Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 182
Filter
1.
Cancer Res ; 79(20): 5316-5327, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31395607

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is associated with significant fibrosis. Recent findings have highlighted the profibrotic activity of tissue-resident macrophages in the pancreatic cancer microenvironment. Here, we show that neoplastic pancreatic epithelium, as well as a subset of tissue-resident macrophages, expresses the prolactin-receptor (PRLR). High mobility group box 1-induced prolactin expression in the pancreas maintained FAK1 and STAT3 phosphorylation within the epithelium and stroma. Gain-of-function and loss-of-function experiments demonstrated the essential role of prolactin in promoting collagen deposition and fibrosis. Finally, the signaling cascade downstream of prolactin/PRLR activated STAT3 rather than STAT5 in PDAC. These findings suggest that targeting prolactin together with IL6, a known major activator of STAT3, could represent a novel therapeutic strategy for treating pancreatic cancer. SIGNIFICANCE: Prolactin is a key factor in the cross-talk between the stroma and neoplastic epithelium, functioning to promote fibrosis and PDAC progression.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Neoplasms, Hormone-Dependent/pathology , Pancreatic Neoplasms/pathology , Prolactin/pharmacology , Animals , Carcinoma, Pancreatic Ductal/physiopathology , Cell Line, Tumor , Collagen/metabolism , Disease Progression , Epithelium/metabolism , Female , Fibrosis , Focal Adhesion Kinase 1/metabolism , Genes, Reporter , HMGB1 Protein/physiology , Humans , Macrophages/metabolism , Male , Metoclopramide , Mice , Mice, Knockout , Neoplasm Proteins/metabolism , Neoplasms, Hormone-Dependent/physiopathology , Pancreatic Neoplasms/physiopathology , Phosphorylation , Pregnancy , Prolactin/deficiency , Prolactin/physiology , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering/genetics , Receptors, Prolactin/genetics , Receptors, Prolactin/metabolism , Recombinant Proteins/pharmacology , STAT3 Transcription Factor/metabolism , Stromal Cells/metabolism
2.
Endocr Relat Cancer ; 22(6): T177-86, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26381288

ABSTRACT

Breast cancer stem cells (BCSCs) are potent tumor-initiating cells in breast cancer, the most common cancer among women. BCSCs have been suggested to play a key role in tumor initiation which can lead to disease progression and formation of metastases. Moreover, BCSCs are thought to be the unit of selection for therapy-resistant clones since they survive conventional treatments, such as chemotherapy, irradiation, and hormonal therapy. The importance of the role of hormones for both normal mammary gland and breast cancer development is well established, but it was not until recently that the effects of hormones on BCSCs have been investigated. This review will discuss recent studies highlighting how ovarian steroid hormones estrogen and progesterone, as well as therapies against them, can regulate BCSC activity.


Subject(s)
Breast Neoplasms/pathology , Estrogens/physiology , Neoplasms, Hormone-Dependent/pathology , Neoplastic Stem Cells/physiology , Progesterone/physiology , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/physiopathology , Cell Division , Cell Line, Tumor , Clone Cells/physiology , Disease Progression , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/physiopathology , Paracrine Communication , Receptors, Estrogen/physiology , Receptors, Progesterone/physiology , Signal Transduction
3.
Endocr Relat Cancer ; 22(6): 889-900, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26311513

ABSTRACT

Although invasive and metastatic progression via the epithelial-mesenchymal transition (EMT) and acquisition of resistance to castration are both critical steps in prostate cancer, the molecular mechanism of this interaction remains unclear. In this study, we aimed to elucidate the interaction of signaling between castration resistance and EMT, and to apply this information to the development of a novel therapeutic concept using transforming growth factor-ß (TGF-ß) inhibitor SB525334 combined with androgen-deprivation therapy against prostate cancer using an in vivo model. This study revealed that an EMT inducer (TGF-ß) induced full-length androgen receptor (AR) and AR variant expression. In addition, a highly invasive clone showed augmented full-length AR and AR variant expression as well as acquisition of castration resistance. Conversely, full-length AR and AR as well as Twist1 and mesenchymal molecules variant expression were up-regulated in castration-resistant LNCaP xenograft. Finally, TGF-ß inhibitor suppressed Twist1 and AR expression as well as prostate cancer growth combined with castration. Taken together, these results demonstrate that Twist1/AR signaling was augmented in castration resistant as well as mesenchymal-phenotype prostate cancer, indicating the molecular mechanism of mutual and functional crosstalk between EMT and castration resistance, which may play a crucial role in prostate carcinogenesis and progression.


Subject(s)
Adenocarcinoma/pathology , Androgens , Epithelial-Mesenchymal Transition/physiology , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/pathology , Nuclear Proteins/physiology , Prostatic Neoplasms/pathology , Receptors, Androgen/physiology , Twist-Related Protein 1/physiology , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/physiopathology , Adenocarcinoma/surgery , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic , Combined Modality Therapy , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/physiopathology , Neoplasms, Hormone-Dependent/surgery , Orchiectomy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/physiopathology , Prostatic Neoplasms/surgery , Quinoxalines/pharmacology , Quinoxalines/therapeutic use , RNA Interference , RNA, Small Interfering/genetics , Random Allocation , Receptors, Androgen/biosynthesis , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors , Xenograft Model Antitumor Assays
4.
Drugs ; 75(12): 1311-21, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26177891

ABSTRACT

Endocrine therapy is a key component of adjuvant treatment for premenopausal patients with endocrine-responsive tumors. It is commonly well tolerated, although side effects are a main concern in the selection of treatment options. Tamoxifen is still considered an adequate endocrine therapy in a large group of premenopausal patients (e.g. lower-risk patient, presence of co-morbidities, patient preference). Results of the SOFT and TEXT trials addressing new adjuvant endocrine treatment options in premenopausal patients were recently presented. Overall, in the SOFT study the premenopausal population did not benefit from the addition of ovarian function suppression (OFS). Nevertheless, for women at sufficient risk of recurrence to receive adjuvant chemotherapy and who maintained premenopausal estradiol, the addition of OFS to tamoxifen reduced the risk of recurrence. The magnitude of the effect was larger in younger patients. Moreover, in the SOFT and TEXT trials, adjuvant treatment with exemestane plus OFS, as compared with tamoxifen plus OFS, significantly improved disease-free survival, breast cancer-free interval and distant disease-free survival. However, premenopausal patients include heterogeneous subsets of women and tumors where costs and benefits of adjuvant endocrine therapy should be properly weighted. Issues specific for premenopausal patients, related to desire for pregnancy, family planning, safety, quality of life and subjective side effects, should be a priority in the therapeutic algorithm. Therefore, selecting the best-tolerated agent can enhance adherence to therapies and reduce the impact on quality of life and health status for these younger patients.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Estrogen Antagonists/therapeutic use , Neoplasms, Hormone-Dependent/drug therapy , Premenopause , Receptors, Estrogen/antagonists & inhibitors , Age Factors , Antineoplastic Agents, Hormonal/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Aromatase Inhibitors/adverse effects , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Chemotherapy, Adjuvant , Disease Progression , Disease-Free Survival , Estrogen Antagonists/adverse effects , Female , Humans , Neoplasm Recurrence, Local , Neoplasm Staging , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/mortality , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/physiopathology , Ovary/drug effects , Ovary/metabolism , Ovary/physiopathology , Patient Selection , Quality of Life , Receptors, Estrogen/metabolism , Risk Assessment , Risk Factors , Signal Transduction/drug effects , Time Factors , Treatment Outcome
5.
J Clin Invest ; 125(5): 2123-35, 2015 May.
Article in English | MEDLINE | ID: mdl-25893606

ABSTRACT

The G protein-coupled estrogen receptor (GPER) mediates both the genomic and nongenomic effects of estrogen and has been implicated in breast cancer development. Here, we compared GPER expression in cancerous tissue and adjacent normal tissue in patients with invasive ductal carcinoma (IDC) of the breast and determined that GPER is highly upregulated in cancerous cells. Additionally, our studies revealed that GPER stimulation activates yes-associated protein 1 (YAP) and transcriptional coactivator with a PDZ-binding domain (TAZ), 2 homologous transcription coactivators and key effectors of the Hippo tumor suppressor pathway, via the Gαq-11, PLCß/PKC, and Rho/ROCK signaling pathways. TAZ was required for GPER-induced gene transcription, breast cancer cell proliferation and migration, and tumor growth. Moreover, TAZ expression positively correlated with GPER expression in human IDC specimens. Together, our results suggest that the Hippo/YAP/TAZ pathway is a key downstream signaling branch of GPER and plays a critical role in breast tumorigenesis.


Subject(s)
Breast Neoplasms/physiopathology , Carcinoma, Ductal, Breast/physiopathology , Estrogens/physiology , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/physiopathology , Protein Serine-Threonine Kinases/physiology , Receptors, Estrogen/physiology , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Tumor Suppressor Proteins/physiology , Acyltransferases , Adaptor Proteins, Signal Transducing/physiology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Cell Division , Cell Movement , Cell Transformation, Neoplastic , Estrogens/pharmacology , Female , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Gene Expression Regulation, Neoplastic , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/metabolism , Phospholipase C beta/physiology , Phosphoproteins/physiology , Phosphorylation , Protein Kinase C/physiology , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptors, Estrogen/drug effects , Receptors, G-Protein-Coupled/drug effects , Serine-Threonine Kinase 3 , Transcription Factors/physiology , Transcription, Genetic , Tumor Suppressor Proteins/analysis , YAP-Signaling Proteins , rho-Associated Kinases/physiology
6.
Proc Natl Acad Sci U S A ; 111(46): 16514-9, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25368192

ABSTRACT

It recently has been recognized that men develop colonic adenomas and carcinomas at an earlier age and at a higher rate than women. In the Apc(Pirc/+) (Pirc) rat model of early colonic cancer, this sex susceptibility was recapitulated, with male Pirc rats developing twice as many adenomas as females. Analysis of large datasets revealed that the Apc(Min/+) mouse also shows enhanced male susceptibility to adenomagenesis, but only in the colon. In addition, WT mice treated with injections of the carcinogen azoxymethane (AOM) showed increased numbers of colonic adenomas in males. The mechanism underlying these observations was investigated by manipulation of hormonal status. The preponderance of colonic adenomas in the Pirc rat model allowed a statistically significant investigation in vivo of the mechanism of sex hormone action on the development of colonic adenomas. Females depleted of endogenous hormones by ovariectomy did not exhibit a change in prevalence of adenomas, nor was any effect observed with replacement of one or a combination of female hormones. In contrast, depletion of male hormones by orchidectomy (castration) markedly protected the Pirc rat from adenoma development, whereas supplementation with testosterone reversed that effect. These observations were recapitulated in the AOM mouse model. Androgen receptor was undetectable in the colon or adenomas, making it likely that testosterone acts indirectly on the tumor lineage. Our findings suggest that indirect tumor-promoting effects of testosterone likely explain the disparity between the sexes in the development of colonic adenomas.


Subject(s)
Adenoma/epidemiology , Carcinogens/toxicity , Colonic Neoplasms/epidemiology , Dihydrotestosterone/toxicity , Gonadal Steroid Hormones/physiology , Neoplasms, Hormone-Dependent/epidemiology , Adenoma/chemically induced , Adenoma/physiopathology , Adenoma/prevention & control , Adenomatous Polyposis Coli/epidemiology , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/physiopathology , Animals , Animals, Congenic , Azoxymethane/toxicity , Colonic Neoplasms/chemically induced , Colonic Neoplasms/physiopathology , Colonic Neoplasms/prevention & control , Disease Models, Animal , Estradiol/administration & dosage , Estradiol/pharmacology , Female , Genes, APC , Hormone Replacement Therapy , Humans , Male , Medroxyprogesterone Acetate/administration & dosage , Medroxyprogesterone Acetate/pharmacology , Mice , Mice, Inbred C57BL , Mutation , Neoplasms, Hormone-Dependent/physiopathology , Neoplasms, Hormone-Dependent/prevention & control , Orchiectomy , Organ Specificity , Ovariectomy , Postmenopause , RNA, Messenger/analysis , Random Allocation , Rats , Rats, Inbred F344 , Rats, Mutant Strains , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Sex Distribution , Species Specificity
7.
Br J Surg ; 101(8): 939-48, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24687409

ABSTRACT

BACKGROUND: Aromatase inhibitors (AIs) are central to the management of oestrogen receptor-positive breast cancer in the adjuvant and metastatic setting. Levels of circulating steroid hormones (SHs) were measured in patients established on AIs to investigate: the influence of body mass index (BMI) in both the adjuvant and metastatic setting; the class of AI utilized in the adjuvant setting (steroidal versus non-steroidal); and differences in SH levels between women treated adjuvantly and those receiving a second-line AI for locally advanced/metastatic disease. METHODS: Plasma levels of androstenedione, 5-androstene-3ß,17ß-diol, dehydroepiandrosterone, oestradiol and testosterone were measured by radioimmunoassay in women with breast cancer who were receiving AIs in either an adjuvant or a metastatic setting. Differences between mean SH levels by class of AI, BMI, and second-line versus adjuvant therapy were assessed. RESULTS: Sixty-four women were receiving AI therapy, 45 (70 per cent) in an adjuvant setting and 19 (30 per cent) were taking a second-line AI. There was no significant correlation between BMI and SH levels. However, BMI was significantly higher in the second-line AI cohort compared with the adjuvant cohort (29.8 versus 26.2 kg/m2 respectively; P = 0.026). In the adjuvant setting, patients receiving a steroidal AI had significantly higher levels of all five hormones (P < 0.050). In the second-line AI cohort, oestradiol levels were significantly higher than in the adjuvant cohort (4.5 versus 3.3 pg/ml respectively; P = 0.022). Multivariable analysis adjusted for BMI confirmed the higher residual oestradiol level in the second-line AI group (P = 0.063) and a significantly higher androstenedione level (P = 0.022). CONCLUSION: Residual levels of SH were not significantly influenced by BMI. However, the significant differences in residual SH levels between the second-line and adjuvant AI cohort is of relevance in the context of resistance to AI therapy, and warrants further investigation.


Subject(s)
Aromatase Inhibitors/therapeutic use , Body Mass Index , Breast Neoplasms/drug therapy , Neoplasms, Hormone-Dependent/drug therapy , Steroids/metabolism , Adult , Aged , Aged, 80 and over , Analysis of Variance , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Chemotherapy, Adjuvant , Female , Gonadal Hormones/metabolism , Humans , Middle Aged , Neoplasm Metastasis , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/physiopathology , Radioimmunoassay
8.
Mol Cell Biochem ; 386(1-2): 189-98, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24104455

ABSTRACT

Prostate cancer is one of the most common malignancies in men and is predicted to be the second leading cause of cancer-related deaths. After 6-18 months, hormone ablation treatment results in androgen-independent growth of cancer cells, metastasis and progression. The mechanism of androgen-independent growth of prostatic carcinoma cells is still unknown. Identification of factors that facilitate the transition from androgen-dependent to independent states is crucial in designing future diagnostics and medication strategies. To understand the biochemical meaning of hormone dependency deprivation, glycoproteins enriched profiles were compared between DU145 (hormone non-responding) and LNCaP (hormone responding) prostate cancer cells. These results allow for anticipation on the important role of glycosylation in malignant transformation. Both Tn antigen and complex antennary N-oligosaccharides were recognized. Their occurrence might be involved in the development and progression of tumor, and failure of hormone ablation therapy. Among identified proteins in androgen-sensitive cells nucleolin (P19338) was found that is widely described as apoptosis inhibitor, and also transporter of molecules from the membrane to the cytoplasm or nucleus. In addition, 14-3-3 protein family (P27348, P31946, P61981, P63104, P62258, Q04917, and P31947) was investigated across available databases as it forms stable complexes with glycoproteins. Our studies indicate that isoforms: sigma and eta were found in androgen-dependent prostate cancer cells, while other isoforms were present in androgen non-responding cells. 14-3-3 binding partners are involved in cancer pathogenesis. These findings may contribute to a better understanding of prostate cancer tumorigenesis and to a more efficient prognosis and individual therapy in a future. However, it still remains to be revealed how important those changes are for androgen dependency loss in prostate cancer patients carried out on clinically relevant populations.


Subject(s)
Androgens/physiology , Glycopeptides/metabolism , Neoplasms, Hormone-Dependent/physiopathology , Proteome , Blotting, Western , Cell Line, Tumor , Cell Transformation, Neoplastic , Chromatography, Affinity/methods , Electrophoresis, Polyacrylamide Gel , Humans , Male , Neoplasms, Hormone-Dependent/metabolism , Tandem Mass Spectrometry
10.
J Natl Cancer Inst ; 105(19): 1463-73, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-24052624

ABSTRACT

BACKGROUND: L-type amino acid transporters (LATs) uptake neutral amino acids including L-leucine into cells, stimulating mammalian target of rapamycin complex 1 signaling and protein synthesis. LAT1 and LAT3 are overexpressed at different stages of prostate cancer, and they are responsible for increasing nutrients and stimulating cell growth. METHODS: We examined LAT3 protein expression in human prostate cancer tissue microarrays. LAT function was inhibited using a leucine analog (BCH) in androgen-dependent and -independent environments, with gene expression analyzed by microarray. A PC-3 xenograft mouse model was used to study the effects of inhibiting LAT1 and LAT3 expression. Results were analyzed with the Mann-Whitney U or Fisher exact tests. All statistical tests were two-sided. RESULTS: LAT3 protein was expressed at all stages of prostate cancer, with a statistically significant decrease in expression after 4-7 months of neoadjuvant hormone therapy (4-7 month mean = 1.571; 95% confidence interval = 1.155 to 1.987 vs 0 month = 2.098; 95% confidence interval = 1.962 to 2.235; P = .0187). Inhibition of LAT function led to activating transcription factor 4-mediated upregulation of amino acid transporters including ASCT1, ASCT2, and 4F2hc, all of which were also regulated via the androgen receptor. LAT inhibition suppressed M-phase cell cycle genes regulated by E2F family transcription factors including critical castration-resistant prostate cancer regulatory genes UBE2C, CDC20, and CDK1. In silico analysis of BCH-downregulated genes showed that 90.9% are statistically significantly upregulated in metastatic castration-resistant prostate cancer. Finally, LAT1 or LAT3 knockdown in xenografts inhibited tumor growth, cell cycle progression, and spontaneous metastasis in vivo. CONCLUSION: Inhibition of LAT transporters may provide a novel therapeutic target in metastatic castration-resistant prostate cancer, via suppression of mammalian target of rapamycin complex 1 activity and M-phase cell cycle genes.


Subject(s)
Activating Transcription Factor 4/metabolism , Amino Acid Transport Systems, Basic/antagonists & inhibitors , Amino Acid Transport Systems, Basic/metabolism , Amino Acids/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Leucine/antagonists & inhibitors , Neoadjuvant Therapy/methods , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/physiopathology , Receptors, Androgen/metabolism , Activating Transcription Factor 4/drug effects , Amino Acid Transport Systems, Basic/genetics , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Biological Transport/drug effects , Cell Cycle/drug effects , Computer Simulation , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Leucine/metabolism , Luminescent Measurements , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/metabolism , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/physiopathology , Orchiectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Array Analysis , Receptors, Androgen/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
11.
Article in English | MEDLINE | ID: mdl-23714450

ABSTRACT

Adjuvant therapy with antiestrogens targeting estrogen receptor α (ER) signaling prevents disease recurrence in many patients with early-stage ER+ breast cancer. However, a significant number of cases exhibit de novo or acquired endocrine resistance. While other clinical subtypes of breast cancer (HER2+, triple-negative) have disproportionately higher rates of mortality, ER+ breast cancer is responsible for at least as many deaths because it is the most common subtype. Therefore, identifying mechanisms that drive endocrine resistance is a high clinical priority. A large body of experimental evidence indicates that oncogenic signaling pathways underlie endocrine resistance, including growth factor receptor tyrosine kinases (HER2, epidermal growth factor receptor [EGFR], fibroblast growth factor receptor 1/2 [FGFR], insulin-like growth factor-1 receptor [IGF-1R]/ insulin receptor [InsR]), PI3K/AKT/ mTOR, MAPK/ERK, Src, CDK4/CDK6, and ER itself. Combined targeting of ER and such pathways may be the most effective means to combat antiestrogen resistance, and clinical trials testing such strategies show promising results. Herein, we discuss pathways associated with endocrine resistance, biomarkers that may be useful to predict response to targeted agents, and avenues for further exploration to identify strategies for the treatment of patients with endocrine-resistant disease.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Estrogen Receptor Modulators/pharmacology , Estrogens , Neoplasms, Hormone-Dependent/drug therapy , Progesterone , Signal Transduction/drug effects , Antineoplastic Agents, Hormonal/therapeutic use , Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/physiopathology , Cell Cycle/drug effects , Cyclin-Dependent Kinases/physiology , Drug Resistance, Neoplasm/physiology , Estrogen Receptor Modulators/therapeutic use , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intercellular Signaling Peptides and Proteins/physiology , Neoplasm Proteins/drug effects , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/physiopathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptor, ErbB-2/physiology , Receptors, Estrogen/drug effects , Receptors, Growth Factor/drug effects , Receptors, Growth Factor/physiology , Receptors, Progesterone/drug effects , Selective Estrogen Receptor Modulators/pharmacology , Selective Estrogen Receptor Modulators/therapeutic use
12.
Front Biosci (Landmark Ed) ; 18(2): 748-55, 2013 01 01.
Article in English | MEDLINE | ID: mdl-23276959

ABSTRACT

Prostate cancer (PCa) is the most commonly diagnosed cancer among men in Western countries and is one of the leading causes of cancer deaths. The growth and progression of PCa is related to androgen levels. In cancer, nicroRNAs (miRs) function as either oncogenes or tumor suppressor genes. In androgen-dependent PCa, miRs play a role in the growth, development, progression, and metastasis of the disease, and are also involved in the response to therapy and therefore affect the prognosis. In this review, we focus on the role played by miRs concerning the mechanisms of androgen-dependent PCa.


Subject(s)
Androgens/physiology , MicroRNAs/genetics , Neoplasms, Hormone-Dependent/genetics , Prostatic Neoplasms/genetics , Disease Progression , Humans , Male , MicroRNAs/physiology , MicroRNAs/therapeutic use , Neoplasm Metastasis/genetics , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/physiopathology , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Receptors, Androgen/physiology
13.
Klin Padiatr ; 225(1): 3-7, 2013 Jan.
Article in German | MEDLINE | ID: mdl-23329621

ABSTRACT

During the last year, alternative androgen synthesis pathways have been discovered in humans. This review article highlights these new concepts of androgen synthesis.We performed a selective literature research using PubMed.After the discovery of a new androgen synthesis pathway in marsupials, this new path-way of androgen synthesis could be established in humans during the last year from two independent studies. One of them could demonstrate that two pathways of androgen synthesis are needed for male sexual differentiation in humans; the other study established that the new pathway is an important source of androgen synthesis in congenital adrenal hyperplasia due to 21-hydroxylase deficiency. Additionally, it has been shown that an alternative androgen synthesis pathway that bypasses testosterone drives castration resistant prostate cancer.New and alternative androgen path-ways occur in humans. Importantly, these path-ways remain cryptic for the clinician, because the androgen synthesis circumvents classical intermediates like dehydroepiandrosterone, androstenedione and testosterone.


Subject(s)
Adrenal Hyperplasia, Congenital/physiopathology , Androgens/biosynthesis , Sex Differentiation/physiology , Steroid 21-Hydroxylase/physiology , 17-alpha-Hydroxypregnenolone/metabolism , 17-alpha-Hydroxyprogesterone/metabolism , Adrenal Glands/physiopathology , Adrenal Hyperplasia, Congenital/diagnosis , Androstenedione/metabolism , Animals , Child , Child, Preschool , Dehydroepiandrosterone/metabolism , Dihydrotestosterone/metabolism , Female , Humans , Infant , Infant, Newborn , Male , Neoplasms, Hormone-Dependent/physiopathology , Orchiectomy , Pregnancy , Prostatic Neoplasms/physiopathology , Testis/physiopathology , Testosterone/metabolism
14.
Urol Clin North Am ; 39(4): 435-52, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23084522

ABSTRACT

Although androgen-deprivation therapy is the standard therapy for advanced and metastatic prostate cancer, this treatment is only palliative. Prostate cancer recurs then grows despite low circulating testicular androgens, using several mechanisms that remain dependent on androgen-receptor signaling in most cases. This article reviews the diversity of mechanisms used for growth by castration-recurrent prostate cancer.


Subject(s)
Neoplasm Recurrence, Local/physiopathology , Neoplasms, Hormone-Dependent/physiopathology , Prostatic Neoplasms/physiopathology , Receptors, Androgen/physiology , Androgen Antagonists/therapeutic use , Antineoplastic Agents/pharmacology , Gene Amplification , Gene Expression Regulation, Neoplastic/physiology , Humans , Male , Neoplasm Recurrence, Local/pathology , Prostate/chemistry , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Isoforms/physiology , Receptors, Androgen/biosynthesis , Receptors, Androgen/drug effects , Testosterone/biosynthesis , Transcriptional Activation/physiology
17.
Biochem J ; 447(1): 1-16, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22970934

ABSTRACT

Shc (Src homology and collagen homology) proteins are considered prototypical signalling adaptors in mammalian cells. Consisting of four unique members, ShcA, B, C and D, and multiple splice isoforms, the family is represented in nearly every cell type in the body, where it engages in an array of fundamental processes to transduce environmental stimuli. Two decades of investigation have begun to illuminate the mechanisms of the flagship ShcA protein, whereas much remains to be learned about the newest discovery, ShcD. It is clear, however, that the distinctive modular architecture of Shc proteins, their promiscuous phosphotyrosine-based interactions with a multitude of membrane receptors, involvement in central cascades including MAPK (mitogen-activated protein kinase) and Akt, and unconventional contributions to oxidative stress and apoptosis all require intricate regulation, and underlie diverse physiological function. From early cardiovascular development and neuronal differentiation to lifespan determination and tumorigenesis, Shc adaptors have proven to be more ubiquitous, versatile and dynamic than their structures alone suggest.


Subject(s)
Shc Signaling Adaptor Proteins/chemistry , Shc Signaling Adaptor Proteins/physiology , Animals , Apoptosis , Cardiovascular Physiological Phenomena , Female , History, 20th Century , History, 21st Century , Humans , Male , Mice , Mice, Knockout , Models, Biological , Neoplasms/physiopathology , Neoplasms, Hormone-Dependent/physiopathology , Nervous System Physiological Phenomena , Oxidative Stress , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/physiology , Shc Signaling Adaptor Proteins/history , Signal Transduction/physiology , src Homology Domains
18.
Minerva Endocrinol ; 37(2): 173-85, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22691890

ABSTRACT

The major issue currently being faced in the management of prostate cancer is the inability to distinguish between indolent prostate tumors that will not present clinically from more aggressive and metastatic prostate cancers that will impact on men's lives. Only a small proportion of prostate cancers can be accounted for by unmistakable hereditary cancer syndromes and the predominant contribution to the progression of most sporadic cancers is thought to be environmental, with nutrition having the greatest influence. Population studies have clearly implicated metabolic factors as contributors to disease progression and poor response to therapy. It is well established that the IGF system is key in regulating growth and metabolism and mediates the effects of nutrition on these processes. It consists of two ligands (IGF-I and IGF-II), two receptors [type 1 IGF-IR and IGF-II/mannose 6-phosphate receptor], and six high affinity IGF-binding proteins (IGFBP-1 to -6). This review provides evidence from in vitro, in vivo, clinical and epidemiology studies that indicates an important role for the IGF axis in the development of prostate cancer and the likely role that it plays in mediating the effects of nutrition on disease progression. We suggest that the IGF axis is central to understanding how lifestyle impacts on prostate cancer and we highlight this by describing numerous strategies being developed to target this axis.


Subject(s)
Adenocarcinoma/metabolism , Neoplasm Proteins/physiology , Prostatic Neoplasms/metabolism , Receptors, Somatomedin/physiology , Somatomedins/physiology , Adenocarcinoma/epidemiology , Adenocarcinoma/physiopathology , Adenocarcinoma/therapy , Androgens/metabolism , Antineoplastic Agents, Hormonal/therapeutic use , Disease Progression , Energy Metabolism , Humans , Incidence , Insulin-Like Growth Factor Binding Proteins , Male , Molecular Targeted Therapy , Neoplasms, Hormone-Dependent/epidemiology , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/physiopathology , Neoplasms, Hormone-Dependent/therapy , Overnutrition/complications , Overnutrition/metabolism , Overnutrition/physiopathology , Prevalence , Prostatectomy , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/physiopathology , Prostatic Neoplasms/therapy , Tumor Cells, Cultured , Zinc/metabolism
19.
Biol Reprod ; 87(1): 5, 1-10, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22553222

ABSTRACT

The endothelin axis, comprising endothelin-1 (ET-1) and its receptors (ETA and ETB), is involved in the pathophysiology of different human tumors. Here we review conventional approaches and gene expression profiling indicating the association of ET-1 and its cognate receptors with human and rat leiomyomas, the most common benign tumors of myometrium. Specifically, ET-1/ETA interactions affect human and rat leiomyoma cell proliferation through protein kinase C and mitogen-activated protein kinase-dependent signaling pathways. Recent experiments demonstrate that the ET-1 axis exerts a potent antiapoptotic effect involving sphingolipid metabolism and prostaglandin-endoperoxide synthase 2/prostaglandin system in the rat Eker leiomyoma tumor-derived ELT3 cell line. Evidence supports that steroid hormones, growth factors, and extracellular matrix are key regulators of the leiomyoma growth. Interestingly, the ET-1 axis is under steroid hormones and can cooperate with these growth factors. Therefore, ET-1 alone or in association with these factors could contribute to the complex regulation of uterine tumor growth, such as proliferation, survival, and extracellular matrix production. This review summarizes current knowledge and emerging data on ET-1 in uterine leiomyoma pathology.


Subject(s)
Endothelin-1/physiology , Leiomyoma/physiopathology , Uterine Neoplasms/physiopathology , Animals , Cell Proliferation , Endothelin-1/genetics , Female , Humans , Leiomyoma/genetics , Leiomyoma/pathology , Models, Biological , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/physiopathology , Rats , Receptors, Endothelin/genetics , Receptors, Endothelin/physiology , Signal Transduction , Transcriptome , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
20.
PLoS One ; 7(5): e36828, 2012.
Article in English | MEDLINE | ID: mdl-22606295

ABSTRACT

BACKGROUND: Growth factors activating the ErbB receptors have been described in prostate tumors. The androgen dependent prostate cancer cell line, LNCaP, expresses the ErbB-1, ErbB-2 and ErbB-3 receptor tyrosine kinases. Previously, it was demonstrated that NRG activates ErbB-2/ErbB-3 heterodimers to induce LNCaP cell death, whereas, EGF activates ErbB-1/ErbB-1 or ErbB-1/ErbB-2 dimers to induce cell growth and survival. It was also demonstrated that PI3K inhibitors repressed this cell death suggesting that in androgen deprived LNCaP cells, NRG activates a PI3K-dependent pathway associated with cell death. METHODOLOGY/PRINCIPAL FINDINGS: In the present study we demonstrate that NRG induces autophagy in LNCaP cells, using LC3 as a marker. However, the autophagy induced by NRG may be incomplete since p62 levels elevate. We also demonstrated that NRG- induced autophagy is independent of mammalian target of rapamycin (mTOR) inhibition since NRG induces Akt and S6K activation. Interestingly, inhibition of reactive oxygen species (ROS) by N-acetylcysteine (NAC), inhibited NRG-induced autophagy and cell death. Our study also identified JNK and Beclin 1 as important components in NRG-induced autophagy and cell death. NRG induced elevation in JNK phosphorylation that was inhibited by NAC. Moreover, inhibitor of JNK inhibited NRG-induced autophagy and cell death. Also, in cells overexpressing Bcl-2 or cells expressing sh-RNA against Beclin 1, the effects of NRG, namely induction of autophagy and cell death, were inhibited. CONCLUSIONS/SIGNIFICANCE: Thus, in LNCaP cells, NRG-induces incomplete autophagy and cell death that depend on ROS levels. These effects of NRG are mediated by signaling pathway that activates JNK and Beclin 1, but is independent of mTOR inhibition.


Subject(s)
Neuregulins/physiology , Prostatic Neoplasms/pathology , Prostatic Neoplasms/physiopathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Acetylcysteine/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Anthracenes/pharmacology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Autophagy/physiology , Beclin-1 , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Humans , MAP Kinase Signaling System/drug effects , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/pathology , Neoplasms, Hormone-Dependent/physiopathology , Neuregulins/pharmacology , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL