Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Oncogene ; 43(24): 1824-1835, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38654106

ABSTRACT

We have performed a functional in vivo mutagenesis screen to identify genes that, when altered, cooperate with a heterozygous Pten mutation to promote prostate tumour formation. Two genes, Bzw2 and Eif5a2, which have been implicated in the process of protein translation, were selected for further validation. Using prostate organoid models, we show that either Bzw2 downregulation or EIF5A2 overexpression leads to increased organoid size and in vivo prostate growth. We show that both genes impact the PI3K pathway and drive a sustained increase in phospho-AKT expression, with PTEN protein levels reduced in both models. Mechanistic studies reveal that EIF5A2 is directly implicated in PTEN protein translation. Analysis of patient datasets identified EIF5A2 amplifications in many types of human cancer, including the prostate. Human prostate cancer samples in two independent cohorts showed a correlation between increased levels of EIF5A2 and upregulation of a PI3K pathway gene signature. Consistent with this, organoids with high levels of EIF5A2 were sensitive to AKT inhibitors. Our study identified novel genes that promote prostate cancer formation through upregulation of the PI3K pathway, predicting a strategy to treat patients with genetic aberrations in these genes particularly relevant for EIF5A2 amplified tumours.


Subject(s)
Eukaryotic Translation Initiation Factor 5A , PTEN Phosphohydrolase , Peptide Initiation Factors , Phosphatidylinositol 3-Kinases , Prostatic Neoplasms , RNA-Binding Proteins , Signal Transduction , Male , Humans , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Peptide Initiation Factors/genetics , Peptide Initiation Factors/metabolism , Signal Transduction/genetics , Animals , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Mice , Organoids/metabolism , Organoids/pathology , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Cell Line, Tumor
2.
J Pathol ; 262(1): 37-49, 2024 01.
Article in English | MEDLINE | ID: mdl-37792636

ABSTRACT

Salivary gland adenoid cystic carcinoma (ACC) is a rare malignancy with limited treatment options. The development of novel therapies is hindered by a lack of preclinical models. We have generated ACC patient-derived xenograft (PDX) lines that retain the physical and genetic properties of the original tumours, including the presence of the common MYB::NFIB or MYBL1::NFIB translocations. We have developed the conditions for the generation of both 2D and 3D tumour organoid patient-derived ACC models that retain MYB expression and can be used for drug studies. Using these models, we show in vitro and in vivo sensitivity of ACC cells to the bromodomain degrader, dBET6. Molecular studies show a decrease in BRD4 and MYB protein levels and target gene expression with treatment. The most prominent effect of dBET6 on tumours in vivo was a change in the relative composition of ACC cell types expressing either myoepithelial or ductal markers. We show that dBET6 inhibits the progenitor function of ACC cells, particularly in the myoepithelial marker-expressing population, revealing a cell-type-specific sensitivity. These studies uncover a novel mechanistic effect of bromodomain inhibitors on tumours and highlight the need to impact both cell-type populations for more effective treatments in ACC patients. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Carcinoma, Adenoid Cystic , Salivary Gland Neoplasms , Humans , Carcinoma, Adenoid Cystic/drug therapy , Carcinoma, Adenoid Cystic/genetics , Carcinoma, Adenoid Cystic/pathology , Nuclear Proteins/genetics , Transcription Factors/genetics , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/pathology , Cell Cycle Proteins/genetics
3.
Evol Med Public Health ; 9(1): 311-321, 2021.
Article in English | MEDLINE | ID: mdl-34754452

ABSTRACT

BACKGROUND AND OBJECTIVES: Several hundred inherited genetic variants or SNPs that alter the risk of cancer have been identified through genome-wide association studies. In populations of European ancestry, these variants are mostly present at relatively high frequencies. To gain insight into evolutionary origins, we screened a series of genes and SNPs linked to breast or prostate cancer for signatures of historical positive selection. METHODOLOGY: We took advantage of the availability of the 1000 genome data and we performed genomic scans for positive selection in five different Caucasian populations as well as one African reference population. We then used prostate organoid cultures to provide a possible functional explanation for the interplay between the action of evolutionary forces and the disease risk association. RESULTS: Variants in only one gene showed genomic signatures of positive, evolutionary selection within Caucasian populations melanophilin (MLPH). Functional depletion of MLPH in prostate organoids, by CRISPR/Cas9 mutation, impacted lineage commitment of progenitor cells promoting luminal versus basal cell differentiation and on resistance to androgen deprivation. CONCLUSIONS AND IMPLICATIONS: The MLPH variants influencing prostate cancer risk may have been historically selected for their adaptive benefit on skin pigmentation but MLPH is highly expressed in the prostate and the derivative, positively selected, alleles decrease the risk of prostate cancer. Our study suggests a potential functional mechanism via which MLPH and its genetic variants could influence risk of prostate cancer, as a serendipitous consequence of prior evolutionary benefits to another tissue. LAY SUMMARY: We screened a limited series of genomic variants associated with breast and prostate cancer risk for signatures of historical positive selection. Variants within the melanophilin (MLPH) gene fell into this category. Depletion of MLPH in prostate organoid cultures, suggested a potential functional mechanism for impacting on cancer risk, as a serendipitous consequence of prior evolutionary benefits to another tissue.

4.
Br J Cancer ; 124(4): 805-816, 2021 02.
Article in English | MEDLINE | ID: mdl-33214683

ABSTRACT

BACKGROUND: Understanding the pathways that drive adrenocortical carcinoma (ACC) is essential to the development of more effective therapies. This study investigates the role of the transcription factor HOXB9 and other HOX factors in ACC and its treatment. METHODS: We used transgenic mouse models to determine the role of Hoxb9 in adrenal tumour development. Patient transcriptomic data was analysed for the expression of HOX genes and their association with disease. Drug response studies on various adrenocortical models were done to establish novel therapeutic options. RESULTS: Our human ACC dataset analyses showed high expression of HOXB9, and other HOX factors, are associated with poorer prognosis. Transgenic overexpression of Hoxb9 in the adrenal cortex of mice with activated Ctnnb1 led to larger adrenal tumours. This phenotype was preferentially observed in male mice and was characterised by more proliferating cells and an increase in the expression of cell cycle genes, including Ccne1. Adrenal tumour cells were found to be dependent on HOX function for survival and were sensitive to a specific peptide inhibitor. CONCLUSIONS: These studies show Hoxb9 can promote adrenal tumour progression in a sex-dependent manner and have identified HOX factors as potential drug targets, leading to novel therapeutic approaches in ACC.


Subject(s)
Adrenal Cortex Neoplasms/drug therapy , Adrenal Cortex Neoplasms/genetics , Adrenocortical Carcinoma/drug therapy , Adrenocortical Carcinoma/genetics , Homeodomain Proteins/genetics , Peptides/pharmacology , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/pathology , Animals , Cell Proliferation/genetics , Female , Gene Expression , Homeodomain Proteins/biosynthesis , Humans , Male , Mice , Mice, Transgenic , Molecular Targeted Therapy , Peptides/genetics
5.
Oncogene ; 38(28): 5700-5724, 2019 07.
Article in English | MEDLINE | ID: mdl-31043708

ABSTRACT

Androgen receptor (AR) signalling is a key prostate cancer (PC) driver, even in advanced 'castrate-resistant' disease (CRPC). To systematically identify microRNAs (miRs) modulating AR activity in lethal disease, hormone-responsive and -resistant PC cells expressing a luciferase-based AR reporter were transfected with a miR inhibitor library; 78 inhibitors significantly altered AR activity. Upon validation, miR-346, miR-361-3p and miR-197 inhibitors markedly reduced AR transcriptional activity, mRNA and protein levels, increased apoptosis, reduced proliferation, repressed EMT, and inhibited PC migration and invasion, demonstrating additive effects with AR inhibition. Corresponding miRs increased AR activity through a novel and anti-dogmatic mechanism of direct association with AR 6.9 kb 3'UTR and transcript stabilisation. In addition, miR-346 and miR-361-3p modulation altered levels of constitutively active AR variants, and inhibited variant-driven PC cell proliferation, so may contribute to persistent AR signalling in CRPC in the absence of circulating androgens. Pathway analysis of AGO-PAR-CLIP-identified miR targets revealed roles in DNA replication and repair, cell cycle, signal transduction and immune function. Silencing these targets, including tumour suppressors ARHGDIA and TAGLN2, phenocopied miR effects, demonstrating physiological relevance. MiR-346 additionally upregulated the oncogene, YWHAZ, which correlated with grade, biochemical relapse and metastasis in patients. These AR-modulatory miRs and targets correlated with AR activity in patient biopsies, and were elevated in response to long-term enzalutamide treatment of patient-derived CRPC xenografts. In summary, we identified miRs that modulate AR activity in PC and CRPC, via novel mechanisms, and may represent novel PC therapeutic targets.


Subject(s)
MicroRNAs/physiology , Prostatic Neoplasms/drug therapy , Receptors, Androgen/physiology , 3' Untranslated Regions , Antisense Elements (Genetics) , Benzamides , Cell Line, Tumor , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Humans , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/pathology , Signal Transduction
6.
Clin Cancer Res ; 24(13): 3149-3162, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29555663

ABSTRACT

Purpose: Persistent androgen receptor (AR) signaling drives castration-resistant prostate cancer (CRPC) and confers resistance to AR-targeting therapies. Novel therapeutic strategies to overcome this are urgently required. We evaluated how bromodomain and extra-terminal (BET) protein inhibitors (BETi) abrogate aberrant AR signaling in CRPC.Experimental Design: We determined associations between BET expression, AR-driven transcription, and patient outcome; and the effect and mechanism by which chemical BETi (JQ1 and GSK1210151A; I-BET151) and BET family protein knockdown regulates AR-V7 expression and AR signaling in prostate cancer models.Results: Nuclear BRD4 protein expression increases significantly (P ≤ 0.01) with castration resistance in same patient treatment-naïve (median H-score; interquartile range: 100; 100-170) and CRPC (150; 110-200) biopsies, with higher expression at diagnosis associating with worse outcome (HR, 3.25; 95% CI, 1.50-7.01; P ≤ 0.001). BRD2, BRD3, and BRD4 RNA expression in CRPC biopsies correlates with AR-driven transcription (all P ≤ 0.001). Chemical BETi, and combined BET family protein knockdown, reduce AR-V7 expression and AR signaling. This was not recapitulated by C-MYC knockdown. In addition, we show that BETi regulates RNA processing thereby reducing alternative splicing and AR-V7 expression. Furthermore, BETi reduce growth of prostate cancer cells and patient-derived organoids with known AR mutations, AR amplification and AR-V7 expression. Finally, BETi, unlike enzalutamide, decreases persistent AR signaling and growth (P ≤ 0.001) of a patient-derived xenograft model of CRPC with AR amplification and AR-V7 expression.Conclusions: BETi merit clinical evaluation as inhibitors of AR splicing and function, with trials demonstrating their blockade in proof-of-mechanism pharmacodynamic studies. Clin Cancer Res; 24(13); 3149-62. ©2018 AACR.


Subject(s)
Antineoplastic Agents/therapeutic use , Molecular Targeted Therapy , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Proteins/antagonists & inhibitors , Proteins/metabolism , Alternative Splicing , Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Cell Cycle Proteins , Cell Line, Tumor , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Prognosis , Prostatic Neoplasms, Castration-Resistant/mortality , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Isoforms , RNA, Small Interfering/genetics , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Treatment Outcome
7.
Oncotarget ; 9(7): 7604-7615, 2018 Jan 26.
Article in English | MEDLINE | ID: mdl-29484136

ABSTRACT

Aggressive lethal prostate cancer is characterised by tumour invasion, metastasis and androgen resistance. Understanding the mechanisms by which localised disease progresses to advanced lethal stages is key to the development of effective therapies. Here we have identified a novel role for the transcription factor, SOX9, as a driver of aggressive invasive prostate cancer. Using genetically modified mouse models, we show that increased Sox9 expression in the prostate epithelia of animals with Pten loss leads to a highly invasive phenotype and metastasis. In depth analysis of these mice and related in vitro models reveals that SOX9 acts a key regulator of various processes that together promote tumour progression. We show that this factor promotes cell lineage plasticity with cells acquiring properties of basal stem cells and an increase in proliferation. In addition, increased SOX9 leads to changes in cytoskeleton and adhesion, deposition of extracellular matrix and epithelia to mesenchyme transition, properties of highly invasive cells. Analysis of castrated mice showed that the invasive phenotype driven by SOX9 is independent of androgen levels. Our study has identified a novel driver of prostate cancer progression and highlighted the cellular and molecular processes that are regulated by Sox9 to achieve invasive disease.

8.
Article in English | MEDLINE | ID: mdl-29229667

ABSTRACT

The prostate is a male exocrine gland that secretes components of the seminal fluid. In men, prostate tumors are one of the most prevalent cancers. Studies on the development of the prostate have given a better understanding of the processes and genes that are important in the formation of this organ and have provided insights into the mechanisms of prostate tumorigenesis. These developmental studies have provided evidence that some of the genes and signaling pathways involved in development are reactivated or deregulated during prostate cancer. The prostate goes through a number of different stages during organogenesis, which include organ specification, epithelial budding, branching morphogenesis, canalization, and cytodifferentiation. During development, these processes are tightly regulated, many of which are controlled by the male hormone androgens. The majority of prostate tumors remain hormone regulated, and antiandrogen therapy is a first-line therapy, highlighting the important link between prostate organogenesis and cancer. In this review, we describe some of the data on genes that have important roles during prostate development that also have strong evidence linking them to prostate cancer.


Subject(s)
Organogenesis/physiology , Prostate/embryology , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/physiology , Cell Differentiation/physiology , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/physiology , Humans , Male , Mice , Models, Animal , Prostate/cytology , Prostate/growth & development , Prostatic Neoplasms/etiology , Receptors, Androgen/physiology , SOX9 Transcription Factor/physiology , Signal Transduction/physiology , Testosterone/physiology
9.
PLoS One ; 10(6): e0129470, 2015.
Article in English | MEDLINE | ID: mdl-26076167

ABSTRACT

Gene expression and functional studies have indicated that the molecular programmes involved in prostate development are also active in prostate cancer. PTEN has been implicated in human prostate cancer and is frequently mutated in this disease. Here, using the Nkx3.1:Cre mouse strain and a genetic deletion approach, we investigate the role of Pten specifically in the developing mouse prostate epithelia. In contrast to its role in other developing organs, this gene is dispensable for the initial developmental processes such as budding and branching. However, as cytodifferentiation progresses, abnormal luminal cells fill the ductal lumens together with augmented epithelial proliferation. This phenotype resembles the hyperplasia seen in postnatal Pten deletion models that develop neoplasia at later stages. Consistent with this, gene expression analysis showed a number of genes affected that are shared with Pten mutant prostate cancer models, including a decrease in androgen receptor regulated genes. In depth analysis of the phenotype of these mice during development revealed that loss of Pten leads to the precocious differentiation of epithelial cells towards a luminal cell fate. This study provides novel insight into the role of Pten in prostate development as part of the process of coordinating the differentiation and proliferation of cell types in time and space to form a functional organ.


Subject(s)
Cell Differentiation/genetics , Epithelial Cells/cytology , PTEN Phosphohydrolase/physiology , Prostate/embryology , Animals , Antigens, Neoplasm/metabolism , Biomarkers/metabolism , Cell Adhesion Molecules/metabolism , Cluster Analysis , Clusterin/metabolism , Gene Deletion , Male , Mice , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prostate/cytology , Signal Transduction
10.
J Pathol ; 236(2): 186-200, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25692405

ABSTRACT

Germline mutations in the tumour suppressor BRCA2 predispose to breast, ovarian and a number of other human cancers. Brca2-deficient mouse models are used for preclinical studies but the pattern of genomic alterations in these tumours has not yet been described in detail. We have performed whole-exome DNA sequencing analysis of mouse mammary tumours from Blg-Cre Brca2(f/f) Trp53(f/f) animals, a model of BRCA2-deficient human cancer. We also used the sequencing data to estimate DNA copy number alterations in these tumours and identified a recurrent copy number gain in Met, which has been found amplified in other mouse mammary cancer models. Through a comparative genomic analysis, we identified several mouse Blg-Cre Brca2(f/f) Trp53(f/f) mammary tumour somatic mutations in genes that are also mutated in human cancer, but few of these genes have been found frequently mutated in human breast cancer. A more detailed analysis of these somatic mutations revealed a set of genes that are mutated in human BRCA2 mutant breast and ovarian tumours and that are also mutated in mouse Brca2-null, Trp53-null mammary tumours. Finally, a DNA deletion surrounded by microhomology signature found in human BRCA1/2-deficient cancers was not common in the genome of these mouse tumours. Although a useful model, there are some differences in the genomic landscape of tumours arising in Blg-Cre Brca2(f/f) Trp53(f/f) mice compared to human BRCA-mutated breast cancers. Therefore, this needs to be taken into account in the use of this model.


Subject(s)
Genes, BRCA2/physiology , Mammary Neoplasms, Experimental/genetics , Tumor Suppressor Protein p53/deficiency , Animals , Antigens, CD/genetics , Breast Neoplasms/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA Copy Number Variations/genetics , DNA, Neoplasm/genetics , Disease Models, Animal , Female , Gene Knockout Techniques , Germ-Line Mutation/genetics , Humans , Mice, Transgenic , Mutation, Missense/genetics , Ovarian Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Immunologic/genetics , Sequence Analysis, DNA , Signaling Lymphocytic Activation Molecule Family
11.
Dev Biol ; 395(2): 209-17, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25261715

ABSTRACT

The mammalian urogenital sinus (UGS) develops in a sex specific manner, giving rise to the prostate in the male and the sinus vagina in the embryonic female. Androgens, produced by the embryonic testis, have been shown to be crucial to this process. In this study we show that retinoic acid signaling is required for the initial stages of bud development from the male UGS. Enzymes involved in retinoic acid synthesis are expressed in the UGS mesenchyme in a sex specific manner and addition of ligand to female tissue is able to induce prostate-like bud formation in the absence of androgens, albeit at reduced potency. Functional studies in mouse organ cultures that faithfully reproduce the initiation of prostate development indicate that one of the roles of retinoic acid signaling in the male is to inhibit the expression of Inhba, which encodes the ßA subunit of Activin, in the UGS mesenchyme. Through in vivo genetic analysis and culture studies we show that inhibition of Activin signaling in the female UGS leads to a similar phenotype to that of retinoic acid treatment, namely bud formation in the absence of androgens. Our data also reveals that both androgens and retinoic acid have extra independent roles to that of repressing Activin signaling in the development of the prostate during fetal stages. This study identifies a novel role for retinoic acid as a mesenchymal factor that acts together with androgens to determine the position and initiation of bud development in the male UGS epithelia.


Subject(s)
Activins/metabolism , Organogenesis/physiology , Signal Transduction/physiology , Tretinoin/metabolism , Urogenital System/embryology , Activins/antagonists & inhibitors , Animals , DNA Primers , Female , Immunohistochemistry , In Situ Hybridization , Inhibin-beta Subunits/antagonists & inhibitors , Male , Mice , Prostate/embryology , Real-Time Polymerase Chain Reaction , Sex Factors , Tretinoin/pharmacology , Urogenital System/metabolism , beta-Galactosidase
12.
PLoS Genet ; 9(1): e1003180, 2013.
Article in English | MEDLINE | ID: mdl-23300485

ABSTRACT

Prostate cancer is a major cause of male death in the Western world, but few frequent genetic alterations that drive prostate cancer initiation and progression have been identified. ß-Catenin is essential for many developmental processes and has been implicated in tumorigenesis in many tissues, including prostate cancer. However, expression studies on human prostate cancer samples are unclear on the role this protein plays in this disease. We have used in vivo genetic studies in the embryo and adult to extend our understanding of the role of ß-Catenin in the normal and neoplastic prostate. Our gene deletion analysis revealed that prostate epithelial ß-Catenin is required for embryonic prostate growth and branching but is dispensable in the normal adult organ. During development, ß-Catenin controls the number of progenitors in the epithelial buds and regulates a discrete network of genes, including c-Myc and Nkx3.1. Deletion of ß-Catenin in a Pten deleted model of castration-resistant prostate cancer demonstrated it is dispensable for disease progression in this setting. Complementary overexpression experiments, through in vivo protein stabilization, showed that ß-Catenin promotes the formation of squamous epithelia during prostate development, even in the absence of androgens. ß-Catenin overexpression in combination with Pten loss was able to drive progression to invasive carcinoma together with squamous metaplasia. These studies demonstrate that ß-Catenin is essential for prostate development and that an inherent property of high levels of this protein in prostate epithelia is to drive squamous fate differentiation. In addition, they show that ß-Catenin overexpression can promote invasive prostate cancer in a clinically relevant model of this disease. These data provide novel information on cancer progression pathways that give rise to lethal prostate disease in humans.


Subject(s)
Carcinoma , PTEN Phosphohydrolase , Prostatic Neoplasms , beta Catenin , Animals , Carcinoma/metabolism , Carcinoma/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Neoplasm Invasiveness/pathology , Orchiectomy , Organ Culture Techniques , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Prostate/growth & development , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , beta Catenin/genetics , beta Catenin/metabolism
13.
PLoS Genet ; 6(6): e1000995, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20585617

ABSTRACT

Epidemiological studies have shown that one of the strongest risk factors for prostate cancer is a family history of the disease, suggesting that inherited factors play a major role in prostate cancer susceptibility. Germline mutations in BRCA2 predispose to breast and ovarian cancer with its predominant tumour suppressor function thought to be the repair of DNA double-strand breaks. BRCA2 has also been implicated in prostate cancer etiology, but it is unclear the impact that mutations in this gene have on prostate tumourigenesis. Here we have undertaken a genetic analysis in the mouse to determine the role of Brca2 in the adult prostate. We show that deletion of Brca2 specifically in prostate epithelia results in focal hyperplasia and low-grade prostate intraepithelial neoplasia (PIN) in animals over 12 months of age. Simultaneous deletion of Brca2 and the tumour suppressor Trp53 in prostate epithelia gave rise to focal hyperplasia and atypical cells at 6 months, leading to high-grade PIN in animals from 12 months. Epithelial cells in these lesions show an increase in DNA damage and have higher levels of proliferation, but also elevated apoptosis. Castration of Brca2;Trp53 mutant animals led to regression of PIN lesions, but atypical cells persisted that continued to proliferate and express nuclear androgen receptor. This study provides evidence that Brca2 can act as a tumour suppressor in the prostate, and the model we describe should prove useful in the development of new therapeutic approaches.


Subject(s)
BRCA2 Protein/metabolism , Cell Transformation, Neoplastic/metabolism , Disease Progression , Prostatic Intraepithelial Neoplasia/metabolism , Prostatic Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , BRCA2 Protein/deficiency , Cell Transformation, Neoplastic/genetics , DNA Damage , Male , Mice , Mice, Knockout , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Tumor Suppressor Protein p53/deficiency
14.
Differentiation ; 76(6): 728-35, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18557758

ABSTRACT

The mammalian prostate arises from the urogenital sinus under the influence of testicular androgens. Few factors have been identified to be important in the early stages of prostate development. Here we review the role of the transcription factor Sox9 in prostate development. Sox9 is a member of the Sox gene family that plays an important role during embryogenesis in the cellular differentiation of various tissues, including testicular Sertoli cells, neural crest cells and chondrocytes. This gene is expressed in the epithelia of all mouse prostatic lobes from the initial stages of their development. Mice with a prostate specific deletion of Sox9 showed a lack of ventral prostate development and abnormal anterior prostate differentiation. In depth analysis of these mutant animals suggested that Sox9 is required for the early differentiation of the prostate bud epithelia, consistent with the function of this factor in other developmental processes. These studies also revealed different phases of prostate bud development. These phases were characterized by being dependent on different molecular pathways and having lobe specific properties. Future studies on the identification of pathways regulated by Sox9 will provide insight into the molecular networks required for prostate epithelia differentiation.


Subject(s)
Prostate/growth & development , Prostatic Neoplasms/physiopathology , SOX9 Transcription Factor/physiology , Animals , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/physiology , Humans , Male , SOX9 Transcription Factor/genetics , Signal Transduction
15.
Dev Dyn ; 234(4): 1006-15, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16245338

ABSTRACT

The Notch family of receptors is involved in a wide variety of developmental processes, including cell fate specification, cell proliferation, and cell survival decisions during cell differentiation and tissue morphogenesis. Notch1 and Notch ligands are expressed in the developing limbs, and Notch signalling has been implicated in the formation of a variety of tissues that comprise the limb, such as the skeleton, musculature, and vasculature. Notch signalling has also been implicated in regulating overall limb size. We have used a conditional allele of Notch1 in combination with two different Cre transgenic lines to delete Notch1 function either in the limb mesenchyme or in the apical ectodermal ridge (AER) and limb ectoderm. We demonstrate that Notch signalling, involving Notch1 and Jagged2, is required to regulate the number of Fgf8-expressing cells that comprise the AER and that regulation of the levels of fibroblast growth factor signalling is important for the freeing of the digits during normal limb formation. Regulation of the extent of the AER is achieved by Notch signalling positively regulating apoptosis in the AER. We also demonstrate that Notch1 is not required for proper formation of all the derivatives of the limb mesenchyme.


Subject(s)
Apoptosis/physiology , Extremities/embryology , Gene Expression Regulation, Developmental , Membrane Proteins/metabolism , Receptor, Notch1/metabolism , Signal Transduction/physiology , Animals , Apoptosis/genetics , Ectoderm/cytology , Ectoderm/metabolism , Fibroblast Growth Factor 8/metabolism , Fluorescent Antibody Technique , In Situ Hybridization , In Situ Nick-End Labeling , Jagged-2 Protein , Limb Buds , Mice , Mice, Transgenic , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...