17914116

ABSTRACT

The high-mobility group A (HMGA) family of proteins orchestrates the assembly of nucleoprotein structures playing important roles in gene transcription, recombination, and chromatin structure through a complex network of protein-DNA and protein-protein interactions. Recently, we have generated transgenic mice carrying wild type or truncated HMGA2 genes under the transcriptional control of the cytomegalovirus promoter. These mice developed pituitary adenomas secreting prolactin and GH mainly due to an increased E2F1 activity, directly consequent to the HMGA2 overexpression. To identify other genes involved in the process of pituitary tumorigenesis induced by the HMGA2 gene, in this study we have analyzed the gene expression profile of three HMGA2-pituitary adenomas in comparison with a pool of ten normal pituitary glands from control mice, using the Affymetrix MG MU11K oligonucleotide array representing approximately 13,000 unique genes. We have identified 82 transcripts that increased and 72 transcripts that decreased at least four-fold in all the mice pituitary adenomas analyzed compared with normal pituitary glands. Among these genes, we focused our attention on the Mia/Cd-rap gene, whose expression was essentially suppressed in all of the pituitary adenomas tested by the microarray. We demonstrated that the HMGA proteins directly bind to the promoter of the Mia/Cd-rap gene and are able to downregulate its expression. In order to understand a possible role of Mia/Cd-rap in pituitary cell growth, we performed a colony assay in GH3 and GH4 cells. Interestingly, Mia/Cd-rap expression inhibits their proliferation, suggesting a potential tumor suppressor role of Mia/Cd-rap in pituitary cells.


Subject(s)
Adenoma/genetics , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Neoplastic , HMGA Proteins/physiology , Pituitary Neoplasms/genetics , Animals , Cell Proliferation , Cluster Analysis , Down-Regulation , Gene Expression Profiling , Mice , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Rats , Tumor Cells, Cultured
14.
Clin Cancer Res ; 13(9): 2738-44, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17473207

ABSTRACT

PURPOSE: We have previously shown that transgenic mice ubiquitously overexpressing the HMGA2 gene develop growth hormone/prolactin-secreting pituitary adenomas. This animal model has been used to evaluate the therapeutic efficacy of SOM230, a somatostatin analogue with high affinity for the somatostatin receptor subtypes 1, 2, 3, and 5, on the growth of the pituitary adenomas. EXPERIMENTAL DESIGN: Four groups of 3- and 9-month-old HMGA2 transgenic mice were treated for 3 months with a continuous s.c. injection of two different dosages of SOM230 (5 or 50 microg/kg/h), one dose of octreotide, corresponding to that used in human therapy, and a placebo, respectively. The development of the tumor before and after therapy was monitored by magnetic resonance imaging of the pituitary region and evaluation of the serum prolactin levels. RESULTS: The highest dose of SOM230 induced a drastic regression of the tumor, whereas octreotide was not able to induce any significant tumor regression, although tumor progression was significantly slowed down. No significant differences were observed between the animals treated with the lowest dose of SOM230 and those receiving placebo. CONCLUSIONS: These results clearly support the efficacy of the SOM230 treatment in human pituitary adenomas secreting prolactin based on the dramatic tumor shrinkage and fall in prolactin levels. This beneficial effect could be of crucial clinical usefulness in patients bearing tumors resistant to dopaminergic drugs.


Subject(s)
Growth Hormone-Secreting Pituitary Adenoma/drug therapy , Pituitary Neoplasms/drug therapy , Somatostatin/analogs & derivatives , Animals , Cell Proliferation/drug effects , Female , Growth Hormone-Secreting Pituitary Adenoma/pathology , HMGA2 Protein/genetics , Mice , Mice, Transgenic , Pituitary Neoplasms/pathology , Somatostatin/pharmacology , Somatostatin/therapeutic use , Treatment Outcome
15.
Cancer Cell ; 9(6): 459-71, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16766265

ABSTRACT

HMGA2 gene amplification and overexpression in human prolactinomas and the development of pituitary adenomas in HMGA2 transgenic mice showed that HMGA2 plays a crucial role in pituitary tumorigenesis. We have explored the pRB/E2F1 pathway to investigate the mechanism by which HMGA2 acts. Here we show that HMGA2 interacts with pRB and induces E2F1 activity in mouse pituitary adenomas by displacing HDAC1 from the pRB/E2F1 complex-a process that results in E2F1 acetylation. We found that loss of E2F1 function (obtained by mating HMGA2 and E2F1(-/-) mice) suppressed pituitary tumorigenesis in HMGA2 mice. Thus, HMGA2-mediated E2F1 activation is a crucial event in the onset of these tumors in transgenic mice and probably also in human prolactinomas.


Subject(s)
E2F1 Transcription Factor/physiology , HMGA2 Protein/physiology , Pituitary Neoplasms/metabolism , Acetylation , Animals , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic , DNA/metabolism , E2F1 Transcription Factor/genetics , Enzyme Activation , HMGA2 Protein/biosynthesis , HMGA2 Protein/genetics , Histone Deacetylase 1 , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Pituitary Neoplasms/pathology , Promoter Regions, Genetic , Protein Binding , Response Elements , Retinoblastoma Protein/metabolism , Signal Transduction
16.
Cancer Res ; 66(5): 2536-43, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16510570

ABSTRACT

The HMGA1 protein is a major factor in chromatin architecture and gene control. It plays a critical role in neoplastic transformation. In fact, blockage of HMGA1 synthesis prevents rat thyroid cell transformation by murine transforming retroviruses, and an adenovirus carrying the HMGA1 gene in the antisense orientation induces apoptotic cell death in anaplastic human thyroid carcinoma cell lines, but not in normal thyroid cells. Moreover, both in vitro and in vivo studies have established the oncogenic role of the HMGA1 gene. In this study, to define HMGA1 function in vivo, we examined the consequences of disrupting the Hmga1 gene in mice. Both heterozygous and homozygous mice for the Hmga1-null allele show cardiac hypertrophy due to the direct role of HMGA1 on cardiomyocytic cell growth regulation. These mice also developed hematologic malignancies, including B cell lymphoma and myeloid granuloerythroblastic leukemia. The B cell expansion and the increased expression of the RAG1/2 endonuclease, observed in HMGA1-knockout spleen tissues, might be responsible for the high rate of abnormal IgH rearrangements observed in these neoplasias. Therefore, the data reported here indicate the critical role of HMGA1 in heart development and growth, and reveal an unsuspected antioncogenic potential for this gene in hematologic malignancies.


Subject(s)
Cardiomegaly/genetics , HMGA1a Protein/genetics , Lymphoproliferative Disorders/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cardiomegaly/immunology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Female , Gene Expression Regulation, Neoplastic , HMGA1a Protein/deficiency , HMGA1a Protein/immunology , Haploidy , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Interleukins/biosynthesis , Interleukins/genetics , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Lymphoproliferative Disorders/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Wistar
17.
Oncogene ; 24(21): 3427-35, 2005 May 12.
Article in English | MEDLINE | ID: mdl-15735694

ABSTRACT

Overexpression of HMGA1 proteins is a constant feature of human carcinomas. Moreover, rearrangements of this gene have been detected in several human benign tumors of mesenchymal origin. To define the role of these proteins in cell transformation in vivo, we have generated transgenic mice overexpressing ubiquitously the HMGA1 gene. These mice developed mixed growth hormone/prolactin cell pituitary adenomas and natural killer (NK)-T/NK cell lymphomas. The HMGA1-induced expression of IL-2 and IL-15 proteins and their receptors may account for the onset of these lymphomas. At odds with mice overexpressing a wild-type or a truncated HMGA2 protein, adrenal medullar hyperplasia and pancreatic islet cell hyperplasia frequently occurred and no increase in body size and weight was observed in HMGA1 mice. Taken together, these data indicate an oncogenic role of the HMGA1 gene also in vivo.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Profiling , HMGA1a Protein/biosynthesis , HMGA1a Protein/genetics , Lymphoma/genetics , Pituitary Neoplasms/genetics , Prolactinoma/genetics , Adrenal Medulla/pathology , Animals , Human Growth Hormone/metabolism , Hyperplasia , Immunohistochemistry , Islets of Langerhans/pathology , Killer Cells, Natural/immunology , Lymphoma/physiopathology , Lymphoma/veterinary , Mice , Mice, Transgenic , Pituitary Neoplasms/physiopathology , Pituitary Neoplasms/veterinary , Prolactinoma/physiopathology , Prolactinoma/veterinary , Reverse Transcriptase Polymerase Chain Reaction
18.
Biochem J ; 389(Pt 1): 91-7, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15713121

ABSTRACT

HMGA1 (high-mobility-group A1) proteins are architectural transcription factors that are found overexpressed in embryogenesis and malignant tumours. We have shown previously that they have a role in lymphopoiesis, since the loss of HMGA1 expression leads to an impairment of T-cell development and to an increase in B-cell population. Since RAGs (recombination activating genes) are key regulators of lymphoid differentiation, in the present study we investigate whether RAG2 expression is dependent on HMGA1 activity. We show that RAG2 gene expression is up-regulated in Hmga1-/- ES (embryonic stem) cells and EBs (embryoid bodies) as well as in yolk sacs and fibroblasts from Hmga1-/- mice, suggesting that HMGA1 proteins control RAG2 gene expression both in vitro and in vivo. We show that the effect of HMGA1 on RAG2 expression is direct, identify the responsible region in the RAG2 promoter and demonstrate binding to the promoter in vivo using chromatin immunoprecipitation. Since RAG2 is necessary for lymphoid cell development, our results suggest a novel mechanism by which HMGA1 might regulate lymphoid differentiation.


Subject(s)
DNA-Binding Proteins/genetics , Down-Regulation , HMGA Proteins/metabolism , Animals , Binding Sites , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Humans , Mice , Mice, Knockout , Promoter Regions, Genetic/genetics , Protein Binding , Stem Cells/metabolism , Up-Regulation , Yolk Sac/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...