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










Publication year range
1.
Oncogene ; 35(35): 4663-74, 2016 09 01.
Article in English | MEDLINE | ID: mdl-26876207

ABSTRACT

Targeted therapies using small-molecule inhibitors (SMIs) are commonly used in metastatic renal cell cancer (mRCC) patients; patients often develop drug resistance and eventually succumb to disease. Currently, understanding of mechanisms leading to SMIs resistance and any identifiable predictive marker(s) are still lacking. We discovered that DAB2IP, a novel Ras-GTPase-activating protein, was frequently epigenetically silenced in RCC, and DAB2IP loss was correlated with the overall survival of RCC patients. Loss of DAB2IP in RCC cells enhances their sensitivities to growth factor stimulation and resistances to SMI (such as mammalian target of rapamycin (mTOR) inhibitors). Mechanistically, loss of DAB2IP results in the activation of extracellular signal-regulated kinase/RSK1 and phosphoinositide-3 kinase/mTOR pathway, which synergizes the induction of hypoxia-inducible factor (HIF)-2α expression. Consequently, elevated HIF-2α suppresses p21/WAF1 expression that is associated with resistance to mTOR inhibitors. Thus combinatorial targeting both pathways resulted in a synergistic tumor inhibition. DAB2IP appears to be a new prognostic/predictive marker for mRCC patients, and its function provides a new insight into the molecular mechanisms of drug resistance to mTOR inhibitors, which also can be used to develop new strategies to overcome drug-resistant mRCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , TOR Serine-Threonine Kinases/genetics , ras GTPase-Activating Proteins/genetics , Adult , Aged , Animals , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Middle Aged , Molecular Targeted Therapy , Protein Kinase Inhibitors/administration & dosage , Signal Transduction/genetics , Sirolimus/administration & dosage , TOR Serine-Threonine Kinases/antagonists & inhibitors , ras GTPase-Activating Proteins/biosynthesis
3.
An Sist Sanit Navar ; 35(1): 9-18, 2012.
Article in Spanish | MEDLINE | ID: mdl-22552124

ABSTRACT

BACKGROUND: To study the biochemical progression-free survival (BPFS) achieved by a group of high risk patients in accordance with D'Amico's classification treated with radical prostatectomy. To identify the clinical-pathological variables which are influential in biochemical progression-free survival and, if possible, use them to design a prognostic model. MATERIAL AND METHODS: The study involves 232 patients, out of a series of 1,054, diagnosed with clinically localized prostate cancer, qualified as high risk on D'Amico's classification (PSA>20 ng/ml or Gleason score 8-10 or T3) treated with radical prostatectomy. The BPFS is studied and the clinical-pathological variables obtained (PSA, Gleason score of the biopsy and of the piece, clinical and pathological study, unilateral or bilateral affectation, margins of the prostatectomy piece, Ki-67 expression) are analyzed to identify whether they influenced the BPFS. Contingency tables and tables for survival analysis: Kaplan-Meyer, log-rank and Cox models were used for the statistical study. RESULTS: Descriptive study: PSA: 23.3 ng/ml (median); cGleason 2-6: 33%; 7: 13%; 8-10: 54%; T2: 58%; Bilateral affectation in the diagnostic biopsy: 59%; RNM T2: 60%; RNM T3: 40%. pGleason 2-6: 24%; 7: 28%; 8-10: 48%; pT2: 43%; pT3a: 30%; pT3b: 27%; Affected margin: 51%; N1:13%. Progression-free survival: with a mean and median follow-up of 64 months; 53% show biochemical progression. The median until progression: 42 months. Progression-free survival at 5 and 10 years is 43±3% and 26±7%. The multivariate study (Cox models) shows that the variables that are independently influential in the BPFS are the affectation of margins (HR: 3.5; 95% IC.1.9-6.7; p<0001); and Ki67 >10% (HR: 2.3; 95% IC: 1.2-4.3; P: 0.009). Risk groups: using the two influential variables and employing Cox models, three risk groups emerged as the best model: Group 1 (0 variables present); Group 2 (1 variable); Group 3 (2 variables). The progression-free survival is 69±8%; 27±6% and 18±11% at 5 years. The differences amongst the three groups are significant. CONCLUSION: The high risk group according to the D'Amico classification is heterogeneous in relation to biochemical progression and can be broken down into three risk groups using the two independently influential variables (affected margins and Ki67 percentage).


Subject(s)
Adenocarcinoma/surgery , Prostatectomy , Prostatic Neoplasms/surgery , Disease-Free Survival , Humans , Male , Prognosis , Risk Factors
4.
Oncogene ; 30(18): 2147-60, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21217781

ABSTRACT

As the ultimate electron acceptor in oxidative phosphorylation, oxygen plays a critical role in metabolism. When oxygen levels drop, heterodimeric hypoxia-inducible factor (Hif) transcription factors become active and facilitate adaptation to hypoxia. Hif regulation by oxygen requires the protein von Hippel-Lindau (pVhl) and pVhl disruption results in constitutive Hif activation. The liver is a critical organ for metabolic homeostasis, and Vhl inactivation in hepatocytes results in a Hif-dependent shortening in life span. While albumin-Cre;Vhl(F/F) mice develop hepatic steatosis and impaired fatty acid oxidation, the variable penetrance and unpredictable life expectancy has made the cause of death elusive. Using a system in which Vhl is acutely disrupted and a combination of ex vivo liver perfusion studies and in vivo oxygen measurements, we demonstrate that Vhl is essential for mitochondrial respiration in vivo. Adenovirus-Cre mediated acute Vhl disruption in the liver caused death within days. Deprived of pVhl, livers accumulated tryglicerides and circulating ketone and glucose levels dropped. The phenotype was reminiscent of inborn defects in fatty acid oxidation and of fasted PPARα-deficient mice and while death was unaffected by pharmacologic PPARα activation, it was delayed by glucose administration. Ex vivo liver perfusion analyses and acylcarnitine profiles showed mitochondrial impairment and a profound inhibition of liver ketone and glucose production. By contrast, other mitochondrial functions, such as ureagenesis, were unaffected. Oxygen consumption studies revealed a marked suppression of mitochondrial respiration, which, as determined by magnetic resonance oximetry in live mice, was accompanied by a corresponding increase in liver pO(2). Importantly, simultaneous inactivation of Hif-1ß suppressed liver steatosis and rescued the mice from death. These data demonstrate that constitutive Hif activation in mice is sufficient to suppress mitochondrial respiration in vivo and that no other pathway exists in the liver that can allow oxygen utilization when Hif is active precluding thereby metabolic collapse.


Subject(s)
Hypoglycemia/pathology , Hypoxia/metabolism , Ketones/blood , Liver/metabolism , Oxygen/metabolism , Signal Transduction , Animals , Gluconeogenesis , Hypoglycemia/metabolism , Mice , Reverse Transcriptase Polymerase Chain Reaction , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/physiology
6.
Kidney Int ; 56(5): 1691-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10571777

ABSTRACT

BACKGROUND: Diabetic nephropathy is characterized by glomerular hypertrophy. We have recently shown that experimental diabetes mellitus is associated with an increase in glomerular expression of the cyclin kinase inhibitor p21WAF1/CIP1 (p21). Furthermore, in vitro glucose-induced mesangial cell hypertrophy is also associated with an up-regulated expression of p21. In this study, we tested the hypothesis that p21 mediates diabetic glomerular hypertrophy in vivo. METHODS: Experimental diabetes mellitus was induced by streptozotocin in mice in which p21 was genetically deleted (p21 -/-) and in wild-type mice (p21 +/+). Kidney biopsies were obtained from diabetic and control (citrate injected) p21 +/+ and p21 -/- mice at day 60. The tissue was used for morphologic studies of glomerular size (measured by computer image-analysis system), glomerular cellularity (cell count), glomerular matrix expansion (silver stain), apoptosis (TUNEL), and expression of transforming growth factor-beta1 (TGF-beta1) by in situ hybridization. RESULTS: The glomerular tuft area increased 11.21% in diabetic p21 +/+ mice at day 60 compared with control (3329.98 +/- 244.05 micrometer(2) vs. 2994. 39 +/- 176.22 micrometer(2), P = 0.03), and the glomerular cell count did not change in diabetic p21 +/+ mice at day 60 compared with the control. These findings are consistent with glomerular hypertrophy. In contrast, the glomerular tuft area did not increase in diabetic p21 -/- mice at day 60 compared with the control (3544.15 +/- 826.49 vs. 3449.15 +/- 109.65, P = 0.82), nor was there an increase in glomerular cell count (41.41 +/- 13.18 vs. 46.95 +/- 3.00, P = 0.43). Diabetic p21 +/+ mice, but not p21 -/- mice, developed an increase in proteinuria at day 60 compared with the control. Tubular cell proliferation, measured by proliferating cell nuclear antigen immunostaining, was increased in both diabetic p21 +/+ (2.1-fold) and p21 -/- (7.61-fold) mice compared with controls. Glomerular cell apoptosis did not increase in diabetic mice. Although glomerular TGF-beta1 mRNA levels increased in both strains of diabetic mice at day 60, the glomerular matrix did not expand. CONCLUSIONS: Hyperglycemia was associated with glomerular hypertrophy in p21 +/+ mice. Despite the increase in TGF-beta1 mRNA, diabetic p21 -/- mice did not develop glomerular hypertrophy, providing evidence that the cyclin kinase inhibitor p21 may be required for diabetic glomerular hypertrophy induced by TGF-beta1. The loss of p21 increases tubular but not glomerular cell proliferation in diabetic nephropathy. The absence of glomerular hypertrophy appears protective of renal function in diabetic mice.


Subject(s)
Cyclins/physiology , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/pathology , Kidney Glomerulus/pathology , Animals , Blood Glucose/analysis , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA/biosynthesis , Hypertrophy , Kidney Tubules/metabolism , Mice , Mice, Knockout , Proteinuria/etiology , RNA, Messenger/analysis , Streptozocin , Transforming Growth Factor beta/genetics
7.
Kidney Int ; 55(6): 2349-61, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10354282

ABSTRACT

BACKGROUND: During glomerulogenesis, visceral glomerular epithelial cells (VECs) exit the cell cycle and become terminally differentiated and quiescent. In contrast to other resident glomerular cells, VECs undergo little if any proliferation in response to injury. However, the mechanisms for this remain unclear. Cell proliferation is controlled by cell-cycle regulatory proteins where the cyclin-dependent kinase inhibitor p21Cip1,WAF1 (p21) inhibits cell proliferation and is required for differentiation of many nonrenal cell types. METHODS: To test the hypothesis that p21 is required to maintain a differentiated and quiescent VEC phenotype, experimental glomerulonephritis was induced in p21 knockout (-/-) and p21 wild-type (+/+) mice with antiglomerular antibody. DNA synthesis (proliferating cell nuclear antigen, bromodeoxyuridine staining), VEC proliferation (multilayers of cells in Bowman's space), matrix accumulation (periodic acid-Schiff, silver staining), apoptosis (TUNEL), and renal function (serum urea nitrogen) were studied on days 5 and 14 (N = 6 per time point). VECs were identified by location, morphology, ezrin staining, and electron microscopy. VEC differentiation was measured by staining for Wilms' tumor-1 gene. RESULTS: Kidneys from unmanipulated p21-/- mice were histologically normal and did not have increased DNA synthesis, suggesting that p21 was not required for the induction of VEC terminal differentiation. Proliferating cell nuclear antigen and bromodeoxyuridine staining was increased 4.3- and 3.3-fold, respectively, in p21-/- mice with glomerulonephritis (P < 0.0001 vs. p21+/+ mice). At each time point, VEC proliferation was also increased in nephritic p21-/- mice (P < 0.0001 vs. p21+/+ mice). VEC re-entry into the cell cycle was associated with the loss of Wilms' tumor-1 gene staining. Nephritic p21-/- mice had increased extracellular matrix protein accumulation and apoptosis and decreased renal function (serum urea nitrogen) compared with p21+/+ mice (P < 0.001). CONCLUSION: These results show that the cyclin kinase inhibitor p21 is not required by VECs to attain a terminally differentiated VEC phenotype. However, the loss of p21, in disease states, is associated with VEC re-entry into the cell cycle and the development of a dedifferentiated proliferative phenotype.


Subject(s)
Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/physiology , Glomerulonephritis/pathology , Glomerulonephritis/physiopathology , Animals , Apoptosis , Cell Division , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA/biosynthesis , Enzyme Inhibitors/metabolism , Epithelium/pathology , Genes, Wilms Tumor , Glomerulonephritis/genetics , Kidney Glomerulus/pathology , Kidney Glomerulus/physiopathology , Mice , Mice, Knockout
8.
Proc Natl Acad Sci U S A ; 96(3): 1002-7, 1999 Feb 02.
Article in English | MEDLINE | ID: mdl-9927683

ABSTRACT

In mammalian cells, activation of certain checkpoint pathways as a result of exposure to genotoxic agents results in cell cycle arrest. The integrity of these arrest pathways is critical to the ability of the cell to repair mutations that otherwise might compromise viability or contribute to deregulation of cellular growth and proliferation. Here we examine the mechanism through which DNA damaging agents result in a G1 arrest that depends on the tumor suppressor p53 and its transcriptional target p21. By using primary cell lines lacking specific cell cycle regulators, we demonstrate that this pathway functions through the growth suppressive properties of the retinoblastoma protein (pRB) tumor suppressor. Specifically, gamma-irradiation inhibits the phosphorylation of pRB at cyclin-dependent kinase 2-specific, but not cyclin-dependent kinase 4-specific, sites in a p21-dependent manner. Most importantly, we show that pRB is a critical component of this DNA damage checkpoint. These data indicate that the p53 --> p21 checkpoint pathway uses the normal cell cycle regulatory machinery to induce the accumulation of the growth suppressive form of pRB and suggest that loss of pRB during the course of tumorigenesis disrupts the function of an important DNA damage checkpoint.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle/radiation effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Retinoblastoma Protein/metabolism , Animals , Cell Cycle/physiology , Cells, Cultured , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , DNA Damage , Embryo, Mammalian , Enzyme Inhibitors/metabolism , Fibroblasts/physiology , Fibroblasts/radiation effects , G1 Phase , Gamma Rays , Mice , Peptide Mapping , Phosphopeptides/chemistry , Phosphopeptides/isolation & purification , Phosphorylation , Retinoblastoma Protein/radiation effects
9.
Mol Cell Biol ; 18(7): 4385-90, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9632822

ABSTRACT

Disruption of the mouse Atm gene, whose human counterpart is consistently mutated in ataxia-telangiectasia (A-T) patients, creates an A-T mouse model exhibiting most of the A-T-related systematic and cellular defects. While ATM plays a major role in signaling the p53 response to DNA strand break damage, Atm-/- p53(-/-) mice develop lymphomas earlier than Atm-/- or p53(-/-) mice, indicating that mutations in these two genes lead to synergy in tumorigenesis. The cell cycle G1/S checkpoint is abolished in Atm-/- p53(-/-) mouse embryonic fibroblasts (MEFs) following gamma-irradiation, suggesting that the partial G1 cell cycle arrest in Atm-/- cells following gamma-irradiation is due to the residual p53 response in these cells. In addition, the Atm-/- p21(-/-) MEFs are more severely defective in their cell cycle G1 arrest following gamma-irradiation than Atm-/- and p21(-/-) MEFs. The Atm-/- MEFs exhibit multiple cellular proliferative defects in culture, and an increased constitutive level of p21 in these cells might account for these cellular proliferation defects. Consistent with this notion, Atm-/- p21(-/-) MEFs proliferate similarly to wild-type MEFs and exhibit no premature senescence. These cellular proliferative defects are also rescued in Atm-/- p53(-/-) MEFs and little p21 can be detected in these cells, indicating that the abnormal p21 protein level in Atm-/- cells is also p53 dependent and leads to the cellular proliferative defects in these cells. However, the p21 mRNA level in Atm-/- MEFs is lower than that in Atm+/+ MEFs, suggesting that the higher level of constitutive p21 protein in Atm-/- MEFs is likely due to increased stability of the p21 protein.


Subject(s)
Cyclins/physiology , Protein Serine-Threonine Kinases , Proteins/physiology , Tumor Suppressor Protein p53/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , Carcinogenicity Tests , Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA-Binding Proteins , G1 Phase , Gamma Rays , Humans , Lymphoma/etiology , Mice , Phenotype , Proteins/genetics , RNA, Messenger , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins
10.
J Cell Biol ; 141(2): 503-14, 1998 Apr 20.
Article in English | MEDLINE | ID: mdl-9548727

ABSTRACT

Proliferation in mammalian cells is controlled primarily in the G1-phase of the cell cycle through the action of the G1 cyclin-dependent kinases, CDK4 and CDK2. To explore the mechanism of cellular response to extrinsic factors, specific loss of function mutations were generated in two negative regulators of G1 progression, p21 and pRB. Individually, these mutations were shown to have significant effects in G1 regulation, and when combined, Rb and p21 mutations caused more profound defects in G1. Moreover, cells deficient for pRB and p21 were uniquely capable of anchorage-independent growth. In contrast, combined absence of pRB and p21 function was not sufficient to overcome contact inhibition of growth nor for tumor formation in nude mice. Finally, animals with the genotype Rb+/-;p21(-/-) succumbed to tumors more rapidly than Rb+/- mice, suggesting that in certain contexts mutations in these two cell cycle regulators can cooperate in tumor development.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins , Cyclins/physiology , Fibroblasts/cytology , G1 Phase/genetics , Proto-Oncogene Proteins , Retinoblastoma Protein/physiology , Tumor Suppressor Proteins , Animals , Blood , Cell Adhesion , Cell Communication , Cell Cycle/physiology , Cell Division , Cell Size , Cell Transformation, Neoplastic , Cells, Cultured , Cyclin D1/analysis , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclins/genetics , Enzyme Inhibitors , Mice , Mice, Knockout , Mice, Nude , Microtubule-Associated Proteins/analysis , Mutation , Neoplasms, Experimental/etiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Retinoblastoma Protein/genetics , Signal Transduction/physiology
13.
EMBO J ; 15(14): 3693-701, 1996 Jul 15.
Article in English | MEDLINE | ID: mdl-8758936

ABSTRACT

The p53 tumor suppressor limits cellular proliferation by inducing either G1 arrest or apoptosis, depending on the cellular context. To determine if these pathways are mechanistically distinct, we have examined the effects of different p53 mutants in p53 null primary mouse embryo fibroblasts. We chose this system as it is highly physiological and ensures that the interpretation of the results will not be confounded by the presence of endogenous p53 or oncoproteins which target p53. Using single cell microinjection assays for both G1 arrest and apoptosis, with loss-of-function and chimeric gain-of-function mutants, we have demonstrated that transcriptional activation is critical for both processes. Replacement of the p53 activation domain with that of VP16, or replacement of the p53 oligomerization domain with that of GCN4, reconstituted both G1 arrest and apoptosis activities. However, despite the importance of transcriptional activation in both processes, the target gene requirements are different. The p21 cyclin-dependent kinase inhibitor, which has been shown to be a direct target of p53 and a component of the radiation-induced G1 arrest response, is dispensable for oncogene-induced apoptosis, suggesting that these two p53-dependent transcriptional pathways are distinct.


Subject(s)
Apoptosis , Cyclins/physiology , Oncogenes , Transcriptional Activation , Tumor Suppressor Protein p53/physiology , Adenovirus E1A Proteins/biosynthesis , Animals , Apoptosis/genetics , Apoptosis/physiology , Binding Sites , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , G1 Phase , Mice , Mutagenesis , Signal Transduction , Tumor Suppressor Protein p53/genetics
14.
Nature ; 377(6549): 552-7, 1995 Oct 12.
Article in English | MEDLINE | ID: mdl-7566157

ABSTRACT

The protein p21 is a dual inhibitor of cyclin-dependent kinases and proliferating-cell nuclear antigen (PCNA), both of which are required for passage through the cell cycle. The p21 gene is under the transcriptional control of p53 (ref. 5), suggesting that p21 might promote p53-dependent cell cycle arrest or apoptosis. p21 has also been implicated in cell senescence and in cell-cycle withdrawal upon terminal differentiation. Here we investigate the role of p21 in these processes using chimaeric mice composed partly of p21-/- and partly of p21+/+ cells. Immunohistochemical studies of the p21+/+ and p21-/- components of adult small intestine indicated that deletion of p21 has no detectable effect on the migration-associated differentiation of the four principal intestinal epithelial cell lineages or on p53-dependent apoptosis following irradiation. However, p21-/- mouse embryo fibroblasts are impaired in their ability to undergo G1 arrest following DNA damage.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle/physiology , Cyclins/physiology , Animals , Apoptosis/physiology , Cell Cycle/radiation effects , Cell Differentiation/physiology , Cell Division/physiology , Cell Line , Cells, Cultured , Chimera , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Cyclins/deficiency , Cyclins/genetics , Enzyme Inhibitors , Gamma Rays , Gene Targeting , Intestine, Small/cytology , Intestine, Small/embryology , Mice , Mice, Inbred C57BL , Proliferating Cell Nuclear Antigen , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...