Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
2.
Sci Rep ; 10(1): 7057, 2020 04 27.
Article in English | MEDLINE | ID: mdl-32341372

ABSTRACT

We have recently identified and characterized two pseudogenes (HMGA1P6 and HMGA1P7) of the HMGA1 gene, which has a critical role in malignant cell transformation and cancer progression. HMGA1P6 and HMGAP17 act as microRNA decoy for HMGA1 and other cancer-related genes upregulating their protein levels. We have previously shown that they are upregulated in several human carcinomas, and their expression positively correlates with a poor prognosis and an advanced cancer stage. To evaluate in vivo oncogenic activity of HMGA1 pseudogenes, we have generated a HMGA1P7 transgenic mouse line overexpressing this pseudogene. By a mean age of 12 months, about 50% of the transgenic mice developed splenomegaly and accumulation of lymphoid cells in several body compartments. For these mice FACS and immunohistochemical analyses suggested the diagnosis of B-cell lymphoma that was further supported by clonality analyses and RNA expression profile of the pathological tissues of the HMGA1P7 transgenic tissues. Therefore, these results clearly demonstrate the oncogenic activity of HMGA1 pseudogenes in vivo.


Subject(s)
HMGA1a Protein/metabolism , Lymphoma, B-Cell/metabolism , Animals , Flow Cytometry , HMGA1a Protein/genetics , Immunohistochemistry , Lymphocytes/metabolism , Lymphoma, B-Cell/genetics , Mice , Mice, Transgenic , NIH 3T3 Cells , Pseudogenes/genetics , RNA-Seq
3.
Genes (Basel) ; 10(5)2019 05 15.
Article in English | MEDLINE | ID: mdl-31096664

ABSTRACT

Endometrioid endometrial carcinomas (EEC) are the most common malignant gynecologic tumors. Despite the increase in EEC molecular knowledge, the identification of new biomarkers involved in disease's development and/or progression would represent an improvement in its course. High-mobility group A protein (HMGA) family members are frequently overexpressed in a wide range of malignancies, correlating with a poor prognosis. Thus, the aim of this study was to analyze HMGA1 and HMGA2 expression pattern and their potential role as EEC biomarkers. HMGA1 and HMGA2 expression was initially evaluated in a series of 46 EEC tumors (stages IA to IV), and the findings were then validated in The Cancer Genome Atlas (TCGA) EEC cohort, comprising 381 EEC tumors (stages IA to IV). Our results reveal that HMGA1 and HMGA2 mRNA and protein are overexpressed in ECC, but only HMGA1 expression is associated with increased histological grade and tumor size. Moreover, HMGA1 but not HMGA2 overexpression was identified as a negative prognostic factor to EEC patients. Finally, a positive correlation between expression of HMGA1 pseudogenes-HMGA1-P6 and HMGA1-P7-and HMGA1 itself was detected, suggesting HMGA1 pseudogenes may play a role in HMGA1 expression regulation in EEC. Thus, these results indicate that HMGA1 overexpression possesses a potential role as a prognostic biomarker for EEC.


Subject(s)
Carcinoma, Endometrioid/genetics , Endometrial Neoplasms/genetics , HMGA1a Protein/genetics , HMGA2 Protein/genetics , Adult , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Carcinoma, Endometrioid/metabolism , Endometrial Neoplasms/metabolism , Female , HMGA1a Protein/biosynthesis , HMGA2 Protein/biosynthesis , Humans , Middle Aged , Prognosis , Transcriptome
4.
Mol Cancer Ther ; 18(2): 482-493, 2019 02.
Article in English | MEDLINE | ID: mdl-30425132

ABSTRACT

The CDK inhibitor, p27kip1, encoded by the Cdkn1b gene can negatively modulate cell proliferation. The control of p27 activity during the cell cycle is regulated at multiple levels, including transcription, translation, and protein stability. The last residue of p27 (threonine 198 in human, threonine 197 in mouse) is involved in the control of protein stability. We have generated a murine knock-in model (Cdkn1b T197A) in which threonine 197 is replaced by alanine, which renders p27 protein highly unstable due to a high rate of proteasomal degradation. Expectedly, Cdkn1b T197A/T197A mice present with increased body size and weight, organomegaly, and multiple organ hyperplasia, similar to what is observed in Cdkn1b KO/KO mice. We investigated the effects exerted by the restoration of normal levels of p27 protein in the tissue of Cdkn1b T197A/T197A mice. We found that proteasome inhibition with bortezomib rescues the hyperplasia induced by the lack of p27 expression in Cdkn1b T197A/T197A but not in Cdkn1b KO/KO mice. However, BAY 11-7082, a proteasome inhibitor that stabilizes IκB but not p27, fails to rescue hyperplasia in Cdkn1b T197A/T197A mice. Bortezomib increases p27 half-life and reduces the proliferation in MEFs derived from Cdkn1b T197A/T197A but not from Cdkn1b WT/WT mice, whereas BAY 11-7082 had no effect on the protein levels of p27 and on the proliferation rate of Cdkn1b T197A/T197A MEFs.The results presented here demonstrate that Cdkn1b T197A/T197A mice represent an attractive in vivo model to investigate whether the targeting of p27 degradation machinery might prove beneficial in the treatment of a variety of human proliferative disorders caused by increased turnover of p27 protein.


Subject(s)
Amino Acid Substitution , Bortezomib/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/chemistry , Cyclin-Dependent Kinase Inhibitor p27/genetics , Models, Animal , Animals , Gene Knock-In Techniques , Hyperplasia , Mice , Nitriles/pharmacology , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis , Sulfones/pharmacology
6.
BMC Cancer ; 17(1): 170, 2017 03 04.
Article in English | MEDLINE | ID: mdl-28259135

ABSTRACT

BACKGROUND: Loss of CBX7 expression has been described in several malignant neoplasias, including human colon and thyroid carcinomas proposing CBX7 as a tumor suppressor gene with a key role in cancer progression. This role is supported from the development of benign and malignant neoplasias in Cbx7 null mice. The aim of our work has been to investigate the mechanisms underlying the CBX7 oncosuppressor activity by analyzing the microRNAs (miRNAs) regulated by CBX7. METHODS: The miRNA expression profiles of the mouse embryonic fibroblasts (MEFs) null for Cbx7 and the wild-type counterpart were analyzed by the miRNACHIP microarray and then validated by qRT-PCR. To asses KRAS as target of miR-155 we evaluated the protein levels after transfection of the synthetic miR-155. Human colon carcinoma samples have been investigated for the expression of CBX7 and miR-155. RESULTS: Twenty miRNAs were found upregulated and nine, including miR-155, downregulated in cbx7-null MEFS in comparison with the wild-type ones. Then, we focused on miR-155 since several studies have shown its deregulated expression in several human malignancies and, moreover, was the most downregulated miRNA. Subsequently, we searched for miR-155 target genes demonstrating that KRAS protein levels are directly modulated by miR-155. A direct significant correlation (r = 0.6779) between CBX7 and miR-155 expression levels was found in a set of human colon carcinoma tissue samples. CONCLUSION: miR-155 is positively regulated by CBX7 in MEFs and colon carcinomas, and has KRAS as one of the target genes likely accounting for the anti-apoptotic activity ascribed to miR-155 in some tissue contexts.


Subject(s)
Colonic Neoplasms/metabolism , Fibroblasts/metabolism , Genes, ras , MicroRNAs/metabolism , Polycomb Repressive Complex 1/metabolism , Signal Transduction , Animals , Cell Line , Colonic Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation , Humans , Mice
7.
Sci Rep ; 6: 37622, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27874091

ABSTRACT

Recent studies have revealed that pseudogene transcripts can function as competing endogenous RNAs, and thereby can also contribute to cancer when dysregulated. We have recently identified two pseudogenes, HMGA1P6 and HMGA1P7 for the HMGA1 gene whose overexpression has a critical role in cancer progression. These pseudogenes work as competitive endogenous RNA decoys for HMGA1 and other cancer related genes suggesting their role in carcinogenesis. Looking for new HMGA1 pseudogene ceRNAs, we performed RNA sequencing technology on mouse embryonic fibroblasts deriving from transgenic mice overexpressing HMGA1P7. Here, we report that HMGA1P7 mRNA sustains the H19 and Igf2 overexpression by acting as miRNA decoy. Lastly, the expression of HMGA1P7 was significantly correlated with H19 and IGF2 levels in human breast cancer thereby suggesting a role for HMGA1P7 deregulation in this neoplasia.


Subject(s)
Insulin-Like Growth Factor II/genetics , Pseudogenes/genetics , RNA, Long Noncoding/genetics , Animals , Breast Neoplasms/genetics , Embryo, Mammalian/metabolism , Female , Fibroblasts/metabolism , Humans , Insulin-Like Growth Factor II/metabolism , MCF-7 Cells , Mice , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , NIH 3T3 Cells , RNA, Long Noncoding/metabolism , Reproducibility of Results , Sequence Analysis, RNA , Up-Regulation
8.
Oncotarget ; 7(19): 28724-35, 2016 May 10.
Article in English | MEDLINE | ID: mdl-26895108

ABSTRACT

Pseudogenes are DNA sequences with high homology to the corresponding functional gene, but, because of the accumulation of various mutations, they have lost their initial functions to code for proteins. Consequently, pseudogenes have been considered until few years ago dysfunctional relatives of the corresponding ancestral genes, and then useless in the course of genome evolution. However, several studies have recently established that pseudogenes are owners of key biological functions. Indeed, some pseudogenes control the expression of functional genes by competitively binding to the miRNAs, some of them generate small interference RNAs to negatively modulate the expression of functional genes, and some of them even encode functional mutated proteins. Here, we concentrate our attention on the pseudogenes of the HMGA1 gene, that codes for the HMGA1a and HMGA1b proteins having a critical role in development and cancer progression. In this review, we analyze the family of HMGA1 pseudogenes through three aspects: classification, characterization, and their possible function and involvement in cancer.


Subject(s)
HMGA1a Protein/genetics , HMGA1b Protein/genetics , Neoplasms/genetics , Pseudogenes/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , HMGA1a Protein/metabolism , HMGA1b Protein/metabolism , Humans , Models, Genetic , Mutation , Neoplasms/metabolism , Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
Am J Cancer Res ; 5(5): 1594-601, 2015.
Article in English | MEDLINE | ID: mdl-26175930

ABSTRACT

CBX7 is a polycomb protein that participates in the formation of polycomb repressive complex 1. Apart from few exceptions, CBX7 expression is lost in human malignant neoplasias and a clear correlation between its downregulated expression and a cancer aggressiveness and poor prognosis has been observed. These findings indicate a critical role of CBX7 in cancer progression. Consistently, CBX7 is able to differentially regulate crucial genes involved in cancer progression and in epithelial-mesenchymal transition, as osteopontin and E-cadherin. Recent evidences indicate a role of CBX7 also in the modulation of response to therapy. In conclusion, CBX7 represents an important prognostic factor, whose loss of expression in general indicates a bad prognosis and a progression towards a fully malignant phenotype.

10.
Naunyn Schmiedebergs Arch Pharmacol ; 388(11): 1179-86, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26216446

ABSTRACT

Lung cancer is one of the most common causes of cancer-related death worldwide in men and women, and, despite the recent remarkable scientific advances, drug treatment is still unsatisfactory. Polycomb protein chromobox homolog 7 (CBX7) is involved in several biological processes, including development and cancer progression, indeed the lack of CBX7 protein correlates with a highly malignant phenotype and a poor prognosis. However, its role in lung cancer still remains unknown. Since CBX7 is drastically downregulated in human lung carcinomas, we investigated whether restoration of CBX7 expression could affect growth property of lung cancer cells and modulate their sensitivity to treatment with irinotecan and etoposide, two chemoterapy drugs most commonly used in lung cancer therapy. Here, we demonstrate that restoration of CBX7 in two human lung carcinoma cell lines (A549 and H1299), in which this protein is not detectable, leads to a decreased proliferation (at least in part through a downregulation of phosphorylated ERK and phosphorylated p38) and an increased apoptotic cell death after drug exposure (at least in part through the downregulation of Bcl-2, phosphorylated Akt, and phosphorylated JNK). Taken together, these results suggest that the retention of CBX7 expression may play a role in the modulation of chemosensitivity of lung cancer patients to the treatment with irinotecan and etoposide.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Camptothecin/analogs & derivatives , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic , Polycomb Repressive Complex 1/genetics , Apoptosis/drug effects , Camptothecin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Resistance, Neoplasm , Humans , Irinotecan , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Polycomb Repressive Complex 1/metabolism , RNA, Messenger/metabolism
11.
Oncotarget ; 6(5): 2680-92, 2015 Feb 20.
Article in English | MEDLINE | ID: mdl-25595895

ABSTRACT

Several recent studies have reported the Polycomb Repressive Complex 1 member CBX7 as a tumor-suppressor gene whose expression progressively decreases in different human carcinomas in relation with tumor grade, malignant stage and poor prognosis. We have previously demonstrated that CBX7 is able to inhibit the expression of the SPP1 gene, encoding the chemokine osteopontin that is over-expressed in cancer and has a critical role in cancer progression. Here, we have analyzed the mechanism by which CBX7 regulates the SPP1 gene expression. We show that the SPP1 transcriptional regulation mechanism involves the CBX7-interacting protein HMGA1b, that acts as a positive regulator of the SPP1 gene. In fact, we demonstrate that, in contrast with the transcriptional activity of CBX7, HMGA1b is able to increase the SPP1 expression by inducing the activity of its promoter. Moreover, we show that CBX7 interferes with HMGA1b on the SPP1 promoter and counteracts the positive transcriptional activity of HMGA1b on the SPP1 expression. Furthermore, since we found that also the NF-κB complex resulted involved in the modulation of the SPP1 expression in thyroid cells, we suppose that CBX7/HMGA1b/NF-κB could take part in the same transcriptional mechanism that finally leads to the regulation of the SPP1 gene expression. Taken together, our data show the important role played by CBX7 in the negative regulation of the SPP1 gene expression, thus contributing to prevent the acquisition of a malignant phenotype.


Subject(s)
Carcinoma/metabolism , HMGA1b Protein/metabolism , Osteopontin/metabolism , Polycomb Repressive Complex 1/metabolism , Thyroid Neoplasms/metabolism , Binding Sites , Carcinoma/genetics , Carcinoma/pathology , Carcinoma, Papillary , Cell Line, Tumor , Cell Movement , Gene Expression Regulation , HEK293 Cells , HMGA1b Protein/genetics , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Osteopontin/genetics , Polycomb Repressive Complex 1/genetics , Promoter Regions, Genetic , Signal Transduction , Thyroid Cancer, Papillary , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Transcription, Genetic , Transfection
12.
Cell Cycle ; 13(23): 3636-9, 2014.
Article in English | MEDLINE | ID: mdl-25483074

ABSTRACT

Two pseudogenes for HMGA1, whose overexpression has a critical role in cancer progression, have been identified. They act as decoy for miRNAs that are able to target the HMGA1 gene then enhancing cell proliferation and migration. Moreover, these pseudogenes contain sequences that are potential target sites for cancer-related miRNAs. Interestingly, HMGA1 pseudogenes are highly expressed in human anaplastic thyroid carcinomas, that is one of the most aggressive tumor in mankind, but almost undetectable in well differentiated thyroid carcinomas.


Subject(s)
Disease Progression , Gene Expression Regulation, Neoplastic , HMGA Proteins/biosynthesis , Neoplasms/metabolism , Pseudogenes/physiology , Animals , Cell Proliferation/physiology , HMGA Proteins/genetics , Humans , MicroRNAs/biosynthesis , Neoplasms/genetics , Neoplasms/pathology
13.
Oncotarget ; 5(18): 8341-54, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25268743

ABSTRACT

The High Mobility Group A (HMGA) are nuclear proteins that participate in the organization of nucleoprotein complexes involved in chromatin structure, replication and gene transcription. HMGA overexpression is a feature of human cancer and plays a causal role in cell transformation. Since non-coding RNAs and pseudogenes are now recognized to be important in physiology and disease, we investigated HMGA1 pseudogenes in cancer settings using bioinformatics analysis. Here we report the identification and characterization of two HMGA1 non-coding pseudogenes, HMGA1P6 and HMGA1P7. We show that their overexpression increases the levels of HMGA1 and other cancer-related proteins by inhibiting the suppression of their synthesis mediated by microRNAs. Consistently, embryonic fibroblasts from HMGA1P7-overexpressing transgenic mice displayed a higher growth rate and reduced susceptibility to senescence. Moreover, HMGA1P6 and HMGA1P7 were overexpressed in human anaplastic thyroid carcinomas, which are highly aggressive, but not in differentiated papillary carcinomas, which are less aggressive. Lastly, the expression of the HMGA1 pseudogenes was significantly correlated with HMGA1 protein levels thereby implicating HMGA1P overexpression in cancer progression. In conclusion, HMGA1P6 and HMGA1P7 are potential proto-oncogenic competitive endogenous RNAs.


Subject(s)
HMGA Proteins/genetics , MicroRNAs/genetics , Ovarian Neoplasms/genetics , Pseudogenes/genetics , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Neoplasms/genetics , Animals , Apoptosis , Cell Proliferation , Cellular Senescence , Computational Biology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HEK293 Cells , HMGA Proteins/metabolism , Humans , MCF-7 Cells , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , RNA Interference , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Time Factors , Transfection
14.
Biol Open ; 3(9): 871-9, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25190058

ABSTRACT

We have recently generated knockout mice for the Cbx7 gene, coding for a polycomb group protein that is downregulated in human malignant neoplasias. These mice develop liver and lung adenomas and carcinomas, which confirms a tumour suppressor role for CBX7. The CBX7 ability to downregulate CCNE1 expression likely accounts for the phenotype of the Cbx7-null mice. Unexpectedly, Cbx7-knockout mice had a higher fat tissue mass than wild-type, suggesting a role of CBX7 in adipogenesis. Consistently, we demonstrate that Cbx7-null mouse embryonic fibroblasts go towards adipocyte differentiation more efficiently than their wild-type counterparts, and this effect is Cbx7 dose-dependent. Similar results were obtained when Cbx7-null embryonic stem cells were induced to differentiate into adipocytes. Conversely, mouse embryonic fibroblasts and human adipose-derived stem cells overexpressing CBX7 show an opposite behaviour. These findings support a negative role of CBX7 in the control of adipocyte cell growth and differentiation.

15.
PLoS One ; 9(5): e98295, 2014.
Article in English | MEDLINE | ID: mdl-24865347

ABSTRACT

BACKGROUND: We have previously shown that the expression of CBX7 is drastically decreased in several human carcinomas and that its expression progressively decreases with the appearance of a highly malignant phenotype. The aim of our study has been to investigate the mechanism by which the loss of CBX7 expression may contribute to the emergence of a more malignant phenotype. METHODS: We analyzed the gene expression profile of a thyroid carcinoma cell line after the restoration of CBX7 and, then, analyzed the transcriptional regulation of identified genes. Finally, we evaluated the expression of CBX7 and regulated genes in a panel of thyroid and lung carcinomas. RESULTS: We found that CBX7 negatively or positively regulates the expression of several genes (such as SPP1, SPINK1, STEAP1, and FOS, FOSB, EGR1, respectively) associated to cancer progression, by interacting with their promoter regions and modulating their transcriptional activity. Quantitative RT-PCR analyses in human thyroid and lung carcinoma tissues revealed a negative correlation between CBX7 and its down-regulated genes, while a positive correlation was observed with up-regulated genes. CONCLUSION: In conclusion, the loss of CBX7 expression might play a critical role in advanced stages of carcinogenesis by deregulating the expression of specific effector genes.


Subject(s)
Genes, Neoplasm , Lung Neoplasms/genetics , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Thyroid Neoplasms/genetics , Animals , Cell Line, Tumor , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , HEK293 Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Rats , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
16.
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.

17.
Cell Cycle ; 11(10): 1888-91, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22544325

ABSTRACT

The generation of knockout mice for the Cbx7 gene validates the tumor suppressor role of CBX7, whose expression is lost in several human malignancies. Indeed, these mice developed liver and lung adenomas and carcinomas. Cyclin E overexpression due to the lack of Cbx7 negative regulation of its expression likely accounts for the phenotype of the Cbx7-KO mice. A similar mechanism is likely involved in human lung carcinogenesis, since cyclin E upregulation associated with the loss of CBX7 expression has been observed in most of the human lung carcinomas analyzed.


Subject(s)
Carcinoma/metabolism , Cell Transformation, Neoplastic , Lung Neoplasms/metabolism , Repressor Proteins/metabolism , Animals , Carcinoma/pathology , Cell Line , Cyclin E/genetics , Cyclin E/metabolism , HMGA Proteins/metabolism , Humans , Lung Neoplasms/pathology , Mice , Mice, Knockout , Polycomb Repressive Complex 1 , Promoter Regions, Genetic , Repressor Proteins/deficiency , Repressor Proteins/genetics , Up-Regulation
18.
J Clin Invest ; 122(2): 612-23, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22214847

ABSTRACT

The CBX7 gene encodes a polycomb group protein that is known to be downregulated in many types of human cancers, although the role of this protein in carcinogenesis remains unclear. To shed light on this issue, we generated mice null for Cbx7. Mouse embryonic fibroblasts derived from these mice had a higher growth rate and reduced susceptibility to senescence compared with their WT counterparts. This was associated with upregulated expression of multiple cell cycle components, including cyclin E, which is known to play a key role in lung carcinogenesis in humans. Adult Cbx7-KO mice developed liver and lung adenomas and carcinomas. In in vivo and in vitro experiments, we demonstrated that CBX7 bound to the CCNE1 promoter in a complex that included HDAC2 and negatively regulated CCNE1 expression. Finally, we found that the lack of CBX7 protein expression in human lung carcinomas correlated with CCNE1 overexpression. These data suggest that CBX7 is a tumor suppressor and that its loss plays a key role in the pathogenesis of cancer.


Subject(s)
Genes, Tumor Suppressor , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , Cyclin E/genetics , Cyclin E/metabolism , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Knockout , Polycomb Repressive Complex 1 , Promoter Regions, Genetic , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics
19.
Genes Cancer ; 1(3): 210-24, 2010 Mar.
Article in English | MEDLINE | ID: mdl-21779448

ABSTRACT

High mobility group A (HMGA) overexpression plays a critical role in neoplastic transformation. To investigate whether HMGA acts by regulating the expression of microRNAs, we analyzed the microRNA expression profile of human breast adenocarcinoma cells (MCF7) transfected with the HMGA1 gene, which results in a highly malignant phenotype. Among the microRNAs induced by HMGA1, we focused on miR-181b, which was overexpressed in several malignant neoplasias including breast carcinomas. We show that miR-181b regulates CBX7 protein levels, which are down-regulated in cancer, and promotes cell cycle progression. We also demonstrate that CBX7, being negatively regulated by HMGA, is able to negatively regulate miR-181b expression. Finally, there was a direct correlation between HMGA1 and miR-181b expression and an inverse correlation between HMGA1 and CBX7 expression in human breast carcinomas. These data indicate the presence of a novel pathway involving HMGA1, miR-181b, and CBX7, which leads to breast cancer progression.

20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...