Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Cell Res ; 1870(5): 119465, 2023 06.
Article in English | MEDLINE | ID: mdl-36935052

ABSTRACT

HIPK2 is an evolutionary conserved serine/threonine kinase with multifunctional roles in stress response, embryonic development and pathological conditions, such as cancer and fibrosis. The heterogeneity of its interactors and targets makes HIPK2 activity strongly dependent on the cellular context, and allows it to modulate multiple signaling pathways, ultimately regulating cell fate and proliferation. HIPK2 is highly expressed in the central and peripheral nervous systems, and its genetic ablation causes neurological defects in mice. Moreover, HIPK2 is involved in processes, such as endoplasmic reticulum stress response and protein aggregate accumulation, and pathways, including TGF-ß and BMP signaling, that are crucial in the pathogenesis of neurological disorders. Here, we review the data about the role of HIPK2 in neuronal development, survival, and homeostasis, highlighting the implications in the pathogenesis of neurological disorders, and pointing out HIPK2 potentiality as therapeutic target and diagnostic or prognostic marker.


Subject(s)
Carrier Proteins , Nervous System Diseases , Animals , Carrier Proteins/metabolism , Nervous System Diseases/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Humans
2.
Cell Death Dis ; 6: e2004, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26633710

ABSTRACT

Homeodomain-interacting protein kinase 2 (HIPK2) is a multitalented coregulator of an increasing number of transcription factors and cofactors involved in cell death and proliferation in several organs and systems. As Hipk2(-/-) mice show behavioral abnormalities consistent with cerebellar dysfunction, we investigated whether Hipk2 is involved in these neurological symptoms. To this aim, we characterized the postnatal developmental expression profile of Hipk2 in the brain cortex, hippocampus, striatum, and cerebellum of mice by real-time PCR, western blot analysis, and immunohistochemistry. Notably, we found that whereas in the brain cortex, hippocampus, and striatum, HIPK2 expression progressively decreased with age, that is, from postnatal day 1 to adulthood, it increased in the cerebellum. Interestingly, mice lacking Hipk2 displayed atrophic lobules and a visibly smaller cerebellum than did wild-type mice. More important, the cerebellum of Hipk2(-/-) mice showed a strong reduction in cerebellar Purkinje neurons during adulthood. Such reduction is due to the activation of an apoptotic process associated with a compromised proteasomal function followed by an unpredicted accumulation of ubiquitinated proteins. In particular, Purkinje cell dysfunction was characterized by a strong accumulation of ubiquitinated ß-catenin. Moreover, our behavioral tests showed that Hipk2(-/-) mice displayed muscle and balance impairment, indicative of Hipk2 involvement in cerebellar function. Taken together, these results indicate that Hipk2 exerts a relevant role in the survival of cerebellar Purkinje cells and that Hipk2 genetic ablation generates cerebellar dysfunction compatible with an ataxic-like phenotype.


Subject(s)
Protein Serine-Threonine Kinases/deficiency , Purkinje Cells/physiology , Animals , Apoptosis/physiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Humans , Mice , Mice, Knockout , Phenotype , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Purkinje Cells/cytology , Purkinje Cells/metabolism , beta Catenin/metabolism
3.
Transl Med UniSa ; 13: 47-58, 2015 Dec.
Article in English | MEDLINE | ID: mdl-27042433

ABSTRACT

There is increasing evidence that diet plays a crucial role in age-related diseases and cancer. Oxidative stress is a conceivable link between diet and diseases, thus food antioxidants, counteracting the damage caused by oxidation, are potential tools for fight age-related diseases and cancer. Resveratrol (RSV), a polyphenolic antioxidant from grapes, has gained enormous attention particularly because of its ability to induce growth arrest and apoptosis in cancer cells, and it has been proposed as both chemopreventive and therapeutic agent for cancer and other diseases. Even though the effects of RSV have been studied in prostate cancer cells and animal models, little is known about its effects on normal cells and tissues. To address this issue, we have investigated the effects of RSV on EPN cells, a human non-transformed prostate cell line, focusing on the relationship between RSV and p66Shc, a redox enzyme whose activities strikingly intersect those of RSV. p66Shc activity is regulated by phosphorylation of serine 36 (Ser36) and has been related to mitochondrial oxidative stress, apoptosis induction, regulation of cell proliferation and migration. Here we show that RSV inhibits adhesion, proliferation and migration of EPN cells, and that these effects are associated to induction of dose- and time-dependent p66Shc-Ser36 phosphorylation and ERK1/2 de-phosphorylation. Moreover, we found that RSV is able to activate also p52Shc, another member of the Shc protein family. These data show that RSV affects non-transformed prostate epithelial cells and suggest that Shc proteins may be key contributors of RSV effects on prostate cells.

4.
Cell Death Dis ; 4: e963, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24336083

ABSTRACT

PATZ1 is a transcriptional factor functioning either as an activator or a repressor of gene transcription depending upon the cellular context. It appears to have a dual oncogenic/anti-oncogenic activity. Indeed, it is overexpressed in colon carcinomas, and its silencing inhibits colon cancer cell proliferation or increases sensitivity to apoptotic stimuli of glioma cells, suggesting an oncogenic role. Conversely, the development of B-cell lymphomas, sarcomas, hepatocellular carcinomas and lung adenomas in Patz1-knockout (ko) mice supports its tumour suppressor function. PATZ1 role in mouse lymphomagenesis is mainly because of the involvement of PATZ1 in BCL6-negative autoregulation. However, this does not exclude that PATZ1 may be involved in tumorigenesis by other mechanisms. Here, we report that PATZ1 interacts with the tumour suppressor p53 and binds p53-dependent gene promoters, including those of BAX, CDKN1A and MDM2. Knockdown of PATZ1 in HEK293 cells reduces promoter activity of these genes and inhibits their expression, suggesting a role of PATZ in enhancing p53 transcriptional activity. Consistently, Patz1-ko mouse embryonic fibroblasts (MEFs) show decreased expression of Bax, Cdkn1a and Mdm2 compared with wild-type (wt) MEFs. Moreover, Patz1-ko MEFs show a decreased percentage of apoptotic cells, either spontaneous or induced by treatment with 5-fluorouracil (5FU), compared with wt controls, suggesting a pro-apoptotic role for PATZ1 in these cells. However, PATZ1 binds p53-target genes also independently from p53, exerting, in the absence of p53, an opposite function on their expression. Indeed, knockdown of PATZ1 in p53-null osteosarcoma cells upregulates BAX expression and decreases survival of 5FU-treated cells, then suggesting an anti-apoptotic role of PATZ1 in p53-null cancer cells. Therefore, these data support a PATZ1 tumour-suppressive function based on its ability to enhance p53-dependent transcription and apoptosis. Conversely, its opposite and anti-apoptotic role in p53-null cancer cells provides the perspective of PATZ1 silencing as a possible adjuvant in the treatment of p53-null cancer.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Line , Cell Survival/genetics , Cell Survival/physiology , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Protein Binding , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Repressor Proteins/genetics , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
5.
Cell Death Dis ; 4: e729, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23868062

ABSTRACT

The transcription factor Pax8, a member of the Paired-box gene family, is a critical regulator required for proper development and differentiation of thyroid follicular cells. Despite being Pax8 well characterized with respect to its role in regulating genes responsible for thyroid differentiation, its involvement in cell survival and proliferation has been hypothesized but remains unclear. Here, we show that Pax8 overexpression significantly increases proliferation and colony-forming efficiency of Fischer rat thyroid line 5 epithelial cells, although it is not sufficient to overcome their hormone dependence. More interestingly, we show that Pax8-specific silencing induces apoptosis through a p53-dependent pathway that involves caspase-3 activation and cleavage of poly(ADP)ribose polymerase. Our data indicate that tumor protein 53 induced nuclear protein 1 (tp53inp1), a positive regulator of p53-dependent cell cycle arrest and apoptosis, is a transcriptional target of Pax8 and is upregulated by Pax8 knockdown. Remarkably, tp53inp1 silencing significantly abolishes Pax8-induced apoptosis thus suggesting that tp53inp1 may be the mediator of the observed effects. In conclusion, our data highlight that Pax8 is required for the survival of differentiated epithelial cells and its expression levels are able to modulate the proliferation rate of such cells.


Subject(s)
Cell Proliferation , Cell Survival , Epithelial Cells/physiology , Paired Box Transcription Factors/physiology , Animals , Apoptosis , Apoptosis Regulatory Proteins , Cell Cycle , Cell Line , Gene Knockdown Techniques , Heat-Shock Proteins/metabolism , Mice , Nuclear Proteins/metabolism , PAX8 Transcription Factor , RNA Interference , Rats
6.
Cell Death Dis ; 3: e383, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22932725

ABSTRACT

The high-mobility group A (HMGA) proteins are a family of non-histone chromatin factors, encoded by the HMGA1 and HMGA2 genes. Several studies demonstrate that HMGA proteins have a critical role in neoplastic transformation, and their overexpression is mainly associated with a highly malignant phenotype, also representing a poor prognostic index. Even though a cytoplasmic localization of these proteins has been previously reported in some highly malignant neoplasias, a clear role for this localization has not been defined. Here, we first confirm the localization of the HMGA1 proteins in the cytoplasm of cancer cells, and then we report a novel mechanism through which HMGA1 inhibits p53-mitochondrial apoptosis by counteracting the binding of p53 to the anti-apoptotic factor Bcl-2. Indeed, we demonstrate a physical and functional interaction between HMGA1 and Bcl-2 proteins. This interaction occurs at mitochondria interfering with the ability of p53 protein to bind Bcl-2, thus counteracting p53-mediated mitochondrial apoptosis. This effect is associated with the inhibition of cytochrome c release and activation of caspases. Consistent with this mechanism, a strong correlation between HMGA1 cytoplasmic localization and a more aggressive histotype of thyroid, breast and colon carcinomas has been observed. Therefore, cytoplasmic localization of HMGA1 proteins in malignant tissues is a novel mechanism of inactivation of p53 apoptotic function.


Subject(s)
Apoptosis/drug effects , HMGA1a Protein/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/pharmacology , Caspases/metabolism , Cell Line , Cytochromes c/metabolism , HEK293 Cells , HMGA1a Protein/genetics , HeLa Cells , Humans , MCF-7 Cells , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays
7.
J Biomed Biotechnol ; 2012: 201689, 2012.
Article in English | MEDLINE | ID: mdl-22665981

ABSTRACT

Orthodontics is a branch of dentistry that aims at the resolution of dental malocclusions. The specialist carries out the treatment using intraoral or extraoral orthodontic appliances that require forces of a given load level to obtain a tooth movement in a certain direction in dental arches. Orthodontic tooth movement is dependent on efficient remodeling of periodontal ligament and alveolar bone, correlated with several biological and mechanical responses of the tissues surrounding the teeth. A periodontal ligament placed under pressure will result in bone resorption whereas a periodontal ligament under tension results in bone formation. In the primary stage of the application of orthodontic forces, an acute inflammation occurs in periodontium. Several proinflammatory cytokines are produced by immune-competent cells migrating by means of dilated capillaries. In this paper we summarize, also through the utilization of animal models, the role of some of these molecules, namely, interleukin-1ß and vascular endothelial growth factor, that are some proliferation markers of osteoclasts and osteoblasts, and the macrophage colony stimulating factor.


Subject(s)
Cytokines/biosynthesis , Disease Models, Animal , Malocclusion/therapy , Tooth Movement Techniques , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Malocclusion/metabolism , Malocclusion/pathology , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Rats
8.
Anticancer Res ; 31(12): 4211-8, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22199283

ABSTRACT

BACKGROUND: N-Cadherin (CDH2) is a calcium-dependent adhesion protein, whose de novo expression, re-expression, up-regulation and down-regulation in human tumors has been demonstrated. The aim of the present work was to define the prognostic role of N-Cadherin in a large series of oral squamous cell carcinomas (OSCCs). MATERIALS AND METHODS: A total of 94 selected OSCCs were quantitatively and qualitatively analyzed by immunohistochemistry for N-Cadherin. The association between protein expression and clinico-pathological parameters was assessed by statistical analysis. RESULTS: In neoplastic tissue, N-Cadherin levels were more evident than in normal peritumoral epithelium (p<0.05). Protein staining was mainly detected in the neoplastic cells, and only focal nuclear positivity was observed. Expression of cytoplasmic N-Cadherin correlated significantly with poor histological differentiation (p<0.05). Furthermore, we have observed significant a statistical trend for stage and a correlation with worst patient outcome, also confirmed by Kaplan-Meier estimates. CONCLUSION: Our work has underlined the key-role of N-Cadherin in oral carcinogenesis and in the prognostic stratification of patients.


Subject(s)
Cadherins/biosynthesis , Carcinoma, Squamous Cell/metabolism , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/metabolism , Adult , Aged , Carcinoma, Squamous Cell/diagnosis , Disease Progression , Disease-Free Survival , Female , Humans , Immunohistochemistry/methods , Male , Medical Oncology/methods , Middle Aged , Mouth Neoplasms/diagnosis , Prognosis
9.
J Cell Physiol ; 226(11): 2997-3003, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21302296

ABSTRACT

Steroid hormones are expressed at low levels in mesenchymal cells and are highly expressed in soft tissue sarcoma. In human soft tissue fibrosarcoma cell line (HT-1080), the epidermal growth factor (EGF) stimulates the express of matrix metal (MMPs) expression through a Src-dependent mechanism. In human fibrosarcomas, increased expression of MMPs correlates with the metastatic progression. Our recent data in human breast cancer cell line MCF-7, demonstrates that EGF stimulates estradiol receptor (ER) phosphorylation on tyrosine at position 537 thereby promoting the association of a complex among EGF receptor (EGFR), androgen receptor (AR), ER, and Src that activates EGF-dependent signaling pathway. In the present study, we demonstrate that, in HT-1080 cells, the Src kinase activity is involved in EGFR phosphorylation and this activity is regulated by an interplay between Src, steroid receptors, and EGFR. In these cells, estradiol (E(2) )/ER and synthetic androgen (R1881)/AR trans-activate EGFR leading to the downstream signaling and to ERK activation. Indeed, the association between ER/AR and EGFR enhances metastatic progression of fibrosarcoma tumors. A population pilot study performed on 16 patients with soft tissue neoplasias highlights that MMPs expression correlates with progression of anaplastic sarcoma as well as overexpression of EGFR. These findings suggest that there is a crosstalk among AR, ER, and EGFR that lead to src activation also in fibrosarcoma cells.


Subject(s)
Receptors, Steroid/metabolism , Sarcoma/pathology , Soft Tissue Neoplasms/pathology , src-Family Kinases/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Child , Child, Preschool , Disease Progression , ErbB Receptors/metabolism , Estradiol/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Male , Metribolone/pharmacology , Middle Aged , Pilot Projects , Signal Transduction , Young Adult
10.
Oncogene ; 29(30): 4341-51, 2010 Jul 29.
Article in English | MEDLINE | ID: mdl-20498639

ABSTRACT

RET/papillary thyroid carcinoma 1 (PTC1) oncogene is frequently activated in human PTCs. It is characterized by the fusion of the intracellular kinase-encoding domain of RET to the first 101 amino acids of CCDC6. The aim of our work is to characterize the function of the CCDC6 protein to better understand the function of its truncation, that results in the loss of the expression of one allele, in the process of thyroid carcinogenesis. Here, we report that CCDC6 interacts with CREB1 and represses its transcriptional activity by recruiting histone deacetylase 1 and protein phosphatase 1 proteins at the CRE site of the CREB1 target genes. Finally, we show an increased CREB1 phosphorylation and activity in PTCs carrying the RET/PTC1 oncogene. Consistently, an increased expression of two known CREB1 target genes, AREG and cyclin A, was observed in this subgroup of thyroid papillary carcinomas. Therefore, the repression of CREB1 activity by CCDC6 has a critical function in the development of human thyroid papillary carcinomas carrying RET/PTC1 activation.


Subject(s)
Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cytoskeletal Proteins/physiology , Histone Deacetylase 1/metabolism , Protein Phosphatase 1/metabolism , Repressor Proteins/physiology , Thyroid Neoplasms/etiology , Amphiregulin , Cell Line, Tumor , Cyclic AMP Response Element-Binding Protein/genetics , EGF Family of Proteins , Glycoproteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Oncogene Proteins, Fusion/genetics , Phosphorylation , Promoter Regions, Genetic , Protein-Tyrosine Kinases/genetics , Transcription, Genetic
11.
J Pathol ; 214(1): 58-64, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17935122

ABSTRACT

The high-mobility group A (HMGA) non-histone chromosomal proteins HMGA1 and HMGA2 are architectural factors. They are abundantly expressed during embryogenesis and in most malignant neoplasias, whereas their expression is low or absent in normal adult tissues. Their over-expression is known to have a causal role in cellular neoplastic transformation. Previous studies from our group have shown that their expression is restricted to specific germinal cells. In this study we have evaluated, by immunohistochemistry, the expression of HMGA1 and HMGA2 in a series of post-pubertal testicular tumours of different histological types, including 30 seminomas, 15 teratomas, 15 embryonal carcinomas and 10 mixed germinal tumours with a prominent yolk sac tumour component. HMGA1 protein expression was detected in all seminomas and embryonal carcinomas analysed, but not in teratomas or yolk sac carcinomas. Conversely, HMGA2 was present only in embryonal carcinomas and yolk sac carcinomas, but not in seminomas or teratomas. The immunohistochemical data were further confirmed by Western blot and, at the mRNA level, by RT-PCR analyses. These findings indicate that HMGA1 and HMGA2 are differently expressed with respect to the state of differentiation of testicular germ cell tumours (TGCTs), with over-expression of both proteins in pluripotential embryonal carcinoma cells and loss of expression of HMGA1 in yolk sac tumours and of both proteins in the mature adult tissue of teratoma areas. Therefore, the different profiles of HMGA1 and HMGA2 protein expression could represent a valuable diagnostic tool in some cases in which the histological differential diagnosis is problematic.


Subject(s)
Biomarkers, Tumor/metabolism , HMGA1a Protein/metabolism , HMGA2 Protein/metabolism , Neoplasms, Germ Cell and Embryonal/diagnosis , Testicular Neoplasms/diagnosis , Adult , Biomarkers, Tumor/genetics , Blotting, Western , Gene Expression , HMGA1a Protein/genetics , HMGA2 Protein/genetics , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Testicular Neoplasms/pathology
12.
Cell Death Differ ; 13(9): 1554-63, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16341121

ABSTRACT

HMGA gene overexpression and rearrangements are frequent in several tumours, but their oncogenic function is still unclear. Here we report of a physical and functional interaction between High Mobility Group A1 (HMGA1) protein and p53 oncosuppressor. We found that HMGA1 binds p53 in vitro and in vivo, and both proteins are present in the same complexes bound to the Bax gene promoter. HMGA1 interferes with the p53-mediated transcription of p53 effectors Bax and p21(waf1) while cooperates with p53 in the transcriptional activation of the p53 inhibitor mdm2. This transcriptional modulation is associated with a reduced p53-dependent apoptosis in cells expressing exogenous HMGA1 and p53, or in cells expressing endogenously the proteins and in which p53 was activated by UV-irradiation. Furthermore, antisense inhibition of HMGA1b expression dramatically increases the UV-induced p53-mediated apoptosis. These data define a new physical and functional interaction between HMGA1 and p53 that modulates transcription of p53 target genes and inhibits apoptosis.


Subject(s)
Apoptosis , HMGA1b Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Humans , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-mdm2/metabolism , Transcription, Genetic , Transcriptional Activation , Ultraviolet Rays , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
13.
Oncogene ; 20(43): 6132-41, 2001 Sep 27.
Article in English | MEDLINE | ID: mdl-11593421

ABSTRACT

The HMGI proteins (HMGI, HMGY and HMGI-C) have an important role in the chromatin organization and interact with different transcriptional factors. The HMGI genes are expressed at very low levels in normal adult tissues, whereas they are very abundant during embryonic development and in several experimental and human tumours. In order to isolate proteins interacting with the HMGI(Y) proteins, a yeast two-hybrid screening was performed using the HMGI(Y) protein as bait. This analysis led to the isolation of homeodomain-interacting protein kinase-2 (HIPK2), a serine/threonine nuclear kinase. HIPK2 co-immunoprecipitates with the HMGI(Y) protein in 293T cells. The interaction between HIPK2 and HMGI(Y) occurs through the PEST domain of HIPK2 and it is direct because in vitro translated HIPK2 binds HMGI(Y). We also show that HIPK2 is able to phosphorylate the HMGI(Y) protein by an in vitro kinase assay. In order to understand a possible role of HIPK2 gene in cell growth we performed a colony assay which showed an impressive HIPK2 inhibitory effect on normal thyroid cells. Flow cytometric analysis would indicate the block of cell growth at the G2/M phase of the cell cycle. Since normal thyroid cells do not express detectable HMGI(Y) protein levels, we assume that the HIPK2 inhibitory effect is independent from the interaction with the HMGI(Y) protein.


Subject(s)
Carrier Proteins/metabolism , High Mobility Group Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Binding Sites , Bromodeoxyuridine/metabolism , Carrier Proteins/chemistry , Carrier Proteins/genetics , Catalysis , Cell Division/drug effects , Cell Line , Cell Nucleus/enzymology , DNA, Complementary/metabolism , Flow Cytometry , Gene Library , HMGA1a Protein , High Mobility Group Proteins/chemistry , High Mobility Group Proteins/genetics , Humans , Models, Genetic , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Protein Binding , Protein Biosynthesis , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transfection , Two-Hybrid System Techniques
14.
Am J Pathol ; 159(4): 1225-30, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11583949

ABSTRACT

The RING-finger protein RNF4 modulates both steroid-receptor-dependent and basal transcription and interacts with a variety of nuclear proteins involved in cell growth control. RNF4 is expressed at very high levels in testis and at much lower levels in several other tissues. We show that in germ cells RNF4 expression is strongly modulated during progression of spermatogonia to spermatids, with a peak in spermatocytes. Analysis of human testicular germ cell tumors shows that RNF4 is not expressed in all tumors analyzed including seminomas, the highly malignant embryonal carcinomas, yolk sac, and mixed germ cell tumors. We also show that the ectopically expressed RNF4 gene inhibits cell proliferation of both somatic and germ cell tumor-derived cells. Mutation of critical cysteine residues in the RING finger domain abolished the RNF4 growth inhibition activity. Our results suggest that the lack of RNF4 expression may play a role in the progression of testicular tumors.


Subject(s)
DNA-Binding Proteins/metabolism , Growth Inhibitors/metabolism , Nuclear Proteins , Spermatozoa/metabolism , Testicular Neoplasms/metabolism , Transcription Factors , Animals , Cell Division/drug effects , Cellular Senescence/physiology , DNA-Binding Proteins/pharmacology , Growth Inhibitors/pharmacology , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred Strains , Reference Values , Spermatozoa/cytology , Spermatozoa/physiology , Testis/metabolism , Tumor Cells, Cultured , Ubiquitin-Protein Ligases
15.
Cancer Res ; 61(11): 4583-90, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11389094

ABSTRACT

The high mobility group (HMG) proteins (HMGA1a, HMGA1b, and HMGA2) bind to DNA and interact with various transcriptional factors. Therefore, they play an important role in chromatin organization. HMGA protein expression is low in normal adult tissues, but abundant during embryonic development and in several experimental and human tumors. Blockage of HMGA expression inhibits the transformation of rat thyroid PC Cl 3 cells treated with oncogene-carrying retroviruses, thus implicating HMGA in rat thyroid transformation. To better understand the role of HMGA and to establish whether its up-regulated expression is sufficient to induce the transformed phenotype, we generated PC Cl 3 cells that overexpress the protein. We demonstrate that HMGA1b protein overexpression does not transform normal rat thyroid PC Cl 3 cells, but it deregulates their cell cycle: cells enter S-phase earlier and the G(2)-M transition is delayed. HMGA1-overexpressing cells undergo apoptosis through a pathway involving caspase-3 activation, probably consequent to the conflict between mitogenic pressure and the inability to proceed through the cell cycle. Using various HMGA1b gene mutations, we found that the third AT-hook domain and the acetylation site K60 are the protein regions required for induction of apoptosis in PC Cl 3 cells. In conclusion, although HMGA1 protein overexpression is associated with the malignant phenotype of rat and human thyroid cells, it does not transform normal thyroid cells in culture but leads them to programmed cell death.


Subject(s)
Apoptosis/physiology , High Mobility Group Proteins/biosynthesis , Thyroid Gland/cytology , Thyroid Gland/metabolism , Animals , CDC2 Protein Kinase/metabolism , Cell Cycle/physiology , Cell Division/physiology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cyclins/biosynthesis , DNA, Complementary/genetics , Gene Expression , High Mobility Group Proteins/genetics , Peptide Mapping , Protein Isoforms , Rats , Transfection
16.
Oncogene ; 20(5): 599-608, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11313992

ABSTRACT

The RET/PTC3 oncogene arises from the fusion between the N-terminal encoding domain of the RFG gene and the tyrosine kinase encoding domain of RET receptor. RET/PTC3 is very frequent in papillary thyroid carcinomas, especially in children exposed to the Chernobyl accident. We have studied the functional consequences of the RFG-RET fusion. Here we show that the N-terminal coiled-coil domain of RGF mediates oligomerization and activation of the kinase and of the transforming capability of RET/PTC3. In addition, the RFG coiled-coil domain mediates a physical association between RET/PTC3 and RGF proteins, rendering RFG a bona fide substrate of RET/PTC3 kinase. Finally, we show that the coiled-coil domain of RGF is essential for the distribution of the RET/PTC3 protein at the membrane/particulate cell compartment level, where also most of the RFG protein is localized. We propose that fusion to the RFG coiled-coil domain provides RET kinase with a scaffold that mediates oligomerization and re-localization of the RET/PTC3 protein, a process that may be crucial for the signalling of this specific RET/PTC variant.


Subject(s)
Drosophila Proteins , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , 3T3 Cells , Animals , Cell Line , Cell Membrane/enzymology , Cell Membrane/metabolism , Cell Transformation, Neoplastic , Enzyme Activation , Epithelial Cells/cytology , Humans , Mice , Oncogene Proteins, Fusion/genetics , Protein Structure, Tertiary , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , Rats , Receptor Protein-Tyrosine Kinases/genetics , Thyroid Gland/cytology , Transfection
17.
Mol Cell Biol ; 21(7): 2485-95, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259597

ABSTRACT

The high-mobility group I (HMGI) nonhistone chromosomal proteins HMGI(Y) and HMGI-C have been implicated in defining chromatin structure and in regulating the transcription of several genes. These proteins have been implicated in adipocyte homeostasis: a severe deficiency of fat tissue is found in mice with targeted disruption of the HMGI-C locus, and lipomagenesis in humans is frequently associated with somatic mutations of HMGI genes. The aim of this study was to examine the role of HMGI(Y) proteins in adipocytic cell growth and differentiation. First, we found that differentiation of the preadipocytic 3T3-L1 cell line caused early induction of HMGI(Y) gene expression. Suppression of HMGI(Y) expression by antisense technology dramatically increased the growth rate and impaired adipocytic differentiation in these cells. The process of adipogenic differentiation involves the interplay of several transcription factors, among which is the CCAAT/enhancer-binding protein (C/EBP) family of proteins. These factors are required for the transcriptional activation of adipocyte-specific genes. We also tested the hypothesis that HMGI(Y) might participate in transcriptional control of adipocyte-specific promoters. We found that HMGI(Y) proteins bind C/EBPbeta in vivo and in vitro. Furthermore, we show that HMGI(Y) strongly potentiates the capacity of C/EBPbeta to transactivate the leptin promoter, an adipose-specific promoter. Taken together, these results indicate that the HMGI(Y) proteins play a critical role in adipocytic cell growth and differentiation.


Subject(s)
Adipocytes/cytology , Adipocytes/physiology , High Mobility Group Proteins/physiology , Transcription Factors/physiology , 3T3 Cells , Animals , Cell Differentiation/physiology , Cell Division/physiology , HMGA1a Protein , Mice
18.
Oncogene ; 17(3): 377-85, 1998 Jul 23.
Article in English | MEDLINE | ID: mdl-9690519

ABSTRACT

We have recently reported that neoplastic transformation of two rat thyroid epithelial cell lines by retroviruses carrying the v-mos and v-ras Ki oncogenes is associated with a drastic increase of AP-1 activity. The most important effects were represented by the dramatic junB and fra-1 gene induction, which was abolished by the block of the transformation-induced HMGI-C protein synthesis. Here, we have further characterized the transformation-dependent AP-1 activity, by analysing the expression of different jun- and fos-related components, in rat thyroid cell lines transformed by several oncogenes, in human thyroid carcinoma cell lines, and in naturally occurring human thyroid tumours. A significant increase of Fra-1 and JunB protein levels was detected in all oncogene transformed rat thyroid cell lines. Fra-1 gene induction was demonstrated to occur also in human thyroid carcinoma cell lines and tissues. Conversely, c-Jun and JunD proteins, rather than JunB, accumulated in human thyroid carcinoma cell lines. An induction of AP-1 target genes was also detected both in rat and human thyroid transformed cell lines. Therefore, in vivo and in vitro thyroid cell transformation is associated with important compositional changes in the AP-1 complex and an increased transcriptional activity.


Subject(s)
Cell Transformation, Neoplastic , Oncogenes , Thyroid Gland/metabolism , Thyroid Neoplasms/genetics , Transcription Factor AP-1/metabolism , Animals , Epithelial Cells/metabolism , Epithelial Cells/pathology , Genes, jun , HMGA2 Protein , High Mobility Group Proteins/biosynthesis , Humans , Neoplasm Proteins/biosynthesis , Polymerase Chain Reaction , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-jun/biosynthesis , Rats , Thyroglobulin/biosynthesis , Thyroid Gland/pathology , Thyrotropin/pharmacology
19.
EMBO J ; 16(17): 5310-21, 1997 Sep 01.
Article in English | MEDLINE | ID: mdl-9311991

ABSTRACT

The expression of the high mobility group I (HMGI)-C chromatin component was shown previously to be essential for the establishment of the neoplastic phenotype in retrovirally transformed thyroid cell lines. To identify possible targets of the HMGI-C gene product, we have analyzed the AP-1 complex in normal, fully transformed and antisense HMGI-C-expressing rat thyroid cells. We show that neoplastic transformation is associated with a drastic increase in AP-1 activity, which reflects multiple compositional changes. The strongest effect is represented by the dramatic junB and fra-1 gene induction, which is prevented in cell lines expressing the antisense HMGI-C. These results indicate that the HMGI-C gene product is essential for the junB and fra-1 transcriptional induction associated with neoplastic transformation. The inhibition of Fra-1 protein synthesis by stable transfection with a fra-1 antisense RNA vector significantly reduces the malignant phenotype of the transformed thyroid cells, indicating a pivotal role for the fra-1 gene product in the process of cellular transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , High Mobility Group Proteins/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Thyroid Neoplasms/genetics , Animals , Gene Expression Regulation, Neoplastic , HMGA2 Protein , High Mobility Group Proteins/genetics , Protein Binding , RNA, Antisense , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Rats , Thyroid Gland/cytology , Transcription Factor AP-1/metabolism , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...