37 results on '"Franco, Capozza"'
Search Results
2. Supplementary Figure Legends 1-2 from CAV1 Inhibits Metastatic Potential in Melanomas through Suppression of the Integrin/Src/FAK Signaling Pathway
- Author
-
Franco Capozza, Michael P. Lisanti, Federica Sotgia, Richard G. Pestell, Andrew E. Aplin, Kristin M. Daumer, Janet N. Milliman, Gloria Bonuccelli, Diana Whitaker-Menezes, and Casey Trimmer
- Abstract
Supplementary Figure Legends 1-2 from CAV1 Inhibits Metastatic Potential in Melanomas through Suppression of the Integrin/Src/FAK Signaling Pathway
- Published
- 2023
3. Supplementary Methods from CAV1 Inhibits Metastatic Potential in Melanomas through Suppression of the Integrin/Src/FAK Signaling Pathway
- Author
-
Franco Capozza, Michael P. Lisanti, Federica Sotgia, Richard G. Pestell, Andrew E. Aplin, Kristin M. Daumer, Janet N. Milliman, Gloria Bonuccelli, Diana Whitaker-Menezes, and Casey Trimmer
- Abstract
Supplementary Methods from CAV1 Inhibits Metastatic Potential in Melanomas through Suppression of the Integrin/Src/FAK Signaling Pathway
- Published
- 2023
4. Supplementary Figure Legend from Genetic Ablation of Cav1 Differentially Affects Melanoma Tumor Growth and Metastasis in Mice: Role of Cav1 in Shh Heterotypic Signaling and Transendothelial Migration
- Author
-
Michael P. Lisanti, Richard G. Pestell, Federica Sotgia, Gemma Llaverias, Marco Crosariol, Antonia Follenzi, Diana Whitaker-Menezes, Sanjay Katiyar, Remedios Castello-Cros, Casey Trimmer, and Franco Capozza
- Abstract
PDF file - 72K
- Published
- 2023
5. Data from CAV1 Inhibits Metastatic Potential in Melanomas through Suppression of the Integrin/Src/FAK Signaling Pathway
- Author
-
Franco Capozza, Michael P. Lisanti, Federica Sotgia, Richard G. Pestell, Andrew E. Aplin, Kristin M. Daumer, Janet N. Milliman, Gloria Bonuccelli, Diana Whitaker-Menezes, and Casey Trimmer
- 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 β3 in B16F10 melanoma cells. In summary, our findings provide experimental evidence that CAV1 may function as an antimetastatic gene in malignant melanoma. Cancer Res; 70(19); 7489–99. ©2010 AACR.
- Published
- 2023
6. Supplementary Tables 1-2, Figures 1-2 from CAV1 Inhibits Metastatic Potential in Melanomas through Suppression of the Integrin/Src/FAK Signaling Pathway
- Author
-
Franco Capozza, Michael P. Lisanti, Federica Sotgia, Richard G. Pestell, Andrew E. Aplin, Kristin M. Daumer, Janet N. Milliman, Gloria Bonuccelli, Diana Whitaker-Menezes, and Casey Trimmer
- Abstract
Supplementary Tables 1-2, Figures 1-2 from CAV1 Inhibits Metastatic Potential in Melanomas through Suppression of the Integrin/Src/FAK Signaling Pathway
- Published
- 2023
7. Genetic Ablation of Cav1 Differentially Affects Melanoma Tumor Growth and Metastasis in Mice: Role of Cav1 in Shh Heterotypic Signaling and Transendothelial Migration
- Author
-
Casey Trimmer, Franco Capozza, Federica Sotgia, Antonia Follenzi, Gemma Llaverias, Michael P. Lisanti, Diana Whitaker-Menezes, Richard G. Pestell, Remedios Castello-Cros, Sanjay Katiyar, and Marco Crosariol
- Subjects
Scaffold protein ,Cancer Research ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,Caveolin 2 ,medicine.medical_treatment ,Caveolin 1 ,Melanoma, Experimental ,Mice, Nude ,Cell Growth Processes ,Biology ,Article ,Metastasis ,Mice ,Paracrine signalling ,Cell Movement ,Human Umbilical Vein Endothelial Cells ,medicine ,Animals ,Humans ,Hedgehog Proteins ,Neoplasm Metastasis ,neoplasms ,Mice, Knockout ,Melanoma ,Cancer ,Fibroblasts ,medicine.disease ,Primary tumor ,Coculture Techniques ,Mice, Inbred C57BL ,Cytokine ,Oncology ,Cancer research ,Cytokines ,Female - 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. Cancer Res; 72(9); 2262–74. ©2012 AACR.
- Published
- 2012
8. CAV1 Inhibits Metastatic Potential in Melanomas through Suppression of the Integrin/Src/FAK Signaling Pathway
- Author
-
Kristin M. Daumer, Casey Trimmer, Franco Capozza, Andrew E. Aplin, Richard G. Pestell, Janet N. Milliman, Federica Sotgia, Gloria Bonuccelli, Michael P. Lisanti, and Diana Whitaker-Menezes
- Subjects
Cancer Research ,Lung Neoplasms ,Caveolin 1 ,Integrin ,Melanoma, Experimental ,Cell Growth Processes ,Article ,Metastasis ,Focal adhesion ,Mice ,Cell Movement ,Cell Line, Tumor ,Caveolae ,medicine ,Animals ,Humans ,Melanoma ,biology ,business.industry ,Cell Membrane ,medicine.disease ,Mice, Inbred C57BL ,src-Family Kinases ,Oncology ,Cell culture ,Focal Adhesion Protein-Tyrosine Kinases ,Cancer research ,biology.protein ,Melanocytes ,Female ,Signal transduction ,business ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src - 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 β3 in B16F10 melanoma cells. In summary, our findings provide experimental evidence that CAV1 may function as an antimetastatic gene in malignant melanoma. Cancer Res; 70(19); 7489–99. ©2010 AACR.
- Published
- 2010
9. The reverse Warburg Effect: Glycolysis inhibitors prevent the tumor promoting effects of caveolin-1 deficient cancer associated fibroblasts
- Author
-
Alessandro Fatatis, Barbara Chiavarina, Agnieszka K. Witkiewicz, Franco Capozza, Federica Sotgia, Diana Whitaker-Menezes, Matthew G. Vander Heiden, Michael P. Lisanti, Neal Flomenberg, Gloria Bonuccelli, Stephanos Pavlides, Richard G. Pestell, Remedios Castello-Cros, Gemma Migneco, Ubaldo E. Martinez-Outschoorn, and Philippe G. Frank
- Subjects
Proteomics ,medicine.medical_specialty ,Stromal cell ,Angiogenesis ,Blotting, Western ,Caveolin 1 ,Pyruvate Kinase ,Mice, Nude ,Deoxyglucose ,Biology ,PKM2 ,Metastasis ,Mice ,Cell Line, Tumor ,Internal medicine ,medicine ,Animals ,Humans ,Electrophoresis, Gel, Two-Dimensional ,Reverse Warburg effect ,Lactate Dehydrogenases ,Molecular Biology ,Dichloroacetic Acid ,Cell Biology ,Fibroblasts ,medicine.disease ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Warburg effect ,Endocrinology ,Microscopy, Fluorescence ,Anaerobic glycolysis ,Culture Media, Conditioned ,cardiovascular system ,Cancer research ,Cancer-Associated Fibroblasts ,Electrophoresis, Polyacrylamide Gel ,Glycolysis ,Developmental Biology - 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.
- Published
- 2010
10. Regulation of insulin receptor substrate-1 expression levels by caveolin-1
- Author
-
Tiziana DeAngelis, Jia Chen, An Wu, Renato Baserga, Michael P. Lisanti, Franco Capozza, and Hongzhi Sun
- Subjects
Phosphotyrosine binding ,Insulin Receptor Substrate Proteins ,Physiology ,Blotting, Western ,Caveolin 1 ,Clinical Biochemistry ,Down-Regulation ,Gene Expression ,Transfection ,Mice ,Growth factor receptor ,Insulin receptor substrate ,Animals ,Immunoprecipitation ,RNA, Messenger ,RNA, Small Interfering ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Insulin-like growth factor 1 receptor ,Mice, Knockout ,biology ,Cell Differentiation ,Cell Biology ,Fibroblasts ,Embryo, Mammalian ,IRS2 ,IRS1 ,Cell biology ,Insulin receptor ,Gene Expression Regulation ,biology.protein ,RNA Interference - 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.
- Published
- 2008
11. Stromal and Epithelial Caveolin-1 Both Confer a Protective Effect Against Mammary Hyperplasia and Tumorigenesis
- Author
-
Michael P. Lisanti, Terence M. Williams, Dolores Di Vizio, Hyangkyu Lee, Hallgeir Rui, Richard G. Pestell, Gloria Bonuccelli, Franco Capozza, Federica Sotgia, Isabelle Mercier, and Ghada S. Hassan
- Subjects
Mammary tumor ,Tumor microenvironment ,Stromal cell ,Mammary gland ,Hyperplasia ,Biology ,medicine.disease ,Pathology and Forensic Medicine ,Paracrine signalling ,medicine.anatomical_structure ,Cyclin D1 ,Caveolin 1 ,cardiovascular system ,medicine ,Cancer research - 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.
- Published
- 2006
12. Muscle-specific interaction of caveolin isoforms: differential complex formation between caveolins in fibroblastic vs. muscle cells
- Author
-
Federica Sotgia, Philippe G. Frank, Alex W. Cohen, Michela Battista, Michael P. Lisanti, William Schubert, Hyangkyu Lee, Michelle W.-C. Cheung, and Franco Capozza
- Subjects
Cell type ,Macromolecular Substances ,Octoxynol ,Physiology ,Detergents ,Biology ,Caveolae ,Caveolins ,Cell Line ,Myoblasts ,Mice ,Caveolin ,medicine ,Animals ,Protein Isoforms ,Myocyte ,Mice, Knockout ,Muscles ,Skeletal muscle ,Cell Biology ,Fibroblasts ,Molecular biology ,Cell biology ,Caveolin 3 ,Caveolin 2 ,Retroviridae ,medicine.anatomical_structure ,Caveolin 1 - 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.
- Published
- 2005
13. Tyrosine Phosphorylation of Caveolin-2 at Residue 27: Differences in the Spatial and Temporal Behavior of Phospho-Cav-2 (pY19 and pY27)
- Author
-
Hyangkyu Lee, Shana Marmon, Michael P. Lisanti, James W. Brooks, Roberto Campos-Gonzalez, Xiaobo Wang, Franco Capozza, and Federica Sotgia
- Subjects
Caveolin 2 ,Proto-Oncogene Proteins pp60(c-src) ,Protein tyrosine phosphatase ,Transfection ,SH2 domain ,Caveolins ,Biochemistry ,Receptor tyrosine kinase ,src Homology Domains ,Mice ,chemistry.chemical_compound ,Membrane Microdomains ,Antibody Specificity ,Cell Line, Tumor ,Animals ,Humans ,Protein phosphorylation ,Phosphorylation ,Tyrosine ,Phosphotyrosine ,Adaptor Proteins, Signal Transducing ,Cell Line, Transformed ,Oncogene Proteins ,Alanine ,Epidermal Growth Factor ,biology ,Antibodies, Monoclonal ,Tyrosine phosphorylation ,Molecular biology ,Molecular Weight ,Amino Acid Substitution ,chemistry ,ras GTPase-Activating Proteins ,COS Cells ,NIH 3T3 Cells ,cardiovascular system ,biology.protein ,Binding Sites, Antibody ,Protein Binding ,Subcellular Fractions - Abstract
Caveolin-2 is an accessory molecule and the binding partner of caveolin-1. Previously, we showed that c-Src expression leads to the tyrosine phosphorylation of Cav-2 at position 19. To further investigate the tyrosine phosphorylation of Cav-2, we have now generated a novel phospho-specific antibody directed against phospho-Cav-2 (pY27). Here, we show that Cav-2 is phosphorylated at both tyrosines 19 and 27. We reconstituted this phosphorylation event by recombinantly coexpressing c-Src and Cav-2. We generated a series of Cav-2 constructs harboring the mutation of each tyrosine to alanine, singly or in combination, i.e., Cav-2 Y19A, Y27A, and Y19A/Y27A. Recombinant expression of these mutants in Cos-7 cells demonstrated that neither tyrosine is the unique phosphorylation site, and that double mutation of tyrosines 19 and 27 to alanine abrogates Cav-2 tyrosine phosphorylation. Immunofluorescence analysis of NIH 3T3 cells revealed that the two tyrosine-phosphorylated forms of Cav-2 exhibited some distinct properties. Phospho-Cav-2 (pY19) is concentrated at cell edges and at cell-cell contacts, whereas phospho-Cav-2 (pY27) is distributed in a dotlike pattern throughout the cell surface and cytoplasm. Further functional analysis revealed that tyrosine phosphorylation of Cav-2 has no effect on its targeting to lipid rafts, but clearly disrupts the hetero-oligomerization of Cav-2 with Cav-1. In an attempt to identify upstream mediators, we investigated Cav-2 tyrosine phosphorylation in an endogenous setting. We found that in A431 cells, EGF stimulation is sufficient to induce Cav-2 phosphorylation at tyrosines 19 and 27. However, the behavior of the two phosphorylated forms of Cav-2 diverges upon EGF stimulation. First, phospho-Cav-2 (pY19) and phospho-Cav-2 (pY27) display different localization patterns. In addition, the temporal response to EGF stimulation appears to be different. Cav-2 is phosphorylated at tyrosine 19 in a rapid and transient fashion, whereas phosphorylation at tyrosine 27 is sustained over time. Three SH2 domain-containing proteins, c-Src, Nck, and Ras-GAP, were found to associate with Cav-2 in a phosphorylation-dependent manner. However, phosphorylation at tyrosine 27 appears to be more critical than phosphorylation at tyrosine 19 for this binding to occur. Taken together, these results suggest that, in addition to the common characteristics that these two sites appear to share, phospho-Cav-2 (pY19) and phospho-Cav-2 (pY27) may each possess a set of unique functional roles.
- Published
- 2004
14. Phosphofructokinase Muscle-Specific Isoform Requires Caveolin-3 Expression for Plasma Membrane Recruitment and Caveolar Targeting
- Author
-
Michael P. Lisanti, Carlo Minetti, Federica Sotgia, Scott Eric Woodman, Franco Capozza, Philipp E. Scherer, Robert G. Kemp, and Gloria Bonuccelli
- Subjects
Biology ,Subcellular localization ,medicine.disease ,Pathology and Forensic Medicine ,Cell biology ,Cell membrane ,Caveolin 3 ,medicine.anatomical_structure ,Biochemistry ,Caveolae ,Caveolin 1 ,Caveolin ,cardiovascular system ,medicine ,medicine.symptom ,Muscular dystrophy ,Myopathy - Abstract
Previous co-immunoprecipitation studies have shown that endogenous PFK-M (phosphofructokinase, muscle-specific isoform) associates with caveolin (Cav)-3 under certain metabolic conditions. However, it remains unknown whether Cav-3 expression is required for the plasma membrane recruitment and caveolar targeting of PFK-M. Here, we demonstrate that recombinant expression of Cav-3 dramatically affects the subcellular localization of PFK-M, by targeting PFK-M to the plasma membrane, and by trans-locating PFK-M to caveolae-enriched membrane domains. In addition, we show that the membrane recruitment and caveolar targeting of PFK-M appears to be strictly dependent on the concentration of extracellular glucose. Interestingly, recombinant expression of PFK-M with three Cav-3 mutants [ΔTFT (63 to 65), P104L, and R26Q], which harbor the same mutations as seen in the human patients with Cav-3-related muscle diseases, causes a substantial reduction in PFK-M expression levels, and impedes the membrane recruitment of PFK-M. Analysis of skeletal muscle tissue samples from Cav-3(−/−) mice directly demonstrates that Cav-3 expression regulates the phenotypic behavior of PFK-M. More specifically, in Cav-3-null mice, PFK-M is no longer targeted to the plasma membrane, and is excluded from caveolar membrane domains. As such, our current results may be important in understanding the pathogenesis of Cav-3-related muscle diseases, such as limb-girdle muscular dystrophy-1C, distal myopathy, and rippling muscle disease, that are caused by mutations within the human Cav-3 gene.
- Published
- 2003
15. Phenotypic behavior of caveolin-3 R26Q, a mutant associated with hyperCKemia, distal myopathy, and rippling muscle disease
- Author
-
Scott Eric Woodman, Federica Sotgia, Carlo Minetti, Philipp E. Scherer, Michael P. Lisanti, Gloria Bonuccelli, and Franco Capozza
- Subjects
medicine.medical_specialty ,Caveolin 3 ,Physiology ,Glutamine ,Mutant ,Muscle Proteins ,Limb girdle ,Biology ,Arginine ,medicine.disease_cause ,Caveolins ,Muscular Dystrophies ,Mice ,Muscular Diseases ,Internal medicine ,Chlorocebus aethiops ,Caveolin ,medicine ,Animals ,Muscular dystrophy ,Myopathy ,Creatine Kinase ,Mutation ,Cell Biology ,medicine.disease ,Phenotype ,Endocrinology ,Amino Acid Substitution ,Gene Expression Regulation ,COS Cells ,NIH 3T3 Cells ,medicine.symptom - Abstract
Four different phenotypes have been associated with CAV3 mutations: limb girdle muscular dystrophy-1C (LGMD-1C), rippling muscle disease (RMD), and distal myopathy (DM), as well as idiopathic and familial hyperCKemia (HCK). Detailed molecular characterization of two caveolin-3 mutations (P104L and ΔTFT), associated with LGMD-1C, shows them to impart a dominant-negative effect on wild-type caveolin-3, rendering it dysfunctional through sequestration in the Golgi complex. Interestingly, substitution of glutamine for arginine at amino acid position 26 (R26Q) of caveolin-3 is associated not only with RMD but also with DM and HCK. However, the phenotypic behavior of the caveolin-3 R26Q mutation has never been evaluated in cultured cells. Thus we characterized the cellular and molecular properties of the R26Q mutant protein to better understand how this mutation can manifest as such distinct disease phenotypes. Here, we show that the caveolin-3 R26Q mutant is mostly retained at the level of the Golgi complex. The caveolin-3 R26Q mutant formed oligomers of a much larger size than wild-type caveolin-3 and was excluded from caveolae-enriched membranes. However, caveolin-3 R26Q did not behave in a dominant-negative fashion when coexpressed with wild-type caveolin-3. Thus the R26Q mutation behaves differently from other caveolin-3 mutations (P104L and ΔTFT) that have been previously characterized. These data provide a possible explanation for the scope of the various disease phenotypes associated with the caveolin-3 R26Q mutation. We propose a haploinsufficiency model in which reduced levels of wild-type caveolin-3, although not rendered dysfunctional due to the caveolin-3 R26Q mutant protein, are insufficient for normal muscle cell function.
- Published
- 2003
16. Identification of Novel mRNA Transcripts of thenm23-M1Gene that Are Modulated during Mouse Embryo Development and Are Differently Expressed in Adult Murine Tissues
- Author
-
Franco Capozza, Fabio Gervasi, Tiziana Bruno, Maurizio Fanciulli, and Daniela Lombardi
- Subjects
Protein Folding ,DNA, Complementary ,Molecular Sequence Data ,Biology ,Embryonic and Fetal Development ,Mice ,Genetics ,Homologous chromosome ,Animals ,RNA, Messenger ,Molecular Biology ,Gene ,Monomeric GTP-Binding Proteins ,Messenger RNA ,Base Sequence ,Cell growth ,Embryogenesis ,Gene Expression Regulation, Developmental ,Cell Biology ,General Medicine ,NM23 Nucleoside Diphosphate Kinases ,Molecular biology ,In vitro ,Nucleoside-Diphosphate Kinase ,Nucleic Acid Conformation ,Identification (biology) ,Transcription Factors - Abstract
The nm23-M1, a putative metastasis-suppressor gene, and its homologs are involved in development and differentiation. We have shown previously that in vitro neuronal cell proliferation and differentiation can be modulated by nm23-M1 expression levels. In the present study, by the yeast two-hybrid system, we have shown that, at the onset of mouse tissue differentiation, the Nm23-M1 protein forms either homodimers, or heterodimers with Nm23-M2. Furthermore, we have isolated two cDNA variants of the nm23-M1 gene in the 3'-untranslated region (UTR). The two variants related to novel mRNA transcripts that are modulated in mouse embryo and are differently expressed in adult murine tissues.
- Published
- 1998
17. Cav1 inhibits benign skin tumor development in a two-stage carcinogenesis model by suppressing epidermal proliferation
- Author
-
Casey, Trimmer, Federica, Sotgia, Michael P, Lisanti, and Franco, Capozza
- Subjects
Original Article - 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.
- Published
- 2012
18. The milk protein α-casein functions as a tumor suppressor via activation of STAT1 signaling, effectively preventing breast cancer tumor growth and metastasis
- Author
-
Diana Whitaker-Menezes, Jiao Xuanmao, Ubaldo E. Martinez-Outschoorn, Gloria Bonuccelli, Federica Sotgia, Zhao Lin, Anthony Howell, Franco Capozza, Aristotelis Tsirigos, Michael P. Lisanti, and Remedios Castello-Cros
- Subjects
Apoptosis ,Breast Neoplasms ,Metastasis ,Cell Line ,Mice ,Breast cancer ,Downregulation and upregulation ,Interferon ,Cancer stem cell ,Cell Movement ,Report ,medicine ,Animals ,Humans ,Molecular Biology ,Cell Proliferation ,Mammary tumor ,biology ,Milk, Human ,Gene Expression Profiling ,CD44 ,Caseins ,Cell Biology ,medicine.disease ,Up-Regulation ,Gene expression profiling ,Hyaluronan Receptors ,STAT1 Transcription Factor ,Cancer research ,biology.protein ,Neoplastic Stem Cells ,Female ,Interferons ,Developmental Biology ,medicine.drug ,Signal Transduction - 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.
- Published
- 2012
19. Cav1 suppresses tumor growth and metastasis in a murine model of cutaneous SCC through modulation of MAPK/AP-1 activation
- Author
-
Richard G. Pestell, Sanjay Katiyar, Gloria Bonuccelli, Federica Sotgia, Michael P. Lisanti, Casey Trimmer, and Franco Capozza
- Subjects
MAPK/ERK pathway ,Keratinocytes ,Serum ,Cell signaling ,Skin Neoplasms ,MAP Kinase Signaling System ,Caveolin 1 ,Mice, Nude ,Biology ,Models, Biological ,Pathology and Forensic Medicine ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cyclin D1 ,Epidermal growth factor ,Cell Movement ,Cell Line, Tumor ,Animals ,Humans ,Neoplasm Invasiveness ,Neoplasm Metastasis ,Protein kinase A ,030304 developmental biology ,Cell Proliferation ,0303 health sciences ,Gene knockdown ,Mice, Inbred BALB C ,Epidermal Growth Factor ,Keratin-18 ,Activator (genetics) ,Cell growth ,Regular Article ,3. Good health ,Enzyme Activation ,Transcription Factor AP-1 ,Disease Models, Animal ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Cancer research ,Carcinoma, Squamous Cell ,Mitogen-Activated Protein Kinases - 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.
- Published
- 2012
20. Eating for two: how stromal fibroblasts might nurture adjacent carcinoma cells
- Author
-
Diana Whitaker-Menezes, Michael P. Lisanti, Alex Molchansky, Remedios Castello-Cros, Agnieszka K. Witkiewicz, Gloria Bonuccelli, Richard G. Pestell, Federica Sotgia, Ruth Birbe, Anthony Howell, and Franco Capozza
- Subjects
Stromal cell ,Angiogenesis ,Caveolin 1 ,Transplantation, Heterologous ,Breast Neoplasms ,Biology ,Metastasis ,Extracellular matrix ,Mice ,Cell Line, Tumor ,Neoplasms ,Report ,Plasminogen Activator Inhibitor 1 ,Plasminogen Activator Inhibitor 2 ,Tumor Microenvironment ,medicine ,Autophagy ,Animals ,Humans ,Neoplasm Metastasis ,Fibroblast ,Molecular Biology ,Tumor microenvironment ,Cell Biology ,Fibroblasts ,medicine.disease ,Coculture Techniques ,Cell biology ,Extracellular Matrix ,Mitochondria ,medicine.anatomical_structure ,Cancer cell ,Cancer-Associated Fibroblasts ,Stromal Cells ,Developmental Biology - 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.
- Published
- 2011
21. Caveolin-1 and mitochondrial SOD2 (MnSOD) function as tumor suppressors in the stromal microenvironment: A new genetically tractable model for human cancer associated fibroblasts
- Author
-
Gregory J. Eaton, Casey Trimmer, Anthony Howell, Franco Capozza, Renato V. Iozzo, Richard G. Pestell, Federica Sotgia, Ubaldo E. Martinez-Outschoorn, Michael P. Lisanti, Stephanos Pavlides, Diana Whitaker-Menezes, Renee M. Balliet, and Philipp E. Scherer
- Subjects
Proteomics ,Cancer Research ,Stromal cell ,Nitric Oxide Synthase Type III ,SOD1 ,Caveolin 1 ,SOD2 ,Mice, Nude ,Mitochondrion ,Biology ,Models, Biological ,03 medical and health sciences ,Paracrine signalling ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,Tumor Microenvironment ,Animals ,Humans ,RNA, Small Interfering ,030304 developmental biology ,Cell Line, Transformed ,Cell Proliferation ,Pharmacology ,0303 health sciences ,Tumor microenvironment ,Cell growth ,Superoxide Dismutase ,Fibroblasts ,Xenograft Model Antitumor Assays ,Recombinant Proteins ,Cell biology ,Mitochondria ,Gene Expression Regulation, Neoplastic ,Oxidative Stress ,Oncology ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,cardiovascular system ,Molecular Medicine ,Female ,Research Paper - 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 overexpression 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 cancerassociated 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-positive breast cancers, which are all associated with a Cav-1 deficient “lethal” tumor microenvironment, driving poor clinical outcome.
- Published
- 2011
22. Autophagy in cancer associated fibroblasts promotes tumor cell survival: Role of hypoxia, HIF1 induction and NFκB activation in the tumor stromal microenvironment
- Author
-
Agnieszka K. Witkiewicz, Ubaldo E. Martinez-Outschoorn, Stephanos Pavlides, Jie Zhou, Barbara Chiavarina, Michael P. Lisanti, Chengwang Wang, Richard G. Pestell, Zhao Lin, Federica Sotgia, Maria P. Martinez-Cantarin, Diana Whitaker-Menezes, Neal Flomenberg, Casey Trimmer, Anthony Howell, Franco Capozza, and Jaime Caro
- Subjects
Caveolin 1 ,Mice ,Caveolin-1 ,Mitophagy ,Tumor Microenvironment ,LC3 ,Reverse Warburg effect ,RNA, Small Interfering ,Hypoxia ,Mice, Knockout ,Intracellular Signaling Peptides and Proteins ,NF-kappa B ,Chloroquine ,Cell Hypoxia ,Cancer associated fibroblasts ,3. Good health ,Cell biology ,Up-Regulation ,Predictive biomarker ,Antirheumatic Agents ,cardiovascular system ,Female ,RNA Interference ,Microtubule-Associated Proteins ,Stromal cell ,BNIP3 ,Cell Survival ,HIF1 ,Breast Neoplasms ,TIGAR ,Biology ,Glutathione Synthase ,Downregulation and upregulation ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Report ,Paracrine Communication ,Autophagy ,Animals ,Humans ,Tumor stroma ,Molecular Biology ,Tumor microenvironment ,Tumor Suppressor Proteins ,Membrane Proteins ,Cell Biology ,Fibroblasts ,Hypoxia-Inducible Factor 1, alpha Subunit ,Coculture Techniques ,Phosphoric Monoester Hydrolases ,Oxidative Stress ,Oxidative stress ,Cancer cell ,Cancer-Associated Fibroblasts ,Stromal Cells ,Apoptosis Regulatory Proteins ,NFκB ,Developmental Biology - Abstract
Recently, using a co-culture system, we demonstrated that MCF7 epithelial cancer cells induce oxidative stress in adjacent cancerassociated 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. © 2010 Landes Bioscience.
- Published
- 2010
23. Caveolin-1 (P132L), a Common Breast Cancer Mutation, Confers Mammary Cell Invasiveness and Defines a Novel Stem Cell/Metastasis-Associated Gene Signature
- Author
-
Janet N. Milliman, Federica Sotgia, Richard G. Pestell, Kristin M. Daumer, Philippe G. Frank, Gloria Bonuccelli, Carlo Minetti, Jie Zhou, Isabelle Mercier, Fabien Alpy, Catherine Tomasetto, Franco Capozza, Manran Liu, Chenguang Wang, Michael P. Lisanti, Sanjay Katiyar, Elliott Dew, Marie Christine Rio, Neal Flomenberg, Mathew C. Casimiro, Peney, Maité, Sidney Kimmel Cancer Center, Jefferson (Philadelphia University + Thomas Jefferson University), Muscular and Neurodegenerative Disease Unit, Università degli studi di Genova = University of Genoa (UniGe), Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), University of Genoa (UNIGE), and Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Strasbourg (UNISTRA)
- Subjects
Blotting, Western ,Caveolin 1 ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Mammary Neoplasms, Animal ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,Metastasis ,Immunoenzyme Techniques ,03 medical and health sciences ,Mice ,0302 clinical medicine ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Cell Movement ,medicine ,Biomarkers, Tumor ,Animals ,Humans ,Neoplasm Invasiveness ,Progenitor cell ,Mammary Glands, Human ,030304 developmental biology ,Cell Proliferation ,Oligonucleotide Array Sequence Analysis ,0303 health sciences ,Mutation ,Cell growth ,Gene Expression Profiling ,Estrogen Receptor alpha ,Cell migration ,medicine.disease ,Prognosis ,3. Good health ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,CYR61 ,Cancer research ,cardiovascular system ,Neoplastic Stem Cells ,Female ,Stem cell ,Regular Articles ,Signal Transduction - Abstract
International audience; 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.
- Published
- 2009
24. Caveolin‐3 KO Mice Develop Dyslipidemia with Impaired Substrate Utilization in Skeletal Muscle
- Author
-
Casey Trimmer, Franco Capozza, and Michael P. Lisanti
- Subjects
medicine.medical_specialty ,Chemistry ,Substrate (chemistry) ,Skeletal muscle ,medicine.disease ,Biochemistry ,Caveolin 3 ,medicine.anatomical_structure ,Endocrinology ,Internal medicine ,Genetics ,medicine ,Molecular Biology ,Dyslipidemia ,Biotechnology - Published
- 2009
25. Loss of caveolin-3 induces a lactogenic microenvironment that is protective against mammary tumor formation
- Author
-
Mathew C. Casimiro, Federica Sotgia, Carlo Minetti, Diana Whitaker-Menezes, Erik S. Knudsen, Ozlem Er, Michael Gormley, Andrew A. Quong, Philippe G. Frank, Richard G. Pestell, Franco Capozza, Janet N. Milliman, Hallgeir Rui, Michael P. Lisanti, Chenguang Wang, Kristin M. Daumer, Jie Zhou, Gloria Bonuccelli, Agnieszka K. Witkiewicz, Isabelle Mercier, and Manran Liu
- Subjects
Male ,medicine.medical_specialty ,Caveolin 3 ,Mammary gland ,Gene Expression ,Biology ,medicine.disease_cause ,Polymerase Chain Reaction ,Pathology and Forensic Medicine ,Mice ,Mammary Glands, Animal ,Cell Movement ,Pregnancy ,Internal medicine ,Lactation ,Gene expression ,medicine ,In Situ Nick-End Labeling ,Animals ,Humans ,Oligonucleotide Array Sequence Analysis ,Mammary tumor ,Milk, Human ,Gene Expression Profiling ,Myoepithelial cell ,Mammary Neoplasms, Experimental ,Hyperplasia ,medicine.disease ,Immunohistochemistry ,Mice, Mutant Strains ,medicine.anatomical_structure ,Endocrinology ,Phenotype ,Cancer research ,cardiovascular system ,Female ,Stem cell ,Carcinogenesis ,Regular Articles - 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.
- Published
- 2009
26. Altered emotionality, spatial memory and cholinergic function in caveolin-1 knock-out mice
- Author
-
Philippe G. Frank, Laura Ricceri, Laura Gioiosa, Carla Raggi, Jean-Francois Jasmin, Franco Capozza, Enrico Alleva, Michael P. Lisanti, Massimo Sargiacomo, and Giovanni Laviola
- Subjects
Male ,medicine.medical_specialty ,Caveolin 1 ,Emotions ,Scopolamine ,Muscarinic Antagonists ,Inhibitory postsynaptic potential ,Choline O-Acetyltransferase ,Behavioral Neuroscience ,Mice ,Sex Factors ,Internal medicine ,Caveolin ,medicine ,Animals ,Memory disorder ,Cerebral Cortex ,Mice, Knockout ,Analysis of Variance ,Memory Disorders ,Behavior, Animal ,Body Weight ,Muscarinic antagonist ,medicine.disease ,Acetylcholine ,Mice, Inbred C57BL ,Endocrinology ,Memory, Short-Term ,Knockout mouse ,Exploratory Behavior ,Cholinergic ,Female ,Signal transduction ,Psychology ,Neuroscience ,medicine.drug - 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.
- Published
- 2007
27. Caveolin-1 is required for the upregulation of fatty acid synthase (FASN), a tumor promoter, during prostate cancer progression
- Author
-
Philippe G. Frank, Federica Sotgia, Ghada S. Hassan, Franco Capozza, Dolores Di Vizio, Richard G. Pestell, Michael R. Freeman, Massimo Loda, Terence M. Williams, and Michael P. Lisanti
- Subjects
Oncology ,PCA3 ,Male ,Cancer Research ,medicine.medical_specialty ,Caveolin 1 ,Mice, Transgenic ,Biology ,Metastasis ,Prostate cancer ,Mice ,Prostate ,Internal medicine ,medicine ,Animals ,Pharmacology ,Cancer ,Prostatic Neoplasms ,medicine.disease ,Up-Regulation ,Fatty acid synthase ,medicine.anatomical_structure ,Tumor progression ,cardiovascular system ,Cancer research ,biology.protein ,Disease Progression ,Molecular Medicine ,Fatty Acid Synthases ,Tramp - 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.
- Published
- 2007
28. Caveolin-1(-/-)- and caveolin-2(-/-)-deficient mice both display numerous skeletal muscle abnormalities, with tubular aggregate formation
- Author
-
Franco Capozza, William Schubert, Michael P. Lisanti, Alex W. Cohen, Carlo Minetti, Federica Sotgia, Salvatore DiMauro, Eduardo Bonilla, Claudio Bruno, and Gloria Bonuccelli
- Subjects
Male ,medicine.medical_specialty ,Caveolin 2 ,Caveolin 1 ,Muscle Fibers, Skeletal ,Biology ,Mitochondrion ,Pathology and Forensic Medicine ,Pathogenesis ,Electron Transport Complex IV ,Myoblasts ,Mice ,Microscopy, Electron, Transmission ,Muscular Diseases ,Precursor cell ,Internal medicine ,medicine ,Myocyte ,Animals ,Genetic Predisposition to Disease ,Muscle, Skeletal ,Mice, Knockout ,Skeletal muscle ,Cadherins ,Mitochondria, Muscle ,Disease Models, Animal ,Endocrinology ,medicine.anatomical_structure ,cardiovascular system ,Stem cell ,Regular Articles - 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.
- Published
- 2007
29. Localized treatment with a novel FDA-approved proteasome inhibitor blocks the degradation of dystrophin and dystrophin-associated proteins in mdx mice
- Author
-
Michael P. Lisanti, Gloria Bonuccelli, Franco Capozza, Federica Sotgia, Elisabetta Gazzerro, and Carlo Minetti
- Subjects
musculoskeletal diseases ,Male ,congenital, hereditary, and neonatal diseases and abnormalities ,Weakness ,Duchenne muscular dystrophy ,Bortezomib ,Dystrophin ,Mice ,medicine ,Animals ,Protease Inhibitors ,Muscular dystrophy ,Muscle, Skeletal ,Molecular Biology ,Cellular localization ,biology ,NF-kappa B ,Cell Biology ,Dipeptides ,medicine.disease ,Molecular biology ,Boronic Acids ,Dystrophin-associated protein ,Muscular Dystrophy, Duchenne ,Microscopy, Fluorescence ,Pyrazines ,Dystrophin-Associated Proteins ,biology.protein ,Cancer research ,Proteasome inhibitor ,Mice, Inbred mdx ,medicine.symptom ,Developmental Biology ,medicine.drug - 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.
- Published
- 2007
30. Caveolin-3 knockout mice show increased adiposity and whole body insulin resistance, with ligand-induced insulin receptor instability in skeletal muscle
- Author
-
Jason K. Kim, Terry P. Combs, Terence M. Williams, William Schubert, So-Young Park, You Ree Cho, Michael P. Lisanti, Franco Capozza, Philipp E. Scherer, Alex W. Cohen, Linda A. Jelicks, and Dawn L. Brasaemle
- Subjects
Blood Glucose ,medicine.medical_specialty ,Physiology ,Caveolin 3 ,Gene Expression ,Caveolins ,Islets of Langerhans ,Mice ,Insulin resistance ,Caveolae ,Internal medicine ,medicine ,Myocyte ,Animals ,Insulin ,Muscle, Skeletal ,Insulin-like growth factor 1 receptor ,Mice, Knockout ,biology ,Skeletal muscle ,Cell Biology ,medicine.disease ,Receptor, Insulin ,Insulin oscillation ,Insulin receptor ,Endocrinology ,medicine.anatomical_structure ,Adipose Tissue ,Liver ,cardiovascular system ,biology.protein ,Body Composition ,Insulin Resistance ,Glycogen ,Signal Transduction - 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 ∼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.
- Published
- 2005
31. Phosphofructokinase muscle-specific isoform requires caveolin-3 expression for plasma membrane recruitment and caveolar targeting: implications for the pathogenesis of caveolin-related muscle diseases
- Author
-
Federica, Sotgia, Gloria, Bonuccelli, Carlo, Minetti, Scott E, Woodman, Franco, Capozza, Robert G, Kemp, Philipp E, Scherer, and Michael P, Lisanti
- Subjects
Mice, Knockout ,Caveolin 3 ,Caveolin 1 ,Cell Membrane ,Extracellular Fluid ,Caveolae ,Caveolins ,Recombinant Proteins ,Cell Line ,Isoenzymes ,Mice ,Glucose ,Phenotype ,Muscular Diseases ,Phosphofructokinases ,COS Cells ,Mutation ,cardiovascular system ,Animals ,Tissue Distribution ,Animal Model ,Muscle, Skeletal - Abstract
Previous co-immunoprecipitation studies have shown that endogenous PFK-M (phosphofructokinase, muscle-specific isoform) associates with caveolin (Cav)-3 under certain metabolic conditions. However, it remains unknown whether Cav-3 expression is required for the plasma membrane recruitment and caveolar targeting of PFK-M. Here, we demonstrate that recombinant expression of Cav-3 dramatically affects the subcellular localization of PFK-M, by targeting PFK-M to the plasma membrane, and by trans-locating PFK-M to caveolae-enriched membrane domains. In addition, we show that the membrane recruitment and caveolar targeting of PFK-M appears to be strictly dependent on the concentration of extracellular glucose. Interestingly, recombinant expression of PFK-M with three Cav-3 mutants [DeltaTFT (63 to 65), P104L, and R26Q], which harbor the same mutations as seen in the human patients with Cav-3-related muscle diseases, causes a substantial reduction in PFK-M expression levels, and impedes the membrane recruitment of PFK-M. Analysis of skeletal muscle tissue samples from Cav-3(-/-) mice directly demonstrates that Cav-3 expression regulates the phenotypic behavior of PFK-M. More specifically, in Cav-3-null mice, PFK-M is no longer targeted to the plasma membrane, and is excluded from caveolar membrane domains. As such, our current results may be important in understanding the pathogenesis of Cav-3-related muscle diseases, such as limb-girdle muscular dystrophy-1C, distal myopathy, and rippling muscle disease, that are caused by mutations within the human Cav-3 gene.
- Published
- 2003
32. Absence of Caveolin-1 Sensitizes Mouse Skin to Carcinogen-Induced Epidermal Hyperplasia and Tumor Formation
- Author
-
Terence M. Williams, Michael P. Lisanti, William Schubert, Franco Capozza, Boumediene Bouzahzah, Federica Sotgia, and Steve A. McClain
- Subjects
Keratinocytes ,Cell type ,Pathology ,medicine.medical_specialty ,Time Factors ,Tumor suppressor gene ,9,10-Dimethyl-1,2-benzanthracene ,Caveolin 1 ,DMBA ,Biology ,medicine.disease_cause ,Caveolins ,Pathology and Forensic Medicine ,Mice ,Cyclin D1 ,medicine ,Animals ,Phosphorylation ,Skin ,Mice, Knockout ,Mitogen-Activated Protein Kinase 1 ,Hyperplasia ,Mitogen-Activated Protein Kinase 3 ,Neoplasms, Experimental ,medicine.disease ,Null allele ,Immunohistochemistry ,Up-Regulation ,Mice, Inbred C57BL ,Microscopy, Electron ,Cancer research ,Carcinogens ,Animal Model ,Epidermis ,Mitogen-Activated Protein Kinases ,Carcinogenesis ,Cell Division - Abstract
Caveolin-1 is the principal protein component of caveolae membrane domains, which are located at the cell surface in most cell types. Evidence has accumulated suggesting that caveolin-1 may function as a suppressor of cell transformation in cultured cells. The human CAV-1 gene is located at a putative tumor suppressor locus (7q31.1/D7S522) and a known fragile site (FRA7G) that is deleted in a variety of epithelial-derived tumors. Mechanistically, caveolin-1 is known to function as a negative regulator of the Ras-p42/44 MAP kinase cascade and as a transcriptional repressor of cyclin D1, possibly explaining its transformation suppressor activity in cultured cells. However, it remains unknown whether caveolin-1 functions as a tumor suppressor gene in vivo. Here, we examine the tumor suppressor function of caveolin-1 using Cav-1 (-/-) null mice as a model system. Cav-1 null mice and their wild-type counterparts were subjected to carcinogen-induced skin tumorigenesis, using 7,12-dimethylbenzanthracene (DMBA). Mice were monitored weekly for the development of tumors. We demonstrate that Cav-1 null mice are dramatically more susceptible to carcinogen-induced tumorigenesis, as they develop skin tumors at an increased rate. After 16 weeks of DMBA-treatment, Cav-1 null mice showed a 10-fold increase in tumor incidence, a 15-fold increase in tumor number per mouse (multiplicity), and a 35-fold increase in tumor area per mouse, as compared with wild-type littermate mice. Moreover, before the development of tumors, DMBA-treatment induced severe epidermal hyperplasia in Cav-1 null mice. Both the basal cell layer and the suprabasal cell layers were expanded in treated Cav-1 null mice, as evidenced by immunostaining with cell-type specific differentiation markers (keratin-10 and keratin-14). In addition, cyclin D1 and phospho-ERK1/2 levels were up-regulated during epidermal hyperplasia, suggesting a possible mechanism for the increased susceptibility of Cav-1 null mice to tumorigenesis. However, the skin of untreated Cav-1 null mice appeared normal, without any evidence of epidermal hyperplasia, despite the fact that Cav-1 null keratinocytes failed to express caveolin-1 and showed a complete ablation of caveolae formation. Thus, Cav-1 null mice require an appropriate oncogenic stimulus, such as DMBA treatment, to reveal their increased susceptibility toward epidermal hyperplasia and skin tumor formation. Our results provide the first genetic evidence that caveolin-1 indeed functions as a tumor suppressor gene in vivo.
- Published
- 2003
33. Intracellular Retention of Glycosylphosphatidyl Inositol-Linked Proteins in Caveolin-Deficient Cells
- Author
-
Babak Razani, William Schubert, Ann Lane Schubert, Franco Capozza, Michael P. Lisanti, Gloria Bonuccelli, J. Thomas Buckley, Carlo Minetti, Hyangkyu Lee, Michela Battista, and Federica Sotgia
- Subjects
Intracellular Fluid ,Glycosylphosphatidylinositols ,Caveolin 1 ,Palmitic Acid ,Biological Transport, Active ,Biology ,medicine.disease_cause ,Transfection ,Caveolins ,CSK Tyrosine-Protein Kinase ,Mice ,Membrane Microdomains ,Palmitoylation ,Caveolae ,Caveolin ,Protein targeting ,Null cell ,medicine ,Animals ,Muscle, Skeletal ,Molecular Biology ,Lipid raft ,Cell Growth and Development ,Lung ,Glycoproteins ,chemistry.chemical_classification ,Mice, Knockout ,Binding Sites ,Cell Biology ,3T3 Cells ,Protein-Tyrosine Kinases ,Cell biology ,Cell Compartmentation ,Kidney Tubules ,src-Family Kinases ,chemistry ,Membrane protein ,Biochemistry ,lipids (amino acids, peptides, and proteins) ,Glycoprotein - Abstract
The relationship between glycosylphosphatidyl inositol (GPI)-linked proteins and caveolins remains controversial. Here, we derived fibroblasts from Cav-1 null mouse embryos to study the behavior of GPI-linked proteins in the absence of caveolins. These cells lack morphological caveolae, do not express caveolin-1, and show a approximately 95% down-regulation in caveolin-2 expression; these cells also do not express caveolin-3, a muscle-specific caveolin family member. As such, these caveolin-deficient cells represent an ideal tool to study the role of caveolins in GPI-linked protein sorting. We show that in Cav-1 null cells GPI-linked proteins are preferentially retained in an intracellular compartment that we identify as the Golgi complex. This intracellular pool of GPI-linked proteins is not degraded and remains associated with intracellular lipid rafts as judged by its Triton insolubility. In contrast, GPI-linked proteins are transported to the plasma membrane in wild-type cells, as expected. Furthermore, recombinant expression of caveolin-1 or caveolin-3, but not caveolin-2, in Cav-1 null cells complements this phenotype and restores the cell surface expression of GPI-linked proteins. This is perhaps surprising, as GPI-linked proteins are confined to the exoplasmic leaflet of the membrane, while caveolins are cytoplasmically oriented membrane proteins. As caveolin-1 normally undergoes palmitoylation on three cysteine residues (133, 143, and 156), we speculated that palmitoylation might mechanistically couple caveolin-1 to GPI-linked proteins. In support of this hypothesis, we show that palmitoylation of caveolin-1 on residues 143 and 156, but not residue 133, is required to restore cell surface expression of GPI-linked proteins in this complementation assay. We also show that another lipid raft-associated protein, c-Src, is retained intracellularly in Cav-1 null cells. Thus, Golgi-associated caveolins and caveola-like vesicles could represent part of the transport machinery that is necessary for efficiently moving lipid rafts and their associated proteins from the trans-Golgi to the plasma membrane. In further support of these findings, GPI-linked proteins were also retained intracellularly in tissue samples derived from Cav-1 null mice (i.e., lung endothelial and renal epithelial cells) and Cav-3 null mice (skeletal muscle fibers).
- Published
- 2002
34. Caveolin-1 expression negatively regulates cell cycle progression by inducing G(0)/G(1) arrest via a p53/p21(WAF1/Cip1)-dependent mechanism
- Author
-
Daniela Volonté, Liang Zhu, Michael P. Lisanti, Richard G. Pestell, Ferruccio Galbiati, Franco Capozza, Philippe G. Frank, and Jun Liu
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Male ,Cell cycle checkpoint ,Recombinant Fusion Proteins ,Cell ,Cyclin A ,Caveolin 1 ,Immunoblotting ,Mice, Transgenic ,Cell Separation ,Caveolins ,Culture Media, Serum-Free ,Article ,Mice ,Genes, Reporter ,Pregnancy ,Cyclins ,medicine ,Animals ,Humans ,CHEK1 ,Enzyme Inhibitors ,Molecular Biology ,S phase ,Cells, Cultured ,biology ,Caspase 3 ,Cell Cycle ,S-phase-promoting factor ,Cell Biology ,Cell cycle ,Fibroblasts ,Flow Cytometry ,Staurosporine ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Caspases ,biology.protein ,Female ,Tumor Suppressor Protein p53 ,Restriction point - Abstract
Caveolin-1 is a principal component of caveolae membranes in vivo. Caveolin-1 mRNA and protein expression are lost or reduced during cell transformation by activated oncogenes. Interestingly, the human caveolin-1 gene is localized to a suspected tumor suppressor locus (7q31.1). However, it remains unknown whether caveolin-1 plays any role in regulating cell cycle progression. Here, we directly demonstrate that caveolin-1 expression arrests cells in the G0/G1phase of the cell cycle. We show that serum starvation induces up-regulation of endogenous caveolin-1 and arrests cells in the G0/G1phase of the cell cycle. Moreover, targeted down-regulation of caveolin-1 induces cells to exit the G0/G1phase. Next, we constructed a green fluorescent protein-tagged caveolin-1 (Cav-1-GFP) to examine the effect of caveolin-1 expression on cell cycle regulation. We directly demonstrate that recombinant expression of Cav-1-GFP induces arrest in the G0/G1phase of the cell cycle. To examine whether caveolin-1 expression is important for modulating cell cycle progression in vivo, we expressed wild-type caveolin-1 as a transgene in mice. Analysis of primary cultures of mouse embryonic fibroblasts from caveolin-1 transgenic mice reveals that caveolin-1 induces 1) cells to exit the S phase of the cell cycle with a concomitant increase in the G0/G1population, 2) a reduction in cellular proliferation, and 3) a reduction in the DNA replication rate. Finally, we demonstrate that caveolin-1-mediated cell cycle arrest occurs through a p53/p21-dependent pathway. Taken together, our results provide the first evidence that caveolin-1 expression plays a critical role in the modulation of cell cycle progression in vivo.
- Published
- 2001
35. Abstract 1631: Cav1 is a key mediator of tumor-stromal interactions in melanoma
- Author
-
Casey Trimmer and Franco Capozza
- Subjects
Cancer Research ,Tumor microenvironment ,Stromal cell ,MMP2 ,Melanoma ,medicine.medical_treatment ,Cancer ,Biology ,medicine.disease ,Metastasis ,Paracrine signalling ,Cytokine ,Oncology ,medicine ,Cancer research - Abstract
Several lines of experimental evidence have demonstrated the importance of the tumor microenvironment in controlling melanoma tumor growth and melanoma metastasis. Caveolin1 (Cav1), the main structural component of the plasma membrane microdomains termed Caveolae, is emerging as an important signaling molecule in the stroma of several tumor types. However, Cav1’s function in the melanoma microenvironment of primary tumors and of metastasis remains largely unexplored. Here, we devise various experimental approaches to elucidate the function of stromal Cav1 in the development of melanoma in mice. We show that loss of Cav1 (but not Cav2) in mice promotes the growth of orthotopically implanted melanoma cells. We use cocultures of fibroblasts and melanoma cells coupled with cytokine analysis to identify various Cav1 regulated factors that function in a paracrine fashion in melanoma. Cav1KO fibroblasts secrete increased amount of ShhN, bFGF, and MMP2/3, cytokines known to promote proliferation of melanoma cells and remodeling of the tumor stroma during melanomagenesis. Intradermal xenografts of fibroblasts and melanoma cells (5:1 ratios) confirmed the tumor promoting role of fibroblasts lacking Cav1. Interestingly, constitutive inhibition of the Shh pathway in melanoma cells reverses the growth and tumor-promoting effects of Cav1KO fibroblasts in co-culture or co-injection experiments suggesting a Cav1 mediated Shh heterotypic interaction between fibroblasts and melanoma cells. In summary, these studies reveal previously undefined functions for Cav1 in the melanoma microenvironment that could potentially be targeted for therapy. Citation Format: Casey Trimmer, Franco Capozza. Cav1 is a key mediator of tumor-stromal interactions in melanoma. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 1631. doi:10.1158/1538-7445.AM2013-1631 Note: This abstract was not presented at the AACR Annual Meeting 2013 because the presenter was unable to attend.
- Published
- 2013
36. Abstract 1083: Caveolin-1 in cutaneous squamous cell carcinoma development
- Author
-
Casey Trimmer, Franco Capozza, Michael P. Lisanti, Sanjay Katiyar, and Richard G. Pestell
- Subjects
Cancer Research ,Cell signaling ,Pathology ,medicine.medical_specialty ,Cell growth ,Melanoma ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,medicine.anatomical_structure ,Oncology ,Caveolin 1 ,medicine ,Cancer research ,Skin cancer ,Carcinogenesis ,Keratinocyte - Abstract
Caveolin-1 (Cav1) is the main protein component of a specialized form of lipid raft termed caveolae and serves to compartmentalize and negatively regulate the activity of signaling molecules involved in cell proliferation and survival. Cav1 has been shown to act as a tumor suppressor in several types of cancer, including breast cancer and melanoma, but its role in the pathogenesis of non-melanoma skin cancer remains largely unexplored. Previous work has indicated that Cav1 is expressed in the basal cell layer of both murine and human epidermis; additionally, loss of Cav1 protein has been shown by our lab to render murine skin more susceptible to papilloma development following treatment with a carcinogenic compound. The purpose of the current study was to further examine the role of Cav1 in non-melanoma skin cancer development using various experimental approaches. Given the previous research, our hypothesis is that Cav1 acts as a suppressor of tumor growth and development in the skin. In order to explore this hypothesis, we have used several experimental systems, including human tissue arrays and loss of function and gain of function studies in animal models of cutaneous squamous cell carcinoma. Manipulation of Cav1 expression in (1) a keratinocyte cell line followed by xenotransplantation and (2) an induced model of skin cancer allowed us to assess the effect of this protein on tumor development, growth, and progression. Using these approaches, we were able to show that Cav1 ablation dramatically increases not only in vitro cell growth and invasion, but also in vivo tumorigenesis, tumor growth, and invasion. In conclusion, these studies provide further evidence in support of the hypothesis that Cav1 acts as a tumor suppressor protein in the skin and serve to identify new potential therapeutic targets in the treatment of cutaneous malignancies. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 1083. doi:10.1158/1538-7445.AM2011-1083
- Published
- 2011
37. Phosphofructokinase Muscle-Specific Isoform Requires Caveolin-3 Expression for Plasma Membrane Recruitment and Caveolar Targeting: Implications for the Pathogenesis of Caveolin-Related Muscle Diseases
- Author
-
Sotgia, F., Bonuccelli, G., Minetti, C., Woodman, S. E., Franco Capozza, Kemp, R. G., Scherer, P. E., and Lisanti, M. P.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.