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.
Cell Cycle ; 9(16): 3256-76, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20814239

ABSTRACT

Loss of stromal fibroblast caveolin-1 (Cav-1) is a powerful single independent predictor of poor prognosis in human breast cancer patients, and is associated with early tumor recurrence, lymph node metastasis and tamoxifen-resistance. We developed a novel co-culture system to understand the mechanism(s) by which a loss of stromal fibroblast Cav-1 induces a "lethal tumor micro-environment." Here, we propose a new paradigm to explain the powerful prognostic value of stromal Cav-1. In this model, cancer cells induce oxidative stress in cancer-associated fibroblasts, which then acts as a "metabolic" and "mutagenic" motor to drive tumor-stroma co-evolution, DNA damage and aneuploidy in cancer cells. More specifically, we show that an acute loss of Cav-1 expression leads to mitochondrial dysfunction, oxidative stress and aerobic glycolysis in cancer associated fibroblasts. Also, we propose that defective mitochondria are removed from cancer-associated fibroblasts by autophagy/mitophagy that is induced by oxidative stress. As a consequence, cancer associated fibroblasts provide nutrients (such as lactate) to stimulate mitochondrial biogenesis and oxidative metabolism in adjacent cancer cells (the "Reverse Warburg Effect"). We provide evidence that oxidative stress in cancer-associated fibroblasts is sufficient to induce genomic instability in adjacent cancer cells, via a bystander effect, potentially increasing their aggressive behavior. Finally, we directly demonstrate that nitric oxide (NO) over-production, secondary to Cav-1 loss, is the root cause for mitochondrial dysfunction in cancer associated fibroblasts. In support of this notion, treatment with anti-oxidants (such as N-acetyl-cysteine, metformin and quercetin) or NO inhibitors (L-NAME) was sufficient to reverse many of the cancer-associated fibroblast phenotypes that we describe. Thus, cancer cells use "oxidative stress" in adjacent fibroblasts (i) as an "engine" to fuel their own survival via the stromal production of nutrients and (ii) to drive their own mutagenic evolution towards a more aggressive phenotype, by promoting genomic instability. We also present evidence that the "field effect" in cancer biology could also be related to the stromal production of ROS and NO species. eNOS-expressing fibroblasts have the ability to downregulate Cav-1 and induce mitochondrial dysfunction in adjacent fibroblasts that do not express eNOS. As such, the effects of stromal oxidative stress can be laterally propagated, amplified and are effectively "contagious"--spread from cell-to-cell like a virus--creating an "oncogenic/mutagenic" field promoting widespread DNA damage.


Subject(s)
Breast Neoplasms/metabolism , Caveolin 1/metabolism , Fibroblasts/metabolism , Genomic Instability , Oxidative Stress , Autophagy , Biological Evolution , Breast Neoplasms/genetics , Caveolin 1/genetics , Cell Line , Coculture Techniques , DNA Damage , Down-Regulation , Female , Histones/metabolism , Humans , Lactic Acid/pharmacology , Mitochondria/metabolism , Mitochondria/physiology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism
2.
Cancer Res ; 70(19): 7489-99, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20709760

ABSTRACT

Caveolin-1 (CAV1) is the main structural component of caveolae, which are plasma membrane invaginations that participate in vesicular trafficking and signal transduction events. Although evidence describing the function of CAV1 in several cancer types has recently accumulated, its role in melanoma tumor formation and progression remains poorly explored. Here, by using B16F10 melanoma cells as an experimental system, we directly explore the function of CAV1 in melanoma tumor growth and metastasis. We first show that CAV1 expression promotes proliferation, whereas it suppresses migration and invasion of B16F10 cells in vitro. When orthotopically implanted in the skin of mice, B16F10 cells expressing CAV1 form tumors that are similar in size to their control counterparts. An experimental metastasis assay shows that CAV1 expression suppresses the ability of B16F10 cells to form lung metastases in C57Bl/6 syngeneic mice. Additionally, CAV1 protein and mRNA levels are found to be significantly reduced in human metastatic melanoma cell lines and human tissue from metastatic lesions. Finally, we show that following integrin activation, B16F10 cells expressing CAV1 display reduced expression levels and activity of FAK and Src proteins. Furthermore, CAV1 expression markedly reduces the expression of integrin ß(3) in B16F10 melanoma cells. In summary, our findings provide experimental evidence that CAV1 may function as an antimetastatic gene in malignant melanoma.


Subject(s)
Caveolin 1/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Melanoma, Experimental/metabolism , Melanoma/metabolism , src-Family Kinases/metabolism , Animals , Caveolin 1/biosynthesis , Caveolin 1/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement/physiology , Female , Humans , Lung Neoplasms/secondary , Melanocytes/metabolism , Melanoma/genetics , Melanoma/pathology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Signal Transduction , src-Family Kinases/antagonists & inhibitors
3.
Cell Cycle ; 9(12): 2423-33, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20562526

ABSTRACT

Loss of stromal caveolin 1 (Cav-1) is a novel biomarker for cancer-associated fibroblasts that predicts poor clinical outcome in breast cancer and DCIS patients. We hypothesized that epithelial cancer cells may have the ability to drive Cav-1 downregulation in adjacent normal fibroblasts, thereby promoting the cancer associated fibroblast phenotype. To test this hypothesis directly, here we developed a novel co-culture model employing (i) human breast cancer cells (MCF7), and (ii) immortalized fibroblasts (hTERT-BJ1), which are grown under defined experimental conditions. Importantly, we show that co-culture of immortalized human fibroblasts with MCF7 breast cancer cells leads to Cav-1 downregulation in fibroblasts. These results were also validated using primary cultures of normal human mammary fibroblasts co-cultured with MCF7 cells. In this system, we show that Cav-1 downregulation is mediated by autophagic/lysosomal degradation, as pre-treatment with lysosome-specific inhibitors rescues Cav-1 expression. Functionally, we demonstrate that fibroblasts co-cultured with MCF7 breast cancer cells acquire a cancer associated fibroblast phenotype, characterized by Cav-1 downregulation, increased expression of myofibroblast markers and extracellular matrix proteins, and constitutive activation of TGFß/Smad2 signaling. siRNA-mediated Cav-1 downregulation mimics several key changes that occur in co-cultured fibroblasts, clearly indicating that a loss of Cav-1 is a critical initiating factor, driving stromal fibroblast activation during tumorigenesis. As such, this co-culture system can now be used as an experimental model for generating "synthetic" cancer associated fibroblasts (CAFs). More specifically, these "synthetic" CAFs could be used for drug screening to identify novel therapeutics that selectively target the Cav-1-negative tumor micro-environment. Our findings also suggest that chloroquine, or other autophagy/lysosome inhibitors, may be useful as anti-cancer agents, to therapeutically restore the expression of stromal Cav-1 in cancer associated fibroblasts. We discuss this possibility, in light of the launch of a new clinical trial that uses chloroquine to treat DCIS patients: PINC (Preventing Invasive Breast Neoplasia with Cholorquine) [See http://clinicaltrials.gov/show/NCT01023477].


Subject(s)
Autophagy/drug effects , Breast Neoplasms/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Caveolin 1/metabolism , Fibroblasts/metabolism , Actins/biosynthesis , Actins/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , Caveolin 1/genetics , Cell Line, Tumor , Chloroquine/pharmacology , Coculture Techniques , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Female , Humans , Microfilament Proteins/biosynthesis , Microfilament Proteins/genetics , Phenotype , Prognosis , Smad2 Protein/biosynthesis , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/metabolism , Vimentin/biosynthesis , Vimentin/genetics , Calponins
4.
Am J Pathol ; 174(4): 1172-90, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19342371

ABSTRACT

Caveolin-1 (Cav-1) loss-of-function mutations are exclusively associated with estrogen receptor-positive (ER(+)) human breast cancers. To dissect the role of Cav-1 loss-of-function in the pathogenesis of human breast cancers, we used Cav-1(-/-) null mice as a model system. First, we demonstrated that Cav-1(-/-) mammary epithelia overexpress two well-established ER co-activator genes, CAPER and Foxa1, in addition to ER-alpha. Thus, the functional loss of Cav-1 may be sufficient to confer estrogen-hypersensitivity in the mammary gland. To test this hypothesis directly, we subjected Cav-1(-/-) mice to ovariectomy and estrogen supplementation. As predicted, Cav-1(-/-) mammary glands were hyper-responsive to estrogen and developed dysplastic mammary lesions with adjacent stromal angiogenesis that resemble human ductal carcinoma in situ. Based on an extensive biomarker analysis, these Cav-1(-/-) mammary lesions contain cells that are hyperproliferative and stain positively with nucleolar (B23/nucleophosmin) and stem/progenitor cell markers (SPRR1A and beta-catenin). Genome-wide transcriptional profiling identified many estrogen-related genes that were over-expressed in Cav-1(-/-) mammary glands, including CAPER--an ER co-activator gene and putative stem/progenitor cell marker. Analysis of human breast cancer samples revealed that CAPER is overexpressed and undergoes a cytoplasmic-to-nuclear shift during the transition from pre-malignancy to ductal carcinoma in situ. Thus, Cav-1(-/-) null mice are a new preclinical model for studying the molecular paradigm of estrogen hypersensitivity and the development of estrogen-dependent ductal carcinoma in situ lesions.


Subject(s)
Carcinoma, Intraductal, Noninfiltrating/genetics , Caveolin 1/genetics , Estrogens/pharmacology , Gene Expression Profiling , Mammary Neoplasms, Experimental/genetics , Animals , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Intraductal, Noninfiltrating/pathology , Caveolin 1/deficiency , Cell Transformation, Neoplastic/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Immunohistochemistry , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Ovariectomy , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Tissue Array Analysis , Trans-Activators/genetics , Trans-Activators/metabolism
5.
Am J Pathol ; 174(5): 1650-62, 2009 May.
Article in English | MEDLINE | ID: mdl-19395651

ABSTRACT

Here we used the Met-1 cell line in an orthotopic transplantation model in FVB/N mice to dissect the role of the Cav-1(P132L) mutation in human breast cancer. Identical experiments were performed in parallel with wild-type Cav-1. Cav-1(P132L) up-regulated the expression of estrogen receptor-alpha as predicted, because only estrogen receptor-alpha-positive patients have been shown to harbor Cav-1(P132L) mutations. In the context of primary tumor formation, Cav-1(P132L) behaved as a loss-of-function mutation, lacking any tumor suppressor activity. In contrast, Cav-1(P132L) caused significant increases in cell migration, invasion, and experimental metastasis, consistent with a gain-of-function mutation. To identify possible molecular mechanism(s) underlying this invasive gain-of-function activity, we performed unbiased gene expression profiling. From this analysis, we show that the Cav-1(P132L) expression signature contains numerous genes that have been previously associated with cell migration, invasion, and metastasis. These include i) secreted growth factors and extracellular matrix proteins (Cyr61, Plf, Pthlh, Serpinb5, Tnc, and Wnt10a), ii) proteases that generate EGF and HGF (Adamts1 and St14), and iii) tyrosine kinase substrates and integrin signaling/adapter proteins (Akap13, Cdcp1, Ddef1, Eps15, Foxf1a, Gab2, Hs2st1, and Itgb4). Several of the P132L-specific genes are also highly expressed in stem/progenitor cells or are associated with myoepithelial cells, suggestive of an epithelial-mesenchymal transition. These results directly support clinical data showing that patients harboring Cav-1 mutations are more likely to undergo recurrence and metastasis.


Subject(s)
Biomarkers, Tumor/genetics , Caveolin 1/genetics , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mutation/genetics , Neoplastic Stem Cells/pathology , Animals , Biomarkers, Tumor/metabolism , Blotting, Western , Caveolin 1/metabolism , Cell Movement , Cell Proliferation , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mammary Neoplasms, Animal/metabolism , Mice , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Prognosis , Signal Transduction
6.
Am J Pathol ; 174(3): 746-61, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19234134

ABSTRACT

Recently, we reported that human breast cancer-associated fibroblasts show functional inactivation of the retinoblastoma (RB) tumor suppressor and down-regulation of caveolin-1 (Cav-1) protein expression. However, it remains unknown whether loss of Cav-1 is sufficient to confer functional RB inactivation in mammary fibroblasts. To establish a direct cause-and-effect relationship, mammary stromal fibroblasts (MSFs) were prepared from Cav-1(-/-) null mice and subjected to phenotypic analysis. Here, we provide evidence that Cav-1(-/-) MSFs share many characteristics with human cancer-associated fibroblasts. The Cav-1(-/-) MSF transcriptome significantly overlaps with human cancer-associated fibroblasts; both show a nearly identical profile of RB/E2F-regulated genes that are up-regulated, which is consistent with RB inactivation. This Cav-1(-/-) MSF gene signature is predictive of poor clinical outcome in breast cancer patients treated with tamoxifen. Consistent with these findings, Cav-1(-/-) MSFs show RB hyperphosphorylation and the up-regulation of estrogen receptor co-activator genes. We also evaluated the paracrine effects of "conditioned media" prepared from Cav-1(-/-) MSFs on wild-type mammary epithelia. Our results indicate that Cav-1(-/-) MSF "conditioned media" is sufficient to induce an epithelial-mesenchymal transition, indicative of an invasive phenotype. Proteomic analysis of this "conditioned media" reveals increased levels of proliferative/angiogenic growth factors. Consistent with these findings, Cav-1(-/-) MSFs are able to undergo endothelial-like transdifferentiation. Thus, these results have important implications for understanding the role of cancer-associated fibroblasts and RB inactivation in promoting tumor angiogenesis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Caveolin 1/deficiency , Caveolin 1/genetics , Fibroblasts/pathology , Stromal Cells/pathology , Blotting, Western , Breast/cytology , Breast/physiology , Breast Neoplasms/mortality , Cell Culture Techniques , Cell Division , Disease Progression , Disease-Free Survival , Epithelial Cells/cytology , Epithelial Cells/pathology , Epithelial Cells/physiology , Female , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Stromal Cells/cytology , Stromal Cells/physiology , Survival Analysis
7.
Am J Pathol ; 174(2): 613-29, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19164602

ABSTRACT

Here, we show that functional loss of a single gene is sufficient to confer constitutive milk protein production and protection against mammary tumor formation. Caveolin-3 (Cav-3), a muscle-specific caveolin-related gene, is highly expressed in muscle cells. We demonstrate that Cav-3 is also expressed in myoepithelial cells within the mammary gland. To determine whether genetic ablation of Cav-3 expression affects adult mammary gland development, we studied the phenotype(s) of Cav-3(-/-)-null mice. Interestingly, Cav-3(-/-) virgin mammary glands developed lobulo-alveolar hyperplasia, akin to the changes normally observed during pregnancy and lactation. Genome-wide expression profiling revealed up-regulation of gene transcripts associated with pregnancy/lactation, mammary stem cells, and human breast cancers, consistent with a constitutive lactogenic phenotype. Expression levels of three key transcriptional regulators of lactation, namely Elf5, Stat5a, and c-Myc, were also significantly elevated. Experiments with pregnant mice directly showed that Cav-3(-/-) mice underwent precocious lactation. Finally, using orthotopic tumor cell implantation, we demonstrated that virgin Cav-3(-/-) mice were dramatically protected against mammary tumor formation. Thus, Cav-3(-/-) mice are a novel preclinical model to study the protective effects of a lactogenic microenvironment on mammary tumor onset and progression. Our current studies have broad implications for using the lactogenic microenvironment as a paradigm to discover new therapies for the prevention and/or treatment of human breast cancers.


Subject(s)
Caveolin 3/genetics , Caveolin 3/metabolism , Gene Expression , Lactation/physiology , Mammary Neoplasms, Experimental/genetics , Animals , Cell Movement/physiology , Female , Gene Expression Profiling , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Mutant Strains , Milk, Human/metabolism , Oligonucleotide Array Sequence Analysis , Phenotype , Polymerase Chain Reaction , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...