Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Androl ; 28(3): 453-60, 2007.
Article in English | MEDLINE | ID: mdl-17251596

ABSTRACT

Cytochrome P450 17alpha-hydroxylase/17, 20-lyase (CYP17) is crucial for cortisol and sex steroid biosynthesis. In a previous study we examined CYP17 function by generating mice with a targeted CYP17 deletion. We found that in addition to its role in steroid biosynthesis, CYP17 is present in germ cells. In the present study we examined the effect of CYP17 on sperm morphology. Disorganization of the sperm midpiece, small sperm mitochondria with reduced inner membranes and matrix, and irregular sperm shape were found to be associated with the CYP17 gene deletion. Treating the mice carrying the CYP17 deletion with testosterone did not alleviate the observed sperm phenotypes, suggesting that CYP17 acts in a testosterone-independent manner. These results suggest that CYP17, in addition to its role in androgen formation, is critical for proper mitochondrial architecture and sperm morphology and thus for sperm function and normal fertility.


Subject(s)
Androgens/physiology , Fertility/physiology , Mitochondria/ultrastructure , Spermatozoa/ultrastructure , Steroid 17-alpha-Hydroxylase/physiology , Animals , Male , Mice , Spermatozoa/cytology
2.
Cancer Res ; 66(20): 9937-47, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17047056

ABSTRACT

Tumors of the Ewing's sarcoma family (ESFT), such as Ewing's sarcoma (EWS) and primitive neuroectodermal tumors (PNET), are highly aggressive malignancies predominantly affecting children and young adults. ESFT express chimeric transcription factors encoded by hybrid genes fusing the EWS gene with several ETS genes, most commonly FLI-1. EWS/FLI-1 proteins are responsible for the malignant phenotype of ESFT, but only few of their transcriptional targets are known. Using antisense and short hairpin RNA-mediated gene expression knockdown, array analyses, chromatin immunoprecipitation methods, and reexpression studies, we show that caveolin-1 (CAV1) is a new direct target of EWS/FLI-1 that is overexpressed in ESFT cell lines and tumor specimens and is necessary for ESFT tumorigenesis. CAV1 knockdown led to up-regulation of Snail and the concomitant loss of E-cadherin expression. Consistently, loss of CAV1 expression inhibited the anchorage-independent growth of EWS cells and markedly reduced the growth of EWS cell-derived tumors in nude mice xenografts, indicating that CAV1 promotes the malignant phenotype in EWS carcinogenesis. Reexpression of CAV1 or E-cadherin in CAV1 knockdown EWS cells rescued the oncogenic phenotype of the original EWS cells, showing that the CAV1/Snail/E-cadherin pathway plays a central role in the expression of the oncogenic transformation functions of EWS/FLI-1. Overall, these data identify CAV1 as a key determinant of the tumorigenicity of ESFT and imply that targeting CAV1 may allow the development of new molecular therapeutic strategies for ESFT patients.


Subject(s)
Caveolin 1/physiology , Oncogene Proteins, Fusion/physiology , Proto-Oncogene Protein c-fli-1/physiology , Sarcoma, Ewing/pathology , Animals , Cadherins/biosynthesis , Cadherins/genetics , Caveolin 1/antagonists & inhibitors , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Adhesion/genetics , Cell Growth Processes/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Down-Regulation , Gene Expression Regulation, Neoplastic , HT29 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , RNA, Antisense/genetics , RNA-Binding Protein EWS , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/genetics , Up-Regulation
3.
Prostate ; 66(7): 708-17, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16425198

ABSTRACT

BACKGROUND: Mouse prostate cancer modeling presents unique obstacles to the study of spontaneous tumor initiation and progression due to the anatomical location of the tissue. RESULTS: High resolution (130 microm(x) x 130 microm(y) x 300 microm(z)), three-dimensional MRI allowed for the visualization, segmentation, and volumetric measurement of the prostate from normal and genetically engineered animals, in vivo. Additionally, MRS performed on the prostate epithelia of probasin-ErbB-2Delta x Pten(+/-) mice identified changes in the relative concentrations of the metabolites choline and citrate, which was not observed in TRAMP mice. METHODS: T1-weighted MRI was performed on normal, TRAMP, probasin-ErbB-2/Her2/Neu (probasin-ErbB-2Delta), and probasin-ErbB-2Delta in the context of decreased Pten activity (probasin-ErbB-2Delta x Pten(+/-)) mice. Volume-localized single-voxel proton magnetic resonance spectroscopy (SVS (1)H MRS) was also performed. CONCLUSIONS: The data presented supports the use of combined MRI and MRS for the measurement of biochemical and morphometric alterations in mouse models of prostate cancer.


Subject(s)
Disease Models, Animal , Prostatic Neoplasms/pathology , Prostatic Neoplasms/veterinary , Animals , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Mice
4.
Cancer Res ; 65(9): 3950-7, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15867396

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR) represents a ligand-dependent nuclear receptor family that regulates multiple metabolic processes associated with fatty acid beta-oxidation, glucose utilization, and cholesterol transport. These and other receptor-mediated actions pertain to their role in hypolipidemic and antidiabetic therapies and as potential targets for cancer chemopreventive agents. The present study evaluated the chemopreventive activity of two highly potent and selective PPARgamma and PPARdelta agonists in a progestin- and carcinogen-induced mouse mammary tumorigenesis model. Animals treated with the PPARgamma agonist GW7845 exhibited a moderate delay in tumor formation. In contrast, animals treated with the PPARdelta agonist GW501516 showed accelerated tumor formation. Significantly, tumors from GW7845-treated mice were predominantly ductal adenocarcinomas, whereas tumors from GW501516-treated animals were adenosquamous and squamous cell carcinomas. Gene expression analysis of tumors arising from GW7845- and GW501516-treated mice identified expression profiles that were distinct from each other and from untreated control tumors of the same histopathology. Only tumors from mice treated with the PPARgamma agonist expressed estrogen receptor-alpha in luminal transit cells, suggesting increased ductal progenitor cell expansion. Tumors from mice treated with the PPARdelta agonist exhibited increased PPARdelta levels and activated 3-phosphoinositide-dependent protein kinase-1 (PDK1), which co-associated, suggesting a link between the known oncogenic activity of PDK1 in mammary epithelium and PPARdelta activation. These results indicate that PPARdelta and PPARgamma agonists produce diverse, yet profound effects on mammary tumorigenesis that give rise to distinctive histopathologic patterns of tumor differentiation and tumor development.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carcinoma, Ductal/prevention & control , Mammary Neoplasms, Experimental/prevention & control , Oxazoles/pharmacology , PPAR delta/agonists , PPAR gamma/agonists , Thiazoles/pharmacology , Tyrosine/analogs & derivatives , Tyrosine/pharmacology , Animals , Carcinoma, Adenosquamous/chemically induced , Carcinoma, Adenosquamous/drug therapy , Carcinoma, Adenosquamous/pathology , Carcinoma, Adenosquamous/prevention & control , Carcinoma, Ductal/chemically induced , Carcinoma, Ductal/drug therapy , Carcinoma, Ductal/pathology , Carcinoma, Squamous Cell/chemically induced , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/prevention & control , Cell Differentiation/drug effects , Disease Progression , Female , Gene Expression Profiling , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Mice
5.
Cancer Res ; 64(20): 7220-5, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15492237

ABSTRACT

Oncogenic signals elevate expression of Id2 in multiple tumor types. When deregulated, Id2 inactivates the tumor suppressor proteins retinoblastoma, p107, and p130. Here, we report a novel and unexpected tumor inhibitory function of Id2 in the intestinal epithelium. First, genetic ablation of Id2 in the mouse prevents differentiation and cell cycle arrest of enterocytes at the time of formation of the crypt-villus unit. Later, these developmental abnormalities evolve toward neoplastic transformation with complete penetrance. Id2-null tumors contain severe dysplastic and metaplastic lesions and express aberrant amounts of beta-catenin. Thus, our data are the first to establish a direct requirement of basic helix-loop-helix inhibitors in driving differentiation and define an unexpected role for the retinoblastoma-binding protein Id2 in preventing tumor formation.


Subject(s)
DNA-Binding Proteins/physiology , Intestinal Mucosa/pathology , Intestinal Neoplasms/pathology , Repressor Proteins/physiology , Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Cycle/genetics , Cell Cycle/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Female , Inhibitor of Differentiation Protein 2 , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Male , Mice , Mice, Knockout , Mutation , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
6.
J Cell Sci ; 117(Pt 15): 3129-40, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15190119

ABSTRACT

Desmoplakin (DP) is a key component of cellular adhesion junctions known as desmosomes; however, recent investigations have revealed a novel location for DP in junctions separate from desmosomes termed complexus adherens junctions. These junctions are found at contact sites between endothelial cells that line capillaries. Few studies have focused on the function of DP in de novo capillary formation (vasculogenesis) and branching (angiogenesis) during tumorigenesis, embryonic development, cardiovascular development or wound healing. Only recently have investigations begun to determine the effect the loss of DP has on capillaries during embryogenesis (i.e. in DP-/- mice). Evidence shows that the loss of desmoplakin in vivo results in leaky capillaries and/or capillary malformation. Consequently, the goal of this study was to determine the function of DP in complexus adherens junctions during capillary formation. To accomplish this goal, we used siRNA technology to knock down desmoplakin expression in endothelial cells before they were induced to form microvascular tubes on matrigel. DP siRNA treated cells sent out filopodia and came in close contact with each other when plated onto matrigel; however, in most cases they failed to form tubes as compared with control endothelial cells. Interestingly, after siRNA degradation, endothelial cells were then capable of forming microvascular tubes. In depth analyses into the function of DP in capillary formation were not previously possible because the tools and experimental approaches only recently have become available (i.e. siRNA). Consequently, fully understanding the role of desmoplakin in capillary formation may lead to a novel approach for inhibiting vasculo- and angiogenesis in tumor formation.


Subject(s)
Cytoskeletal Proteins/physiology , Microcirculation , Alleles , Animals , Antigens, CD , Blotting, Western , Cadherins/metabolism , Capillaries/metabolism , Cell Adhesion , Cell Line , Collagen/pharmacology , Cytoskeletal Proteins/metabolism , Desmoplakins , Desmosomes/metabolism , Drug Combinations , Endothelium, Vascular/metabolism , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Laminin/pharmacology , Mice , Mice, Transgenic , Microscopy, Electron , Microscopy, Fluorescence , Neovascularization, Physiologic , Proteoglycans/pharmacology , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
7.
Cancer Res ; 63(19): 6290-8, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14559816

ABSTRACT

Exposure of normal mouse fibroblasts (MEF3T3) to ionizing radiation (IR) resulted in a dose-dependent increase of mTOR mRNA and protein levels and the shuttling of the mTOR protein from its normal, predominantly mitochondrial location to the cell nucleus. The same IR doses that activated mTOR induced the phosphorylation of p53 on Ser(18) (mouse equivalent to human Ser(15)) and the subsequent transcriptional activation of PUMA, a known proapoptotic p53-target gene, and promoted apoptosis involving increased overall caspase activity, caspase-3 activation, cleavage of poly(ADP-ribose) polymerase (PARP) and classic protein kinase C (PKC) isoforms, and DNA fragmentation. The proapoptotic role of mTOR in this process was demonstrated by the fact that rapamycin, a mTOR inhibitor, blocked p53 Ser(18) phosphorylation, the induction of PUMA, and all other apoptosis events. Furthermore, the proapoptotic function of mTOR was also antagonized by the expression in MEF3T3 cells of the PCPH oncoprotein, known to enhance cell survival by causing partial ATP depletion. Tetracyclin (Tet)-regulated expression of oncogenic PCPH, or overexpression of normal PCPH, blocked both phosphorylation and nuclear shuttling of mTOR in response to IR. These results indicate that alterations in PCPH expression may render tumor cells resistant to IR, and perhaps other DNA-damaging agents, by preventing mTOR activation and signaling.


Subject(s)
Apoptosis/radiation effects , Fibroblasts/radiation effects , Oncogene Proteins/physiology , Protein Kinase Inhibitors , Radiation Tolerance/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , DNA Damage/physiology , Dose-Response Relationship, Radiation , Fibroblasts/cytology , Mice , Mitochondria/metabolism , Mitochondria/radiation effects , Oncogene Proteins/biosynthesis , Phosphorylation/radiation effects , Protein Kinases/biosynthesis , Protein Kinases/metabolism , Protein Kinases/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Radiation Tolerance/drug effects , Signal Transduction/physiology , Signal Transduction/radiation effects , Sirolimus/pharmacology , Swiss 3T3 Cells , TOR Serine-Threonine Kinases , Transcriptional Activation/radiation effects , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...