Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Am J Transl Res ; 5(1): 80-91, 2013.
Article in English | MEDLINE | ID: mdl-23390568

ABSTRACT

Caveolin-1 (Cav1) is the main protein component of the membrane lipid rafts caveolae. Cav1 serves as a scaffolding protein that compartmentalizes a multitude of signaling molecules and sequesters them in their inactive state. Due to its function in the negative regulation of signal transduction, loss of Cav1 has been implicated in the pathogenesis of many cancers, but its role in cutaneous squamous cell carcinoma (cSCC) is largely unexplored. cSCC is a multi-stage disease characterized by the development of benign, premalignant lesions and their progression into malignant cancer. Here, we use a two-stage carcinogenesis protocol to elucidate the function of Cav1 in the different stages of benign papilloma development: initiation and promotion. First, we demonstrate that Cav1 knock-out (KO) mice are more susceptible to benign papilloma development after being subjected to a DMBA/TPA initiation/promotion protocol. Treatment of wild-type (WT) and Cav1 KO mice with DMBA alone shows that both groups have similar rates of apoptosis. In contrast, treatment of these groups with TPA alone indicates that Cav1 KO mice are more susceptible to promoter treatment as evidenced by increased epidermal proliferation. Furthermore, primary keratinocytes isolated from Cav1 KO mice have a proliferative advantage over WT keratinocytes in both low- and high-calcium medium, conditions that promote proliferation and induce differentiation, respectively. Collectively, these data indicate that Cav1 functions to suppress proliferation in the epidermis, and loss of this function promotes the development of benign skin tumors.

2.
Am J Pathol ; 182(3): 992-1004, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23267770

ABSTRACT

Caveolin-1 (Cav1) is a scaffolding protein that serves to regulate the activity of several signaling molecules. Its loss has been implicated in the pathogenesis of several types of cancer, but its role in the development and progression of cutaneous squamous cell carcinoma (cSCC) remains largely unexplored. Herein, we use the keratinocyte cell line PAM212, a murine model of cSCC, to determine the function of Cav1 in skin tumor biology. We first show that Cav1 overexpression decreases cell and tumor growth, whereas Cav1 knockdown increases these attributes in PAM212 cells. In addition, Cav1 knockdown increases the invasive ability and incidence of spontaneous lymph node metastasis. Finally, we demonstrate that Cav1 knockdown increases extracellular signaling-related kinase 1/2 mitogen-activated protein kinase/activator protein-1 pathway activation. We attribute the growth and invasive advantage conferred by Cav1 knockdown to increased expression of activator protein-1 transcriptional targets, including cyclin D1 and keratin 18, which show inverse expression in PAM212 based on the expression level of Cav1. In summary, we demonstrate that loss of Cav1 affects several characteristics associated with aggressive human skin tumors and that this protein may be an important modulator of tumor growth and invasion in cSCC.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Caveolin 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Transcription Factor AP-1/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , Gene Knockdown Techniques , Humans , Keratin-18/metabolism , Keratinocytes/pathology , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Serum
3.
Cell Cycle ; 11(21): 3972-82, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23047602

ABSTRACT

Here, we identified the milk protein α-casein as a novel suppressor of tumor growth and metastasis. Briefly, Met-1 mammary tumor cells expressing α-casein showed a ~5-fold reduction in tumor growth and a near 10-fold decrease in experimental metastasis. To identify the molecular mechanism(s), we performed genome-wide transcriptional profiling. Interestingly, our results show that α-casein upregulates gene transcripts associated with interferon/STAT1 signaling and downregulates genes associated with "stemness." These findings were validated by immunoblot and FACS analysis, which showed the upregulation and hyperactivation of STAT1 and a decrease in the number of CD44(+) "cancer stem cells." These gene signatures were also able to predict clinical outcome in human breast cancer patients. Thus, we conclude that a lactation-based therapeutic strategy using recombinant α-casein would provide a more natural and non-toxic approach to the development of novel anticancer therapies.


Subject(s)
Caseins/pharmacology , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Caseins/metabolism , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Gene Expression Profiling , Humans , Hyaluronan Receptors/metabolism , Interferons/metabolism , Mice , Milk, Human , Neoplastic Stem Cells/metabolism , STAT1 Transcription Factor/genetics , Up-Regulation
4.
Cancer Res ; 72(9): 2262-74, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22396494

ABSTRACT

Both cell-autonomous and non-cell-autonomous factors contribute to tumor growth and metastasis of melanoma. The function of caveolin-1 (Cav1), a multifunctional scaffold protein known to modulate several biologic processes in both normal tissue and cancer, has been recently investigated in melanoma cancer cells, but its role in the melanoma microenvironment remains largely unexplored. Here, we show that orthotopic implantation of B16F10 melanoma cells in the skin of Cav1KO mice increases tumor growth, and co-injection of Cav1-deficient dermal fibroblasts with melanoma cells is sufficient to recapitulate the tumor phenotype observed in Cav1KO mice. Using indirect coculture experiments with fibroblasts and melanoma cells combined with cytokine analysis, we found that Cav1-deficient fibroblasts promoted the growth of melanoma cells via enhanced paracrine cytokine signaling. Specifically, Cav1-deficient fibroblasts displayed increased ShhN expression, which heterotypically enhanced the Shh signaling pathway in melanoma cells. In contrast to primary tumor growth, the ability of B16F10 melanoma cells to form lung metastases was significantly reduced in Cav1KO mice. This phenotype was associated mechanistically with the inability of melanoma cells to adhere to and to transmigrate through a monolayer of endothelial cells lacking Cav1. Together, our findings show that Cav1 may regulate different mechanisms during primary melanoma tumor growth and metastatic dissemination.


Subject(s)
Caveolin 1/deficiency , Cell Movement/genetics , Hedgehog Proteins/metabolism , Melanoma, Experimental/pathology , Animals , Caveolin 1/genetics , Caveolin 1/metabolism , Caveolin 2/deficiency , Caveolin 2/metabolism , Cell Growth Processes/genetics , Coculture Techniques , Cytokines/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasm Metastasis
5.
Cell Cycle ; 10(12): 2021-34, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21646868

ABSTRACT

We have previously demonstrated that loss of stromal caveolin-1 (Cav-1) in cancer-associated fibroblasts is a strong and independent predictor of poor clinical outcome in human breast cancer patients. However, the signaling mechanism(s) by which Cav-1 downregulation leads to this tumor-promoting microenvironment are not well understood. To address this issue, we performed an unbiased comparative proteomic analysis of wild-type (WT) and Cav-1(-/-) null mammary stromal fibroblasts (MSFs). Our results show that plasminogen activator inhibitor type 1 and type 2 (PAI-1 and PAI-2) expression is significantly increased in Cav-1(-/-) MSFs. To establish a direct cause-effect relationship, we next generated immortalized human fibroblast lines stably overexpressing either PAI-1 or PAI-2. Importantly, PAI-1/2(+) fibroblasts promote the growth of MDA-MB-231 tumors (a human breast cancer cell line) in a murine xenograft model, without any increases in angiogenesis. Similarly, PAI-1/2(+) fibroblasts stimulate experimental metastasis of MDA-MB-231 cells using an in vivo lung colonization assay. Further mechanistic studies revealed that fibroblasts overexpressing PAI-1 or PAI-2 display increased autophagy ("self-eating") and are sufficient to induce mitochondrial biogenesis/activity in adjacent cancer cells, in co-culture experiments. In xenografts, PAI-1/2(+) fibroblasts significantly reduce the apoptosis of MDA-MB-231 tumor cells. The current study provides further support for the "Autophagic Tumor Stroma Model of Cancer" and identifies a novel "extracellular matrix"-based signaling mechanism, by which a loss of stromal Cav-1 generates a metastatic phenotype. Thus, the secretion and remodeling of extracellular matrix components (such as PAI-1/2) can directly regulate both (1) autophagy in stromal fibroblasts and (2) epithelial tumor cell mitochondrial metabolism.


Subject(s)
Autophagy , Extracellular Matrix/metabolism , Mitochondria/metabolism , Neoplasm Metastasis , Stromal Cells/pathology , Animals , Breast Neoplasms/pathology , Caveolin 1/metabolism , Cell Line, Tumor , Coculture Techniques , Fibroblasts/pathology , Humans , Mice , Mitochondria/pathology , Neoplasms/pathology , Neoplasms/ultrastructure , Plasminogen Activator Inhibitor 1/metabolism , Plasminogen Activator Inhibitor 2/metabolism , Transplantation, Heterologous , Tumor Microenvironment
6.
Am J Pathol ; 178(1): 402-12, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21224077

ABSTRACT

Diet and obesity are important risk factors for cancer development. Many studies have suggested an important role for several dietary nutrients in the progression and development of breast cancer. However, few studies have specifically addressed the role of components of a Western diet as important factors involved in breast cancer initiation and progression. The present study examined the role of cholesterol in the regulation of tumor progression in a mouse model of mammary tumor formation. The results suggest that cholesterol accelerates and enhances tumor formation. In addition, tumors were more aggressive, and tumor angiogenesis was enhanced. Metabolism of cholesterol was also examined in this mouse model. It was observed that plasma cholesterol levels were reduced during tumor development but not prior to its initiation. These data provide new evidence for an increased utilization of cholesterol by tumors and for its role in tumor formation. Taken together, these results imply that an increase in plasma cholesterol levels accelerates the development of tumors and exacerbates their aggressiveness.


Subject(s)
Breast Neoplasms/etiology , Cell Transformation, Neoplastic , Cholesterol/adverse effects , Diet/adverse effects , Mammary Neoplasms, Experimental/etiology , Animals , Breast Neoplasms/blood , Breast Neoplasms/pathology , Cholesterol/administration & dosage , Cholesterol/blood , Female , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Protein Biosynthesis
7.
Cancer Biol Ther ; 11(4): 383-94, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21150282

ABSTRACT

We have recently proposed a new model for understanding tumor metabolism, termed: "The Autophagic Tumor Stroma Model of Cancer Metabolism". In this new paradigm, catabolism (autophagy) in the tumor stroma fuels the anabolic growth of aggressive cancer cells. Mechanistically, tumor cells induce autophagy in adjacent cancer-associated fibroblasts via the loss of caveolin-1 (Cav-1), which is sufficient to promote oxidative stress in stromal fibroblasts. To further test this hypothesis, here we created human Cav-1 deficient immortalized fibroblasts using a targeted sh-RNA knock-down approach. Relative to control fibroblasts, Cav-1 deficient fibroblasts dramatically promoted tumor growth in xenograft assays employing an aggressive human breast cancer cell line, namely MDA-MB-231 cells. Co-injection of Cav-1 deficient fibroblasts, with MDA-MB-231 cells, increased both tumor mass and tumor volume by ~4-fold. Immuno-staining with CD31 indicated that this paracrine tumor promoting effect was clearly independent of angiogenesis. Mechanistically, proteomic analysis of these human Cav-1 deficient fibroblasts identified > 40 protein biomarkers that were upregulated, most of which were associated with i) myofibroblast differentiation, or ii) oxidative stress/hypoxia. In direct support of these findings, the tumor promoting effects of Cav-1 deficient fibroblasts could be functionally suppressed (nearly 2-fold) by the recombinant over-expression of SOD2 (superoxide dismutase 2), a known mitochondrial enzyme that de-activates superoxide, thereby reducing mitochondrial oxidative stress. In contrast, cytoplasmic soluble SOD1 had no effect, further highlighting a specific role for mitochondrial oxidative stress in this process. In summary, here we provide new evidence directly supporting a key role for a loss of stromal Cav-1 expression and oxidative stress in cancer-associated fibroblasts, in promoting tumor growth, which is consistent with "The Autophagic Tumor Stroma Model of Cancer". The human Cav-1 deficient fibroblasts that we have generated are a new genetically tractable model system for identifying other suppressors of the cancer-associated fibroblast phenotype, via a genetic "complementation" approach. This has important implications for understanding the pathogenesis of triple negative and basal breasts cancers, as well as tamoxifen-resistance in ER+ breast cancers, which are all associated with a Cav-1 deficient "lethal" tumor micro-environment, driving poor clinical outcome.


Subject(s)
Caveolin 1/pharmacology , Fibroblasts/drug effects , Mitochondria/metabolism , Recombinant Proteins/pharmacology , Superoxide Dismutase/pharmacology , Tumor Microenvironment/drug effects , Animals , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Mitochondria/enzymology , Models, Biological , Neoplasms/genetics , Neoplasms/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Proteomics , RNA, Small Interfering , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
8.
Cell Cycle ; 9(17): 3515-33, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20855962

ABSTRACT

Recently, using a co-culture system, we demonstrated that MCF7 epithelial cancer cells induce oxidative stress in adjacent cancer-associated fibroblasts, resulting in the autophagic/lysosomal degradation of stromal caveolin-1 (Cav-1). However, the detailed signaling mechanism(s) underlying this process remain largely unknown. Here, we show that hypoxia is sufficient to induce the autophagic degradation of Cav-1 in stromal fibroblasts, which is blocked by the lysosomal inhibitor chloroquine. Concomitant with the hypoxia-induced degradation of Cav-1, we see the upregulation of a number of well-established autophagy/mitophagy markers, namely LC3, ATG16L, BNIP3, BNIP3L, HIF-1α and NFκB. In addition, pharmacological activation of HIF-1α drives Cav-1 degradation, while pharmacological inactivation of HIF-1 prevents the downregulation of Cav-1. Similarly, pharmacological inactivation of NFκB--another inducer of autophagy-prevents Cav-1 degradation. Moreover, treatment with an inhibitor of glutathione synthase, namely BSO, which induces oxidative stress via depletion of the reduced glutathione pool, is sufficient to induce the autophagic degradation of Cav-1. Thus, it appears that oxidative stress mediated induction of HIF1- and NFκB-activation in fibroblasts drives the autophagic degradation of Cav-1. In direct support of this hypothesis, we show that MCF7 cancer cells activate HIF-1α- and NFκB-driven luciferase reporters in adjacent cancer-associated fibroblasts, via a paracrine mechanism. Consistent with these findings, acute knock-down of Cav-1 in stromal fibroblasts, using an siRNA approach, is indeed sufficient to induce autophagy, with the upregulation of both lysosomal and mitophagy markers. How does the loss of stromal Cav-1 and the induction of stromal autophagy affect cancer cell survival? Interestingly, we show that a loss of Cav-1 in stromal fibroblasts protects adjacent cancer cells against apoptotic cell death. Thus, autophagic cancer-associated fibroblasts, in addition to providing recycled nutrients for cancer cell metabolism, also play a protective role in preventing the death of adjacent epithelial cancer cells. We demonstrate that cancer-associated fibroblasts upregulate the expression of TIGAR in adjacent epithelial cancer cells, thereby conferring resistance to apoptosis and autophagy. Finally, the mammary fat pads derived from Cav-1 (-/-) null mice show a hypoxia-like response in vivo, with the upregulation of autophagy markers, such as LC3 and BNIP3L. Taken together, our results provide direct support for the "Autophagic Tumor Stroma Model of Cancer Metabolism", and explain the exceptional prognostic value of a loss of stromal Cav-1 in cancer patients. Thus, a loss of stromal fibroblast Cav-1 is a biomarker for chronic hypoxia, oxidative stress and autophagy in the tumor microenvironment, consistent with its ability to predict early tumor recurrence, lymph node metastasis and tamoxifen-resistance in human breast cancers. Our results imply that cancer patients lacking stromal Cav-1 should benefit from HIF-inhibitors, NFκB-inhibitors, anti-oxidant therapies, as well as autophagy/lysosomal inhibitors. These complementary targeted therapies could be administered either individually or in combination, to prevent the onset of autophagy in the tumor stromal compartment, which results in a "lethal" tumor microenvironment.


Subject(s)
Autophagy , Breast Neoplasms/metabolism , Fibroblasts/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , NF-kappa B/metabolism , Stromal Cells/metabolism , Animals , Antirheumatic Agents/pharmacology , Apoptosis Regulatory Proteins , Breast Neoplasms/pathology , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Chloroquine/pharmacology , Coculture Techniques , Female , Glutathione Synthase/antagonists & inhibitors , Glutathione Synthase/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Oxidative Stress , Paracrine Communication , Phosphoric Monoester Hydrolases , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Tumor Microenvironment , Tumor Suppressor Proteins/metabolism , Up-Regulation
9.
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
10.
Cell Cycle ; 9(10): 1960-71, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20495363

ABSTRACT

We and others have previously identified a loss of stromal caveolin-1 (Cav-1) in cancer-associated fibroblasts (CAFs) as a powerful single independent predictor of breast cancer patient tumor recurrence, metastasis, tamoxifen-resistance, and poor clinical outcome. However, it remains unknown how loss of stromal Cav-1 mediates these effects clinically. To mechanistically address this issue, we have now generated a novel human tumor xenograft model. In this two-component system, nude mice are co-injected with i) human breast cancer cells (MDA-MB-231), and ii) stromal fibroblasts (wild-type (WT) versus Cav-1 (-/-) deficient). This allowed us to directly evaluate the effects of a Cav-1 deficiency solely in the tumor stromal compartment. Here, we show that Cav-1-deficient stromal fibroblasts are sufficient to promote both tumor growth and angiogenesis, and to recruit Cav-1 (+) micro-vascular cells. Proteomic analysis of Cav-1-deficient stromal fibroblasts indicates that these cells upregulate the expression of glycolytic enzymes, a hallmark of aerobic glycolysis (the Warburg effect). Thus, Cav-1-deficient stromal fibroblasts may contribute towards tumor growth and angiogenesis, by providing energy-rich metabolites in a paracrine fashion. We have previously termed this new idea the "Reverse Warburg Effect". In direct support of this notion, treatment of this xenograft model with glycolysis inhibitors functionally blocks the positive effects of Cav-1-deficient stromal fibroblasts on breast cancer tumor growth. Thus, pharmacologically-induced metabolic restriction (via treatment with glycolysis inhibitors) may be a promising new therapeutic strategy for breast cancer patients that lack stromal Cav-1 expression. We also identify the stromal expression of PKM2 and LDH-B as new candidate biomarkers for the "Reverse Warburg Effect" or "Stromal-Epithelial Metabolic Coupling" in human breast cancers.


Subject(s)
Caveolin 1/deficiency , Fibroblasts/metabolism , Glycolysis/drug effects , Lactate Dehydrogenases/metabolism , Pyruvate Kinase/metabolism , Animals , Blotting, Western , Caveolin 1/genetics , Cell Line, Tumor , Culture Media, Conditioned , Deoxyglucose/pharmacology , Dichloroacetic Acid/pharmacology , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Fibroblasts/cytology , Humans , Immunohistochemistry , Lactate Dehydrogenases/genetics , Mice , Mice, Nude , Microscopy, Fluorescence , Proteomics , Pyruvate Kinase/genetics , Xenograft Model Antitumor Assays
11.
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
12.
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
13.
J Cell Physiol ; 217(1): 281-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18506777

ABSTRACT

The insulin receptor substrate-1 (IRS-1), a docking protein of the type 1 insulin-like growth factor receptor (IGF-IR) plays a significant role in cell proliferation and differentiation. The expression of IRS-1 is down-regulated in mouse embryo fibroblasts (MEFs) with a deletion of caveolin-1 (cav1) genes (KO cells). Levels of IRS-1 mRNA are not affected. Re-introduction of cav1 into KO cells rescues IRS-1 expression. Stabilization of protein levels is reciprocal and a strict correlation between IRS-1 and cav1 levels was confirmed in five cell lines, and in mouse tissues. IRS-1 binds through its phosphotyrosine binding (PTB) domain to tyrosine 14 (Y14) of cav1, the residue phosphorylated by IGF-1 stimulation and by v-src. The down-regulation of IRS-1 in cav-/- cells occurs via the proteasome pathway. These results indicate a novel mechanism for the regulation of IRS-1 expression levels, an important finding in view of IRS-1 role in cell proliferation and transformation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Caveolin 1/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , Adaptor Proteins, Signal Transducing/genetics , Animals , Blotting, Western , Cell Differentiation/physiology , Cell Proliferation , Down-Regulation , Embryo, Mammalian , Fibroblasts/cytology , Gene Expression , Immunoprecipitation , Insulin Receptor Substrate Proteins , Mice , Mice, Knockout , RNA Interference , RNA, Messenger/analysis , RNA, Small Interfering , Transfection
14.
Behav Brain Res ; 188(2): 255-62, 2008 Apr 09.
Article in English | MEDLINE | ID: mdl-18083242

ABSTRACT

Neurological phenotypes associated with loss of caveolin 1 (cav-1) (the defining structural protein in caveolar vesicles, which regulate signal transduction and cholesterol trafficking in cells) in mice have been reported recently. In brain, cav-1 is highly expressed in neurons and glia. We investigated emotional and cognitive behavioural domains in mice deficient in cav-1 (CavKO mice). CavKO mice were more anxious and spent more time in self-directed grooming behaviour than wild-type (wt) mice. In a spatial/working memory task, CavKO mice failed to recognize the object displacement, thus showing a spatial memory impairment. CavKO mice showed higher locomotor activity than wt mice, thus suggesting reduced inhibitory function by CNS cholinergic systems. Behavioural response to the cholinergic muscarinic antagonist, scopolamine (2 mg/Kg), was decreased in CavKO mice. Few behavioural sex differences emerged in mice; whereas the sex differences were generally attenuated or even reverted in the null genotype. Our data confirm a distinct behavioural phenotype in CavKO mice and indicate a selective alteration in central cholinergic function.


Subject(s)
Acetylcholine/physiology , Caveolin 1/deficiency , Emotions/physiology , Memory Disorders/genetics , Memory, Short-Term/physiology , Analysis of Variance , Animals , Behavior, Animal/drug effects , Body Weight/drug effects , Body Weight/genetics , Cerebral Cortex/metabolism , Choline O-Acetyltransferase/metabolism , Exploratory Behavior/drug effects , Female , Male , Memory, Short-Term/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscarinic Antagonists/pharmacology , Scopolamine/pharmacology , Sex Factors
15.
Cancer Biol Ther ; 6(8): 1263-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17786030

ABSTRACT

Prostate cancer is the second leading cause of cancer-related deaths in men. Fatty acid synthase (FASN) is normally upregulated during human prostate cancer onset and metastatic progression and its expression positively correlates with the development of advanced metastatic disease. However, it remains unknown what molecular factor(s) control FASN expression. It has been hypothesized that FASN functions as a tumor promoter during prostate cancer progression in humans. Consistently, an established mouse of model of prostate cancer, termed TRAMP mice, also shows the progressive upregulation of FASN levels during prostate cancer development. Here, we examine the role of caveolin-1 (Cav-1) in regulating FASN expression during prostate cancer progression. For this purpose, we crossed Cav-1-/- null mice with TRAMP mice to generate TRAMP/Cav-1+/+ and TRAMP/Cav-1-/- mice. Then, we assessed the expression of FASN in Cav-1+/+ and Cav-1-/- prostate tumors by immuno-histochemistry and Western blot analysis. Interestingly, our results indicate that FASN fails to be upregulated in Cav-1-/- tumors. Importantly, the tumors examined were the same morphological grade, but Cav-1-/- tumors were dramatically smaller and did not metastasize efficiently. We conclude that Cav-1 expression is normally required for the upregulation of FASN during prostate cancer progression. These results also mechanistically explain why TRAMP/Cav-1-/- mice are dramatically resistant to the development of prostate tumors and lung metastases, as they lack the expression of the FASN tumor promoter. Thus, TRAMP/Cav-1-/- mice will provide a novel model system to elucidate the role of FASN in prostate tumor progression. In addition, our results provide the first molecular genetic evidence that Cav-1 functions upstream of FASN during prostate cancer progression.


Subject(s)
Caveolin 1/physiology , Fatty Acid Synthases/metabolism , Prostatic Neoplasms/pathology , Animals , Caveolin 1/genetics , Disease Progression , Fatty Acid Synthases/genetics , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Up-Regulation
16.
Cell Cycle ; 6(10): 1242-8, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17495527

ABSTRACT

Duchenne Muscular Dystrophy (DMD) is an incurable inherited disease of childhood, characterized by progressive muscle degeneration and weakness. Our previous findings supported the idea that dystrophin and associated proteins, absent or greatly reduced in DMD, are degraded in dystrophin-deficient muscle by the proteasomal-dependent pathway. Indeed, treatment with the proteasome inhibitor MG-132 of skeletal muscles from mdx mice--a spontaneous mouse model of DMD--as well as from DMD patients, effectively rescued the expression and correct cellular localization of dystrophin and associated proteins. These promising results led us to further explore the use of proteasome inhibitors as a therapy for DMD. Therefore, we directed our attention towards two new dipeptide boronic acid inhibitors blocking the proteasomal-dependent degradation pathway: Velcade (bortezomib or PS-341) and MLN273 (PS-273). The exciting aspect of this development is that these drugs have already progressed to preclinical and clinical trials, in different fields than muscular dystrophy. Indeed, Velcade has been already FDA-approved for treatment of multiple myeloma and its side effects had been already explored and managed. Promisingly, MLN273 is currently in the preclinical trial phase. Here, we test the effectiveness of Velcade and MLN273 by local injection into the gastrocnemius muscle of mdx mice. We show the rescue of expression and membrane localization of alpha-dystroglycan, beta-dystroglycan, alpha-sarcoglycan, and dystrophin after Velcade and MLN273 localized treatment, versus untreated (PBS only) mdx mice. Intriguingly, we also show that localized treatment with Velcade and MLN273 reduces the activation of Nuclear Factor-kappaB (NFkB). Because the NFkB pathway has been shown to be involved in inflammation responses in myopathies and DMD, our current results may have important clinical implications. Clearly, more investigations are needed, but our results emphasize the effectiveness of the pharmacological approach as a potential treatment for Duchenne muscular dystrophy.


Subject(s)
Boronic Acids/pharmacology , Dipeptides/pharmacology , Dystrophin-Associated Proteins/metabolism , Dystrophin/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Animals , Bortezomib , Male , Mice , Mice, Inbred mdx , Microscopy, Fluorescence , Muscular Dystrophy, Duchenne/drug therapy , NF-kappa B/metabolism
17.
Am J Pathol ; 170(1): 316-33, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17200204

ABSTRACT

Here, we examine the role of "non-muscle" caveolins (Cav-1 and Cav-2) in skeletal muscle biology. Our results indicate that skeletal muscle fibers from male Cav-1(-/-) and Cav-2(-/-) mice show striking abnormalities, such as tubular aggregates, mitochondrial proliferation/aggregation, and increased numbers of M-cadherin-positive satellite cells. Notably, these skeletal muscle defects were more pronounced with increasing age. Because Cav-2-deficient mice displayed normal expression levels of Cav-1, whereas Cav-1-null mice exhibited an almost complete deficiency in Cav-2, these skeletal muscle abnormalities seem to be due to loss of Cav-2. Thus, Cav-2(-/-) mice represent a novel animal model-and the first genetically well-defined mouse model-that can be used to study the pathogenesis of tubular aggregate formation, which remains a poorly understood age-related skeletal muscle abnormality. Finally, because Cav-1 and Cav-2 were not expressed within mature skeletal myofibers, our results indicate that development of these abnormalities probably originates in stem/precursor cells, such as satellite cells or myoblasts. Consistent with this hypothesis, skeletal muscle isolated from male Cav-3(-/-) mice did not show any of these abnormalities. As such, this is the first study linking stem cells with the genesis of these intriguing muscle defects.


Subject(s)
Caveolin 1/genetics , Caveolin 2/genetics , Mitochondria, Muscle , Muscle Fibers, Skeletal , Muscular Diseases/genetics , Animals , Cadherins/biosynthesis , Caveolin 1/deficiency , Caveolin 2/deficiency , Disease Models, Animal , Electron Transport Complex IV/analysis , Genetic Predisposition to Disease , Male , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/ultrastructure , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal/abnormalities , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Muscular Diseases/metabolism , Muscular Diseases/pathology , Myoblasts/metabolism , Myoblasts/pathology
18.
Am J Pathol ; 169(5): 1784-801, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17071600

ABSTRACT

Here, we investigate the role of caveolin-1 (Cav-1) in breast cancer onset and progression, with a focus on epithelial-stromal interactions, ie, the tumor microenvironment. Cav-1 is highly expressed in adipocytes and is abundant in mammary fat pads (stroma), but it remains unknown whether loss of Cav-1 within mammary stromal cells affects the differentiated state of mammary epithelia via paracrine signaling. To address this issue, we characterized the development of the mammary ductal system in Cav-1-/- mice and performed a series of mammary transplant studies, using both wild-type and Cav-1-/- mammary fat pads. Cav-1-/- mammary epithelia were hyperproliferative in vivo, with dramatic increases in terminal end bud area and mammary ductal thickness as well as increases in bromodeoxyuridine incorporation, extracellular signal-regulated kinase-1/2 hyperactivation, and up-regulation of STAT5a and cyclin D1. Consistent with these findings, loss of Cav-1 dramatically exacerbated mammary lobulo-alveolar hyperplasia in cyclin D1 Tg mice, whereas overexpression of Cav-1 caused reversion of this phenotype. Most importantly, Cav-1-/- mammary stromal cells (fat pads) promoted the growth of both normal mammary ductal epithelia and mammary tumor cells. Thus, Cav-1 expression in both epithelial and stromal cells provides a protective effect against mammary hyperplasia as well as mammary tumorigenesis.


Subject(s)
Caveolin 1/metabolism , Cyclin D1/antagonists & inhibitors , Epithelial Cells/cytology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/pathology , Protective Agents/metabolism , Stromal Cells/cytology , Adipose Tissue/cytology , Adipose Tissue/pathology , Animals , Caveolin 1/deficiency , Cell Proliferation , Cyclin D1/metabolism , Enzyme Activation , Female , Hyperplasia , Male , Mammary Neoplasms, Animal/chemically induced , Mammary Tumor Virus, Mouse , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phenotype , STAT5 Transcription Factor/metabolism , Signal Transduction
19.
Am J Physiol Cell Physiol ; 288(6): C1317-31, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15689413

ABSTRACT

Caveolin-3 (Cav-3) is expressed predominantly in skeletal muscle fibers, where it drives caveolae formation at the muscle cell's plasma membrane. In vitro studies have suggested that Cav-3 may play a positive role in insulin signaling and energy metabolism. We directly address the in vivo metabolic consequences of genetic ablation of Cav-3 in mice as it relates to insulin action, glucose metabolism, and lipid homeostasis. At age 2 mo, Cav-3 null mice are significantly larger than wild-type mice, and display significant postprandial hyperinsulinemia, whole body insulin resistance, and whole body glucose intolerance. Studies using hyperinsulinemic-euglycemic clamps revealed that Cav-3 null mice exhibited 20% and 40% decreases in insulin-stimulated whole body glucose uptake and whole body glycogen synthesis, respectively. Whole body insulin resistance was mostly attributed to 20% and 40% decreases in insulin-stimulated glucose uptake and glucose metabolic flux in the skeletal muscle of Cav-3 null mice. In addition, insulin-mediated suppression of hepatic glucose production was significantly reduced in Cav-3 null mice, indicating hepatic insulin resistance. Insulin-stimulated glucose uptake in white adipose tissue, which does not express Cav-3, was decreased by approximately 70% in Cav-3 null mice, suggestive of an insulin-resistant state for this tissue. During fasting, Cav-3 null mice possess normal insulin receptor protein levels in their skeletal muscle. However, after 15 min of acute insulin stimulation, Cav-3 null mice show dramatically reduced levels of the insulin receptor protein, compared with wild-type mice treated identically. These results suggest that Cav-3 normally functions to increase the stability of the insulin receptor at the plasma membrane, preventing its rapid degradation, i.e., by blocking or slowing ligand-induced receptor downregulation. Thus our results demonstrate the importance of Cav-3 in regulating whole body glucose homeostasis in vivo and its possible role in the development of insulin resistance. These findings may have clinical implications for the early diagnosis and treatment of caveolinopathies.


Subject(s)
Body Composition/physiology , Caveolins/physiology , Insulin Resistance/physiology , Muscle, Skeletal/physiology , Receptor, Insulin/physiology , Adipose Tissue/physiology , Animals , Blood Glucose/physiology , Caveolin 3 , Caveolins/genetics , Gene Expression , Glycogen/metabolism , Insulin/blood , Islets of Langerhans/pathology , Liver/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Receptor, Insulin/metabolism , Signal Transduction
20.
Am J Physiol Cell Physiol ; 288(3): C677-91, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15548572

ABSTRACT

It is generally well accepted that caveolin-3 expression is muscle specific, whereas caveolin-1 and -2 are coexpressed in a variety of cell types, including adipocytes, endothelial cells, epithelial cells, and fibroblasts. Caveolin-1 and -2 are known to form functional hetero-oligomeric complexes in cells where they are coexpressed, whereas caveolin-3 forms homo-oligomeric high molecular mass complexes. Although caveolin-2 might be expected to interact in a similar manner with caveolin-3, most studies indicate that this is not the case. However, this view has recently been challenged as it has been demonstrated that caveolin-2 and -3 are coexpressed in primary cultures of cardiac myocytes, where these two proteins can be coimmunoprecipitated. Thus it remains controversial whether caveolin-2 interacts with caveolin-3. Here, we directly address the issue of caveolin isoform protein-protein interactions by means of three distinct molecular genetic approaches. First, using caveolin-1-deficient mouse embryonic fibroblasts, in which we have stably expressed caveolin-1, -2, or -3, we find that caveolin-1 interacts with caveolin-2 in this setting, whereas caveolin-3 does not, in agreement with most published observations. Next, we used a transfected L6 myoblast cell system expressing all three caveolin proteins. Surprisingly, we found that caveolin-1, -2, and -3 all coimmunoprecipitate in this cell type, suggesting that this interaction is muscle cell specific. Similar results were obtained when the skeletal muscle of caveolin-1 transgenic animals was analyzed for caveolin-1 and caveolin-3 coimmunoprecipitation. Thus we conclude that all three caveolins can interact to form a discrete hetero-oligomeric complex, but that such complex formation is clearly muscle specific.


Subject(s)
Caveolins/metabolism , Fibroblasts/metabolism , Muscles/metabolism , Myoblasts/metabolism , Protein Isoforms/metabolism , Animals , Caveolae/chemistry , Caveolae/metabolism , Caveolins/genetics , Cell Line , Detergents/metabolism , Fibroblasts/ultrastructure , Macromolecular Substances , Mice , Mice, Knockout , Muscles/cytology , Myoblasts/ultrastructure , Octoxynol/metabolism , Protein Isoforms/genetics , Retroviridae/genetics , Retroviridae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...