89 results on '"Frank PG"'
Search Results
2. Proteasome inhibitor (MG-132) treatment of mdx mice rescues the expression and membrane localization of dystrophin-associated proteins
- Author
-
Bonuccelli, G, Sotgia, F, Schubert, W, Park, D, Frank, Pg, Woodman, Se, Insabato, L, Cammer, M, Minetti, Carlo, and Lisanti, Mp
- Published
- 2003
3. Proteasome inhibitor (MG-132) treatment of mdx mice rescues the expression and membrane localization of dystrophin and dystrophin-associated proteins
- Author
-
William Schubert, Michael Cammer, Scott Eric Woodman, Philippe G. Frank, Carlo Minetti, David S. Park, Gloria Bonuccelli, Federica Sotgia, Luigi Insabato, Michael P. Lisanti, Bonuccelli, G, Sotgia, F, Schubert, W, Park, D, Frank, Pg, Woodman, Se, Insabato, Luigi, Cammer, M, Minetti, C, and Lisanti, Mp
- Subjects
musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Leupeptins ,Injections, Subcutaneous ,Duchenne muscular dystrophy ,Cysteine Proteinase Inhibitors ,Injections, Intramuscular ,Drug Administration Schedule ,Pathology and Forensic Medicine ,Dystrophin ,Mice ,Gastrocnemius muscle ,Sarcoglycans ,medicine ,Animals ,Tissue Distribution ,Muscular dystrophy ,Dystroglycans ,Muscle, Skeletal ,Infusion Pumps ,Membrane Glycoproteins ,biology ,Cell Membrane ,Skeletal muscle ,musculoskeletal system ,medicine.disease ,Dystrophin-associated protein ,Cell biology ,Muscular Dystrophy, Duchenne ,Cytoskeletal Proteins ,Membrane glycoproteins ,medicine.anatomical_structure ,Biochemistry ,Mice, Inbred mdx ,biology.protein ,Animal Model ,ITGA7 - Abstract
Dystrophin, the protein product of the Duchenne muscular dystrophy (DMD) gene, is absent in the skeletal muscle of DMD patients and mdx mice. At the plasma membrane of skeletal muscle fibers, dystrophin associates with a multimeric protein complex, termed the dystrophin-glycoprotein complex (DGC). Protein members of this complex are normally absent or greatly reduced in dystrophin-deficient skeletal muscle fibers, and are thought to undergo degradation through an unknown pathway. As such, we reasoned that inhibition of the proteasomal degradation pathway might rescue the expression and subcellular localization of dystrophin-associated proteins. To test this hypothesis, we treated mdx mice with the well-characterized proteasomal inhibitor MG-132. First, we locally injected MG-132 into the gastrocnemius muscle, and observed the outcome after 24 hours. Next, we performed systemic treatment using an osmotic pump that allowed us to deliver different concentrations of the proteasomal inhibitor, over an 8-day period. By immunofluorescence and Western blot analysis, we show that administration of the proteasomal inhibitor MG-132 effectively rescues the expression levels and plasma membrane localization of dystrophin, beta-dystroglycan, alpha-dystroglycan, and alpha-sarcoglycan in skeletal muscle fibers from mdx mice. Furthermore, we show that systemic treatment with the proteasomal inhibitor 1) reduces muscle membrane damage, as revealed by vital staining (with Evans blue dye) of the diaphragm and gastrocnemius muscle isolated from treated mdx mice, and 2) ameliorates the histopathological signs of muscular dystrophy, as judged by hematoxylin and eosin staining of muscle biopsies taken from treated mdx mice. Thus, the current study opens new and important avenues in our understanding of the pathogenesis of DMD. Most importantly, these new findings may have clinical implications for the pharmacological treatment of patients with DMD.
- Published
- 2003
4. Is cholesterol a risk factor for breast cancer incidence and outcome?
- Author
-
Ben Hassen C, Goupille C, Vigor C, Durand T, Guéraud F, Silvente-Poirot S, Poirot M, and Frank PG
- Subjects
- Humans, Female, Incidence, Cholesterol metabolism, Cholesterol Esters metabolism, Risk Factors, Breast Neoplasms metabolism
- Abstract
Cholesterol plays important roles in many physiological processes, including cell membrane structure and function, hormone synthesis, and the regulation of cellular homeostasis. The role of cholesterol in breast cancer is complex, and some studies have suggested that elevated cholesterol levels may be associated with an increased risk of developing breast cancer, while others have found no significant association. On the other hand, other studies have shown that, for total cholesterol and plasma HDL-associated cholesterol levels, there was inverse association with breast cancer risk. One possible mechanism by which cholesterol may contribute to breast cancer risk is as a key precursor of estrogen. Other potential mechanisms by which cholesterol may contribute to breast cancer risk include its role in inflammation and oxidative stress, which have been linked to cancer progression. Cholesterol has also been shown to play a role in signaling pathways regulating the growth and proliferation of cancer cells. In addition, recent studies have shown that cholesterol metabolism can generate tumor promoters such as cholesteryl esters, oncosterone, 27-hydroxycholesterol but also tumor suppressor metabolites such as dendrogenin A. This review summarizes some of the most important clinical studies that have evaluated the role of cholesterol or its derivatives in breast cancer. It also addresses the role of cholesterol and its derivatives at the cellular level., Competing Interests: Conflicts of interest The authors declare no conflict of interest., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
5. Curcumin and NCLX inhibitors share anti-tumoral mechanisms in microsatellite-instability-driven colorectal cancer.
- Author
-
Guéguinou M, Ibrahim S, Bourgeais J, Robert A, Pathak T, Zhang X, Crottès D, Dupuy J, Ternant D, Monbet V, Guibon R, Flores-Romero H, Lefèvre A, Lerondel S, Le Pape A, Dumas JF, Frank PG, Girault A, Chautard R, Guéraud F, García-Sáez AJ, Ouaissi M, Emond P, Sire O, Hérault O, Fromont-Hankard G, Vandier C, Tougeron D, Trebak M, Raoul W, and Lecomte T
- Subjects
- Animals, Calcium metabolism, Calcium Signaling, Humans, Mice, Microsatellite Repeats, Mitochondrial Proteins metabolism, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Curcumin pharmacology, Microsatellite Instability, Sodium-Calcium Exchanger antagonists & inhibitors
- Abstract
Background and Aims: Recent evidences highlight a role of the mitochondria calcium homeostasis in the development of colorectal cancer (CRC). To overcome treatment resistance, we aimed to evaluate the role of the mitochondrial sodium-calcium-lithium exchanger (NCLX) and its targeting in CRC. We also identified curcumin as a new inhibitor of NCLX., Methods: We examined whether curcumin and pharmacological compounds induced the inhibition of NCLX-mediated mitochondrial calcium (mtCa
2+ ) extrusion, the role of redox metabolism in this process. We evaluated their anti-tumorigenic activity in vitro and in a xenograft mouse model. We analyzed NCLX expression and associations with survival in The Cancer Genome Atlas (TCGA) dataset and in tissue microarrays from 381 patients with microsatellite instability (MSI)-driven CRC., Results: In vitro, curcumin exerted strong anti-tumoral activity through its action on NCLX with mtCa2+ and reactive oxygen species overload associated with a mitochondrial membrane depolarization, leading to reduced ATP production and apoptosis. NCLX inhibition with pharmacological and molecular approaches reproduced the effects of curcumin. NCLX inhibitors decreased CRC tumor growth in vivo. Both transcriptomic analysis of TCGA dataset and immunohistochemical analysis of tissue microarrays demonstrated that higher NCLX expression was associated with MSI status, and for the first time, NCLX expression was significantly associated with recurrence-free survival., Conclusions: Our findings highlight a novel anti-tumoral mechanism of curcumin through its action on NCLX and mitochondria calcium overload that could benefit for therapeutic schedule of patients with MSI CRC., (© 2022. The Author(s), under exclusive licence to Springer Nature Switzerland AG.)- Published
- 2022
- Full Text
- View/download PDF
6. Identification of a Positive Association between Mammary Adipose Cholesterol Content and Indicators of Breast Cancer Aggressiveness in a French Population.
- Author
-
Goupille C, Ouldamer L, Pinault M, Guimares C, Arbion F, Jourdan ML, and Frank PG
- Subjects
- Adult, Aged, Aged, 80 and over, Breast Neoplasms epidemiology, Female, France epidemiology, Humans, Middle Aged, Adipose Tissue metabolism, Breast Neoplasms pathology, Cholesterol metabolism, Mammary Glands, Human metabolism, Mammary Glands, Human pathology, Neoplasm Invasiveness pathology
- Abstract
Background: Several studies have recently highlighted important roles for adipose tissue in cancer. However, few have examined adipose tissue cholesterol, and no study has been performed in breast adipose tissue associated with breast tumors., Objectives: The present work was designed to determine if breast adipose tissue cholesterol from the tumor-surrounding area is associated with breast cancer aggressiveness., Methods: Between 2009 and 2011, 215 breast adipose tissue samples were collected at the Tours University Hospital (France) during surgery of women (aged 28-89 y) with invasive breast cancer. Associations of free cholesterol (FC), esterified cholesterol (EC), and total cholesterol (TC) amounts with clinical variables (age, BMI, and treated or untreated hypercholesterolemia) and tumor aggressiveness parameters [phenotype, grade, presence of inflammatory breast cancer (IBC), and multifocality] were tested using Student's t test and after ANOVA., Results: The predominant form of cholesterol in adipose tissue was FC, and 50% of patients had no detectable EC. The adipose tissue FC content (μg/mg total lipid) was 18% greater in patients >70 y old than in those 40-49 y old (P < 0.05) and the TC content tended to be 12% greater in untreated hypercholesterolemic patients than in normocholesterolemic patients (P = 0.06). Breast adipose cholesterol concentrations were increased in tissues obtained from patients with human-epidermal-growth-factor-receptor-2 (HER2) phenotype (+13% FC; P < 0.05 compared with luminal A), IBC (+15% FC; P = 0.06 compared with noninflammatory tumors), as well as with multifocal triple-negative tumors (+34% FC, P < 0.05; +30% TC, P < 0.05, compared with unifocal triple-negative tumors). Among patients with triple-negative tumors, hypercholesterolemia was significantly more common (P < 0.05) in patients with multifocal tumors (64%) than in patients with unifocal tumors (25%)., Conclusions: This study is the first of this magnitude that analyzes cholesterol concentrations in adipose tissue from female breast cancer patients. An increase in breast adipose tissue cholesterol content may contribute to breast cancer aggressiveness (HER2 phenotype, multifocality of triple-negative tumors, and IBC)., (© The Author(s) 2021. Published by Oxford University Press on behalf of the American Society for Nutrition.)
- Published
- 2021
- Full Text
- View/download PDF
7. Low Levels of Omega-3 Long-Chain Polyunsaturated Fatty Acids Are Associated with Bone Metastasis Formation in Premenopausal Women with Breast Cancer: A Retrospective Study.
- Author
-
Goupille C, Frank PG, Arbion F, Jourdan ML, Guimaraes C, Pinault M, Body G, Chevalier S, Bougnoux P, and Ouldamer L
- Subjects
- Adult, Aged, Aged, 80 and over, Breast metabolism, Breast pathology, Breast Neoplasms metabolism, Chromatography, Gas, Female, Humans, Middle Aged, Neoplasm Metastasis, Phenotype, Postmenopause metabolism, Retrospective Studies, Risk Factors, Adipose Tissue metabolism, Bone Neoplasms secondary, Breast Neoplasms pathology, Fatty Acids, Omega-3 metabolism, Premenopause metabolism
- Abstract
In the present study, we investigated various biochemical, clinical, and histological factors associated with bone metastases in a large cohort of pre- and postmenopausal women with breast cancer. Two hundred and sixty-one consecutive women with breast cancer were included in this study. Breast adipose tissue specimens were collected during surgery. After having established the fatty acid profile of breast adipose tissue by gas chromatography, we determined whether there were differences associated with the occurrence of bone metastases in these patients. Regarding the clinical and histological criteria, a majority of the patients with bone metastases (around 70%) had tumors with a luminal phenotype and 59% of them showed axillary lymph node involvement. Moreover, we found a negative association between the levels of n- 3 long-chain polyunsaturated fatty acids (LC-PUFA) in breast adipose tissue and the development of bone metastases in premenopausal women. No significant association was observed in postmenopausal women. In addition to a luminal phenotype and axillary lymph node involvement, low levels of n- 3 LC-PUFA in breast adipose tissue may constitute a risk factor that contributes to breast cancer bone metastases formation in premenopausal women.
- Published
- 2020
- Full Text
- View/download PDF
8. EPA and DHA Fatty Acids Induce a Remodeling of Tumor Vasculature and Potentiate Docetaxel Activity.
- Author
-
Goupille C, Vibet S, Frank PG, and Mahéo K
- Subjects
- Animals, Diet, Fatty Acids, Omega-3 pharmacology, Female, Mammary Neoplasms, Animal pathology, Neovascularization, Pathologic metabolism, Rats, Rats, Sprague-Dawley, Docetaxel pharmacology, Docosahexaenoic Acids pharmacology, Eicosapentaenoic Acid pharmacology, Fatty Acids pharmacology, Mammary Neoplasms, Animal drug therapy, Neovascularization, Pathologic drug therapy
- Abstract
n-3 long chain Polyunsaturated Fatty Acids (n-3 LCPUFA) have been shown to improve the efficacy of conventional chemotherapies used for breast cancer treatment. In addition to their reported ability to increase the chemosensitivity of cancer cells, we hypothesized that n-3 LCPUFA could induce a remodeling of the vascular network in mammary tumors. A contrast-enhanced ultrasound method was used to monitor the vascular architecture during docetaxel treatment of mammary tumors in rats fed either a control or an n-3 LCPUFA-enriched diet (docosahexaenoic acid (DHA)/eicosapentaenoic acid (EPA)). The vascular network was remodeled in favor of smaller vessels (microvascularization), which represented 54% of the vasculature in n-3 LCPUFA tumors but only 26% in control tumors after 2 weeks of chemotherapy. Importantly, vascularization changes occurred both before and during docetaxel treatment. The density of smaller vessels quantified before chemotherapy was correlated with improved tumor size reduction by docetaxel treatment. Furthermore, transcript levels of the angiogenesis-specific genes epiregulin and amphiregulin were reduced by ~4.5- and twofold in tumors obtained from rats fed an n-3 LCPUFA-enriched diet compared to those of rats fed a control diet, respectively. Their expression levels were negatively correlated with tumor regression after chemotherapy. Taken together, this preclinical data strengthen the potential usefulness of n-3 LCPUFA as a complementary clinical strategy to improve drug efficiency via remodeling of the tumor vasculature.
- Published
- 2020
- Full Text
- View/download PDF
9. Development of a Novel High-Performance Thin Layer Chromatography-Based Method for the Simultaneous Quantification of Clinically Relevant Lipids from Cells and Tissue Extracts.
- Author
-
Pinault M, Guimaraes C, Ben Hassen C, Gutierrez-Pajares JL, Chevalier S, Goupille C, Bernard-Savary P, and Frank PG
- Subjects
- Breast pathology, Cell Line, Tumor, Cholesterol analysis, Cholesterol Esters analysis, Chromatography, Thin Layer, Fatty Acids, Nonesterified analysis, Humans, MCF-7 Cells, Triglycerides analysis, Breast chemistry, Lipids analysis, Tissue Extracts chemistry
- Abstract
Lipids such as cholesterol, triacylglycerols, and fatty acids play important roles in the regulation of cellular metabolism and cellular signaling pathways and, as a consequence, in the development of various diseases. It is therefore important to understand how their metabolism is regulated to better define the components involved in the development of various human diseases. In the present work, we describe the development and validation of a high-performance thin layer chromatography (HPTLC) method allowing the separation and quantification of free cholesterol, cholesteryl esters, nonesterified fatty acids, and triacylglycerols. This method will be of interest as the quantification of these lipids in one single assay is difficult to perform., (© 2020 AOCS.)
- Published
- 2020
- Full Text
- View/download PDF
10. Apolipoprotein-mediated regulation of lipid metabolism induces distinctive effects in different types of breast cancer cells.
- Author
-
Ben Hassen C, Gutierrez-Pajares JL, Guimaraes C, Guibon R, Pinault M, Fromont G, and Frank PG
- Subjects
- Animals, Breast Neoplasms classification, Cell Line, Tumor, Cell Movement, Databases, Genetic, Epithelial-Mesenchymal Transition, Female, Humans, Mice, Mice, Nude, Survival Rate, Xenograft Model Antitumor Assays, Apolipoprotein A-I metabolism, Apolipoproteins E metabolism, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Proliferation, Cholesterol metabolism, Lipid Metabolism
- Abstract
Background: The highest incidence of breast cancer is in the Western world. Several aspects of the Western lifestyle are known risk factors for breast cancer. In particular, previous studies have shown that cholesterol levels can play an important role in the regulation of tumor progression., Methods: In the present study, we modulated cholesterol metabolism in the human breast cancer cell lines MCF-7 and MDA-MB-231 using a genetic approach. Apolipoprotein A-I (apoA-I) and apolipoprotein E (apoE) were expressed in these cell lines to modulate cholesterol metabolism. The effects of these apolipoproteins on cancer cell properties were examined., Results: Our results show that both apolipoproteins can regulate cholesterol metabolism and can control the epithelial-to-mesenchymal transition process. However, these effects were different depending on the cell type. We show that expressing apoA-I or apoE stimulates proliferation, migration, and tumor growth of MCF-7 cells. However, apoA-I or apoE reduces proliferation and migration of MDA-MB-231 cells., Conclusions: These data suggest that modulating sterol metabolism may be most effective at limiting tumor progression in models of triple-negative cancers.
- Published
- 2020
- Full Text
- View/download PDF
11. Oxidized Products of α-Linolenic Acid Negatively Regulate Cellular Survival and Motility of Breast Cancer Cells.
- Author
-
Gutierrez-Pajares JL, Ben Hassen C, Oger C, Galano JM, Durand T, and Frank PG
- Subjects
- Antineoplastic Agents chemistry, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Drug Screening Assays, Antitumor, Female, Furans chemistry, Humans, MCF-7 Cells, Oxidation-Reduction, Prostanoic Acids chemistry, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Cell Movement drug effects, Furans pharmacology, Prostanoic Acids pharmacology, alpha-Linolenic Acid chemistry
- Abstract
Despite recent advances in our understanding of the biological processes leading to the development and progression of cancer, there is still a need for new and effective agents to treat this disease. Phytoprostanes (PhytoPs) and phytofurans (PhytoFs) are non-enzymatically oxidized products of α-linolenic acid that are present in seeds and vegetable oils. They have been shown to possess anti-inflammatory and apoptosis-promoting activities in macrophages and leukemia cells, respectively. In this work, seven PhytoPs (PP1-PP7) and one PhytoFs (PF1) were evaluated for their cytotoxic, chemosensitization, and anti-migratory activities using the MCF-7 and MDA-MB-231 breast cancer cell lines. Among the tested compounds, only three PhytoPs had a significant effect on cell viability compared to the control group: Ent -9-L
1 -PhytoP (PP6) decreased cell viability in both cell lines, while 16-F1t -PhytoP (PP1) and 9-L1 -PhytoP (PP5) decreased viability of MCF-7 and MDA-MB-231 cells, respectively. When combined with a sub-cytotoxic dose of doxorubicin, these three PhytoPs displayed significantly enhanced cytotoxic effects on MCF-7 cells while the chemotherapeutic drug alone had no effect. In cellular motility assays, Ent -9-( RS )-12- epi -ST-Δ10 -13-PhytoF could significantly inhibit cellular migration of MDA-MB-231 cells. In addition, Ent -9-( RS )-12- epi -ST-Δ10 -13-PhytoF also enhanced cellular adhesion of MDA-MB-231 cells.- Published
- 2019
- Full Text
- View/download PDF
12. SCN4B acts as a metastasis-suppressor gene preventing hyperactivation of cell migration in breast cancer.
- Author
-
Bon E, Driffort V, Gradek F, Martinez-Caceres C, Anchelin M, Pelegrin P, Cayuela ML, Marionneau-Lambot S, Oullier T, Guibon R, Fromont G, Gutierrez-Pajares JL, Domingo I, Piver E, Moreau A, Burlaud-Gaillard J, Frank PG, Chevalier S, Besson P, and Roger S
- Subjects
- Animals, Breast Neoplasms ultrastructure, Cell Line, Tumor, Cell Proliferation, Disease Progression, Down-Regulation genetics, Epithelial Cells metabolism, Extracellular Matrix metabolism, Female, Gene Expression Regulation, Neoplastic, Human Umbilical Vein Endothelial Cells metabolism, Humans, Ion Channel Gating, Mice, Nude, Neoplasm Invasiveness, Neoplasm Metastasis, Prognosis, Protein Subunits metabolism, Sodium Channels metabolism, Voltage-Gated Sodium Channel beta-4 Subunit metabolism, Zebrafish, rhoA GTP-Binding Protein metabolism, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Movement genetics, Genes, Tumor Suppressor, Voltage-Gated Sodium Channel beta-4 Subunit genetics
- Abstract
The development of metastases largely relies on the capacity of cancer cells to invade extracellular matrices (ECM) using two invasion modes termed 'mesenchymal' and 'amoeboid', with possible transitions between these modes. Here we show that the SCN4B gene, encoding for the β4 protein, initially characterized as an auxiliary subunit of voltage-gated sodium channels (Na
V ) in excitable tissues, is expressed in normal epithelial cells and that reduced β4 protein levels in breast cancer biopsies correlate with high-grade primary and metastatic tumours. In cancer cells, reducing β4 expression increases RhoA activity, potentiates cell migration and invasiveness, primary tumour growth and metastatic spreading, by promoting the acquisition of an amoeboid-mesenchymal hybrid phenotype. This hyperactivated migration is independent of NaV and is prevented by overexpression of the intracellular C-terminus of β4. Conversely, SCN4B overexpression reduces cancer cell invasiveness and tumour progression, indicating that SCN4B/β4 represents a metastasis-suppressor gene.- Published
- 2016
- Full Text
- View/download PDF
13. SR-BI: Linking Cholesterol and Lipoprotein Metabolism with Breast and Prostate Cancer.
- Author
-
Gutierrez-Pajares JL, Ben Hassen C, Chevalier S, and Frank PG
- Abstract
Studies have demonstrated the significant role of cholesterol and lipoprotein metabolism in the progression of cancer. The SCARB1 gene encodes the scavenger receptor class B type I (SR-BI), which is an 82-kDa glycoprotein with two transmembrane domains separated by a large extracellular loop. SR-BI plays an important role in the regulation of cholesterol exchange between cells and high-density lipoproteins. Accordingly, hepatic SR-BI has been shown to play an essential role in the regulation of the reverse cholesterol transport pathway, which promotes the removal and excretion of excess body cholesterol. In the context of atherosclerosis, SR-BI has been implicated in the regulation of intracellular signaling, lipid accumulation, foam cell formation, and cellular apoptosis. Furthermore, since lipid metabolism is a relevant target for cancer treatment, recent studies have focused on examining the role of SR-BI in this pathology. While signaling pathways have initially been explored in non-tumoral cells, studies with cancer cells have now demonstrated SR-BI's function in tumor progression. In this review, we will discuss the role of SR-BI during tumor development and malignant progression. In addition, we will provide insights into the transcriptional and post-transcriptional regulation of the SCARB1 gene. Overall, studying the role of SR-BI in tumor development and progression should allow us to gain useful information for the development of new therapeutic strategies.
- Published
- 2016
- Full Text
- View/download PDF
14. Caveolin-3 Promotes a Vascular Smooth Muscle Contractile Phenotype.
- Author
-
Gutierrez-Pajares JL, Iturrieta J, Dulam V, Wang Y, Pavlides S, Malacari G, Lisanti MP, and Frank PG
- Abstract
Epidemiological studies have demonstrated the importance of cardiovascular diseases in Western countries. Among the cell types associated with a dysfunctional vasculature, smooth muscle (SM) cells are believed to play an essential role in the development of these illnesses. Vascular SM cells are key regulators of the vascular tone and also have an important function in the development of atherosclerosis and restenosis. While in the normal vasculature, contractile SM cells are predominant, in atherosclerotic vascular lesions, synthetic cells migrate toward the neointima, proliferate, and synthetize extracellular matrix proteins. In the present study, we have examined the role of caveolin-3 in the regulation of SM cell phenotype. Caveolin-3 is expressed in vivo in normal arterial SM cells, but its expression appears to be lost in cultured SM cells. Our data show that caveolin-3 expression in the A7r5 SM cell line is associated with increased expression of contractility markers such as SM α-actin, SM myosin heavy chain but decreased expression of the synthetic phenotype markers such as p-Elk and Klf4. Moreover, we also show that caveolin-3 expression can reduce proliferation upon treatment with LDL or PDGF. Finally, we show that caveolin-3-expressing SM cells are less sensitive to apoptosis than control cells upon treatment with oxidized LDL. Taken together, our data suggest that caveolin-3 can regulate the phenotypic switch between contractile and synthetic SM cells. A better understanding of the factors regulating caveolin-3 expression and function in this cell type will permit the development of a better comprehension of the factors regulating SM function in atherosclerosis and restenosis.
- Published
- 2015
- Full Text
- View/download PDF
15. Caveolin-1 regulates the anti-atherogenic properties of macrophages.
- Author
-
Pavlides S, Gutierrez-Pajares JL, Katiyar S, Jasmin JF, Mercier I, Walters R, Pavlides C, Pestell RG, Lisanti MP, and Frank PG
- Subjects
- Animals, Apoptosis drug effects, Atherosclerosis blood, Bone Marrow Transplantation, Caveolin 1 deficiency, Cytokines metabolism, Inflammation pathology, Lipopolysaccharides pharmacology, Lipoproteins blood, Macrophages, Peritoneal drug effects, Mice, Inbred C57BL, Up-Regulation drug effects, Atherosclerosis pathology, Caveolin 1 metabolism, Macrophages, Peritoneal metabolism, Macrophages, Peritoneal pathology
- Abstract
Atherosclerosis is a complex disease initiated by the vascular accumulation of lipoproteins in the sub-endothelial space, followed by the infiltration of monocytes into the arterial intima. Caveolin-1 (Cav-1) plays an essential role in the regulation of cellular cholesterol metabolism and of various signaling pathways. In order to study specifically the role of macrophage Cav-1 in atherosclerosis, we used Cav-1 (-/-) Apoe (-/-) mice and transplanted them with bone marrow (BM) cells obtained from Cav-1 (+/+) Apoe (-/-) or Cav-1 (-/-) Apoe (-/-) mice and vice versa. We found that Cav-1 (+/+) mice harboring Cav-1 (-/-) BM-derived macrophages developed significantly larger lesions than Cav-1 (+/+) mice harboring Cav-1 (+/+) BM-derived macrophages. Cav-1 (-/-) macrophages were more susceptible to apoptosis and more prone to induce inflammation. The present study provides clear evidence that the absence of Cav-1 in macrophage is pro-atherogenic, whereas its absence in endothelial cells protects against atherosclerotic lesion formation. These findings demonstrate the cell-specific role of Cav-1 during the development of this disease.
- Published
- 2014
- Full Text
- View/download PDF
16. Endothelial caveolin-1 plays a major role in the development of atherosclerosis.
- Author
-
Pavlides S, Gutierrez-Pajares JL, Iturrieta J, Lisanti MP, and Frank PG
- Subjects
- Albumins metabolism, Animals, Aorta metabolism, Aorta pathology, Caveolae metabolism, Down-Regulation, Endocytosis, Endothelial Cells pathology, Gene Silencing, Human Umbilical Vein Endothelial Cells metabolism, Human Umbilical Vein Endothelial Cells pathology, Inflammation pathology, Lipoproteins, LDL metabolism, Mice, Mice, Inbred C57BL, NF-kappa B metabolism, RNA, Small Interfering metabolism, Signal Transduction, Atherosclerosis metabolism, Atherosclerosis pathology, Caveolin 1 metabolism, Endothelial Cells metabolism
- Abstract
Clinical studies have established the important impact of atherosclerotic disease in Western societies. This disease is characterized by the accumulation of lipids and the migration of various cell types in the sub-endothelial space of blood vessels. As demonstrated by many studies, endothelial cells play an essential role in the development of this disease. The endothelium acts as a gatekeeper of blood vessel integrity and cardiovascular health status. For instance, the transfer of lipids via the transport of lipoproteins in the arterial intima is believed to be mediated by endothelial cells through a process termed transcytosis. In addition, lipoproteins that accumulate in the sub-endothelial space may also be modified, in a process that can direct the activation of endothelial cells. These steps are essential for the initiation of an atherosclerotic plaque and may be mediated, at least in part, by caveolae and their associated protein caveolin-1. In the present study, we evaluate the role of caveolin-1/caveolae in the regulation of these two steps in endothelial cells. Our data clearly demonstrate that caveolin-1 is involved in the regulation of lipoprotein transcytosis across endothelial cells and in the regulation of vascular inflammation.
- Published
- 2014
- Full Text
- View/download PDF
17. Intra-chain photodimerization of pendant anthracene units as an efficient route to single-chain nanoparticle fabrication.
- Author
-
Frank PG, Tuten BT, Prasher A, Chao D, and Berda EB
- Subjects
- Chromatography, Gel, Dimerization, Microscopy, Electron, Transmission, Spectrophotometry, Ultraviolet, Viscosity, Anthracenes chemistry, Nanoparticles, Photochemistry
- Abstract
An efficient route to architecturally defined, sub-20 nm soft nanoparticles fabricated from single polymer chains via intramolecular photodimerization of pendant anthracene units is presented. Photodimerization is confirmed by the disappearance of the characteristic anthracene π-π* absorption peak at ≈ 360 nm measured by UV-vis spectroscopy. Size exclusion chromatography (SEC) with UV, multi-angle light scattering (MALS), and viscometric detection confirms that as photodimers form, the chains fold to form nanoparticles, demonstrated by shifts in the SEC traces to longer retention times as a function of increased irradiation time. These shifts indicate a reduction in hydrodynamic radius, corroborated and quantified by viscometric data. MALS detector traces reveal the presence of a small amount of chain-chain coupling during this process, but confirm that this is primarily a single-chain phenomenon. Electron microscopy provides visual confirmation of nanoparticle formation., (© 2013 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2014
- Full Text
- View/download PDF
18. Ablation of calcineurin Aβ reveals hyperlipidemia and signaling cross-talks with phosphodiesterases.
- Author
-
Suk HY, Zhou C, Yang TT, Zhu H, Yu RY, Olabisi O, Yang X, Brancho D, Kim JY, Scherer PE, Frank PG, Lisanti MP, Calvert JW, Lefer DJ, Molkentin JD, Ghigo A, Hirsch E, Jin J, and Chow CW
- Subjects
- Aging drug effects, Aging pathology, Amino Acid Sequence, Animals, COS Cells, Calcineurin metabolism, Chlorocebus aethiops, Cyclic AMP metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Cyclic Nucleotide Phosphodiesterases, Type 3 chemistry, Cyclosporine pharmacology, Embryo, Mammalian cytology, Enzyme Activation drug effects, Fibroblasts drug effects, Fibroblasts enzymology, Hyperlipidemias pathology, Insulin Resistance, Lipid Metabolism drug effects, Male, Mice, Mice, Inbred C57BL, Molecular Sequence Data, Phosphodiesterase Inhibitors pharmacology, Receptors, Adrenergic, beta metabolism, Triglycerides biosynthesis, Calcineurin deficiency, Cyclic Nucleotide Phosphodiesterases, Type 3 metabolism, Cyclic Nucleotide Phosphodiesterases, Type 4 metabolism, Hyperlipidemias enzymology, Signal Transduction drug effects
- Abstract
Insulin resistance, hyperlipidemia, and cardiovascular complications are common dysregulations of metabolic syndrome. Transplant patients treated with immunosuppressant drugs such as cyclosporine A (CsA), an inhibitor of calcineurin phosphatase, frequently develop similar metabolic complications. Although calcineurin is known to mediate insulin sensitivity by regulating β-cell growth and adipokine gene transcription, its role in lipid homeostasis is poorly understood. Here, we examined lipid homeostasis in mice lacking calcineurin Aβ (CnAβ(-/-)). We show that mice lacking calcineurin Aβ are hyperlipidemic and develop age-dependent insulin resistance. Hyperlipidemia found in CnAβ(-/-) mice is, in part, due to increased lipolysis in adipose tissues, a process mediated by β-adrenergic G-protein-coupled receptor signaling pathways. CnAβ(-/-) mice also exhibit additional pathophysiological phenotypes caused by the potentiated GPCR signaling pathways. A cell autonomous mechanism with sustained cAMP/PKA activation is found in CnAβ(-/-) mice or upon CsA treatment to inhibit calcineurin. Increased PKA activation and cAMP accumulation in CnAβ(-/-) mice, however, are sensitive to phosphodiesterase inhibitor. Indeed, we show that calcineurin regulates degradation of phosphodiesterase 3B, in addition to phosphodiesterase 4D. These results establish a role for calcineurin in lipid homeostasis. These data also indicate that potentiated cAMP signaling pathway may provide an alternative molecular pathogenesis for the metabolic complications elicited by CsA in transplant patients.
- Published
- 2013
- Full Text
- View/download PDF
19. Scavenger receptor class B type I regulates cellular cholesterol metabolism and cell signaling associated with breast cancer development.
- Author
-
Danilo C, Gutierrez-Pajares JL, Mainieri MA, Mercier I, Lisanti MP, and Frank PG
- Subjects
- Animals, Breast Neoplasms genetics, Breast Neoplasms pathology, CD36 Antigens genetics, Cell Line, Tumor, Cell Movement drug effects, Cell Proliferation, Cholesterol pharmacology, Cholesterol, HDL metabolism, Cholesterol, HDL pharmacology, Disease Models, Animal, Enzyme Activation drug effects, Female, Gene Knockdown Techniques, Heterografts, Humans, Lipoproteins, HDL genetics, Lipoproteins, HDL metabolism, MCF-7 Cells, Mitogen-Activated Protein Kinase 1, Mitogen-Activated Protein Kinase 3, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Receptors, Lipoprotein genetics, Receptors, Lipoprotein metabolism, Tumor Burden genetics, Breast Neoplasms metabolism, CD36 Antigens metabolism, Cell Transformation, Neoplastic, Cholesterol metabolism, Signal Transduction
- Abstract
Introduction: Previous studies have identified cholesterol as an important regulator of breast cancer development. High-density lipoprotein (HDL) and its cellular receptor, the scavenger receptor class B type I (SR-BI) have both been implicated in the regulation of cellular cholesterol homeostasis, but their functions in cancer remain to be established., Methods: In the present study, we have examined the role of HDL and SR-BI in the regulation of cellular signaling pathways in breast cancer cell lines and in the development of tumor in a mouse xenograft model., Results: Our data show that HDL is capable of stimulating migration and can activate signal transduction pathways in the two human breast cancer cell lines, MDA-MB-231 and MCF7. Furthermore, we also show that knockdown of the HDL receptor, SR-BI, attenuates HDL-induced activation of the phosphatidylinositol 3-kinase (PI3K)/protein Kinase B (Akt) pathway in both cell lines. Additional investigations show that inhibition of the PI3K pathway, but not that of the mitogen-activated protein kinase (MAPK) pathway, could lead to a reduction in cellular proliferation in the absence of SR-BI. Importantly, whereas the knockdown of SR-BI led to decreased proliferation and migration in vitro, it also led to a significant reduction in tumor growth in vivo. Most important, we also show that pharmacological inhibition of SR-BI can attenuate signaling and lead to decreased cellular proliferation in vitro. Taken together, our data indicate that both cholesteryl ester entry via HDL-SR-BI and Akt signaling play an essential role in the regulation of cellular proliferation and migration, and, eventually, tumor growth., Conclusions: These results identify SR-BI as a potential target for the treatment of breast cancer.
- Published
- 2013
- Full Text
- View/download PDF
20. Cholesterol and breast cancer development.
- Author
-
Danilo C and Frank PG
- Subjects
- Animals, Breast Neoplasms drug therapy, Cell Movement, Cell Proliferation, Cholesterol metabolism, Drug Design, Female, Humans, Mice, Molecular Targeted Therapy, Neoplasms, Experimental, Signal Transduction, Antineoplastic Agents pharmacology, Breast Neoplasms pathology, Cholesterol blood
- Abstract
Breast cancer is the most commonly occurring type of cancer in the world. Among the environmental factors believed to be responsible for this phenomenon, cholesterol has recently received considerable attention. Epidemiologic studies have provided inconclusive results, indicating that there may be a relationship between abnormal plasma cholesterol levels and breast cancer risk. However, more compelling evidence has been obtained in laboratory studies, and they indicate that cholesterol is capable of regulating proliferation, migration, and signaling pathways in breast cancer. In vivo studies have also indicated that plasma cholesterol levels can regulate tumor growth in mouse models. The recognition of cholesterol as a factor contributing to breast cancer development identifies cholesterol and its metabolism as novel targets for cancer therapy., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
21. Atherosclerosis, caveolae and caveolin-1.
- Author
-
Pavlides S, Gutierrez-Pajares JL, Danilo C, Lisanti MP, and Frank PG
- Subjects
- Animals, Endothelial Cells metabolism, Endothelial Cells pathology, Humans, Macrophages metabolism, Muscle, Smooth, Vascular metabolism, Muscle, Smooth, Vascular pathology, Atherosclerosis metabolism, Atherosclerosis pathology, Caveolae metabolism, Caveolin 1 chemistry, Caveolin 1 deficiency, Caveolin 1 metabolism
- Abstract
Atherosclerosis is a disease of the blood vessel characterized by the development of an arterial occlusion containing lipid and cellular deposits. Caveolae are 50-100 nm cell surface plasma membrane invaginations that are believed to play an important role in the regulation of cellular signaling and transport of molecules among others. These organelles are enriched in sphingolipids and cholesterol and are characterized by the presence of the protein caveolin-1. Caveolin-1 and caveolae are present in most of the cells involved in the development of atherosclerosis. The current literature suggests a rather complex role for caveolin-1 in this disease, with evidence of either pro- or anti-atherogenic functions depending on the cell type examined. In the present chapter, the various roles of caveolae and caveolin-1 in the development of atherosclerosis are examined.
- Published
- 2012
- Full Text
- View/download PDF
22. Alterations in membrane caveolae and BKCa channel activity in skin fibroblasts in Smith-Lemli-Opitz syndrome.
- Author
-
Ren G, Jacob RF, Kaulin Y, Dimuzio P, Xie Y, Mason RP, Tint GS, Steiner RD, Roullet JB, Merkens L, Whitaker-Menezes D, Frank PG, Lisanti MP, Cox RH, and Tulenko TN
- Subjects
- Caveolin 1 metabolism, Cells, Cultured, Dehydrocholesterols chemistry, Genotype, Humans, Immunoblotting, Membranes, Artificial, Microscopy, Electron, Molecular Structure, Skin cytology, Sterols metabolism, X-Ray Diffraction, Caveolae metabolism, Dehydrocholesterols metabolism, Fibroblasts metabolism, Large-Conductance Calcium-Activated Potassium Channel alpha Subunits metabolism, Smith-Lemli-Opitz Syndrome metabolism
- Abstract
The Smith-Lemli-Opitz syndrome (SLOS) is an inherited disorder of cholesterol synthesis caused by mutations in DHCR7 which encodes the final enzyme in the cholesterol synthesis pathway. The immediate precursor to cholesterol synthesis, 7-dehydrocholesterol (7-DHC) accumulates in the plasma and cells of SLOS patients which has led to the idea that the accumulation of abnormal sterols and/or reduction in cholesterol underlies the phenotypic abnormalities of SLOS. We tested the hypothesis that 7-DHC accumulates in membrane caveolae where it disturbs caveolar bilayer structure-function. Membrane caveolae from skin fibroblasts obtained from SLOS patients were isolated and found to accumulate 7-DHC. In caveolar-like model membranes containing 7-DHC, subtle, but complex alterations in intermolecular packing, lipid order and membrane width were observed. In addition, the BK(Ca) K(+) channel, which co-migrates with caveolin-1 in a membrane fraction enriched with cholesterol, was impaired in SLOS cells as reflected by reduced single channel conductance and a 50 mV rightward shift in the channel activation voltage. In addition, a marked decrease in BK(Ca) protein but not mRNA expression levels was seen suggesting post-translational alterations. Accompanying these changes was a reduction in caveolin-1 protein and mRNA levels, but membrane caveolar structure was not altered. These results are consistent with the hypothesis that 7-DHC accumulation in the caveolar membrane results in defective caveolar signaling. However, additional cellular alterations beyond mere changes associated with abnormal sterols in the membrane likely contribute to the pathogenesis of SLOS., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
23. Caveolin-2-deficient mice show increased sensitivity to endotoxemia.
- Author
-
de Almeida CJ, Witkiewicz AK, Jasmin JF, Tanowitz HB, Sotgia F, Frank PG, and Lisanti MP
- Subjects
- Animals, Caveolin 1 deficiency, Caveolin 1 genetics, Caveolin 2 genetics, Chemokines blood, Chemokines immunology, Cytokines blood, Cytokines immunology, Endotoxemia chemically induced, Endotoxemia mortality, Epithelial Cells metabolism, Epithelial Cells ultrastructure, Intestinal Mucosa cytology, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Lipopolysaccharides immunology, Lipopolysaccharides pharmacology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Nitric Oxide metabolism, Nitric Oxide Synthase Type II metabolism, Nitrites metabolism, Permeability, Caveolin 2 deficiency, Endotoxemia immunology
- Abstract
Caveolin proteins are structural components of caveolae and are involved in the regulation of many biological processes. Recent studies have shown that caveolin-1 modulates inflammatory responses and is important for sepsis development. In the present study, we show that caveolin-1 and caveolin-2 have opposite roles in lipopolysaccharide (LPS)-induced sepsis using caveolin-deficient (Cav-1 (-/-) and Cav-2 (-/-) ) mice for each of these proteins. While Cav-1 (-/-) mice displayed delayed mortality following challenge with LPS, Cav-2 (-/-) mice were more sensitive to LPS compared to wild-type (WT). With Cav-2 (-/-) mice, this effect was associated with increased intestinal injury and increased intestinal permeability. This negative outcome was also correlated with enhanced expression of iNOS in epithelial intestinal cells, and enhanced production of nitric oxide (NO). By contrast, Cav-1 (-/-) mice demonstrated a decrease in iNOS expression with decreased NO production, but no alteration in intestinal permeability. The differential expression of iNOS was associated with a significant increase of STAT-1 activation in these mice. Intestinal cells of Cav-2 (-/-) mice showed increased phosphorylation of STAT-1 at tyrosine 701 compared to wild-type. However, Cav-1 (-/-) mice-derived intestinal cells showed decreased levels of phosphorylation of STAT-1 at tyrosine 701. Since caveolin-2 is almost completely absent in Cav-1 (-/-) mice, we conclude that it is not just the absence of caveolin-2 that is responsible for the observed effects, but that the balance between caveolin-1 and caveolin-2 is important for iNOS expression and ultimately for sepsis outcome.
- Published
- 2011
- Full Text
- View/download PDF
24. Role of cholesterol in the development and progression of breast cancer.
- Author
-
Llaverias G, Danilo C, Mercier I, Daumer K, Capozza F, Williams TM, Sotgia F, Lisanti MP, and Frank PG
- Subjects
- 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, Breast Neoplasms etiology, Cell Transformation, Neoplastic, Cholesterol adverse effects, Diet adverse effects, Mammary Neoplasms, Experimental etiology
- 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., (Copyright © 2011 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
25. A Western-type diet accelerates tumor progression in an autochthonous mouse model of prostate cancer.
- Author
-
Llaverias G, Danilo C, Wang Y, Witkiewicz AK, Daumer K, Lisanti MP, and Frank PG
- Subjects
- Adenocarcinoma blood, Adenocarcinoma etiology, Adenocarcinoma metabolism, Animals, Cell Proliferation drug effects, Dietary Fats adverse effects, Dietary Fats pharmacology, Disease Models, Animal, Disease Progression, Lung Neoplasms secondary, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Prostatic Neoplasms blood, Prostatic Neoplasms etiology, Prostatic Neoplasms metabolism, Tumor Burden, Western World, Adenocarcinoma pathology, Diet adverse effects, Prostatic Neoplasms pathology
- Abstract
Epidemiological studies have provided evidence suggesting an important role for diet and obesity in the development of cancer. Specifically, lipid nutrients of the diet have been identified as important regulators of tumor development and progression. In the present study, we have examined the role of dietary fat and cholesterol in the initiation and progression of prostate cancer using the well-characterized TRAMP mouse model. Consumption of a Western-type diet--that is, enriched in both fat and cholesterol--accelerated prostate tumor incidence and tumor burden compared to mice fed a control chow diet. Furthermore, we also show that this diet increased the extent and the histological grade of prostate tumors. These findings were confirmed by the presence of increased levels of protein markers of advanced tumors in prostates obtained from animals fed a Western-type diet compared to those obtained from control animals. Increased lung metastases in animals fed a Western-type diet were also observed. In addition, we found that with a Western diet, animals bearing tumors presented with reduced plasma cholesterol levels compared with animals fed a control diet. Finally, we show that tumors obtained from animals fed a Western-type diet displayed increased expression of the high-density lipoprotein receptor SR-BI and increased angiogenesis. Taken together, our data suggest that dietary fat and cholesterol play an important role in the development of prostate cancer.
- Published
- 2010
- Full Text
- View/download PDF
26. Celecoxib combined with atorvastatin prevents progression of atherosclerosis.
- Author
-
Raval M, Frank PG, Laury-Kleintop L, Yan G, and Lanza-Jacoby S
- Subjects
- Animals, Apolipoproteins E physiology, Atorvastatin, Celecoxib, Chemokine CCL2 blood, Cyclooxygenase 2 blood, Dinoprostone blood, Disease Progression, Drug Therapy, Combination, Immunohistochemistry, Male, Matrix Metalloproteinase 9 blood, Mice, Mice, Inbred C57BL, Vascular Cell Adhesion Molecule-1 genetics, Atherosclerosis drug therapy, Cyclooxygenase 2 Inhibitors administration & dosage, Heptanoic Acids administration & dosage, Hydroxymethylglutaryl-CoA Reductase Inhibitors administration & dosage, Pyrazoles administration & dosage, Pyrroles administration & dosage, Sulfonamides administration & dosage
- Abstract
Background: Increased expression of cyclooxygenase (COX-2) contributes to atherosclerosis. Recent studies suggest that COX-2 inhibitors prevent early plaque development but their effects on established lesions are less clear, while the statins promote plaque stability. The purpose of this study is to investigate whether administering a combination of a COX-2 inhibitor with a statin drug alters plaque progression in apo E-/- mice., Materials and Methods: Apo E-/- mice were fed a Western diet from 6 to 26 wk of age. At 26 wk, the Western diets supplemented with atorvastatin, celecoxib, or atorvastatin plus celecoxib were given for an additional 12 wk., Results: When the mice were 38 wk of age, the total area occupied by the atherosclerotic lesion was 53% less in the mice fed the combination of atorvastatin + celecoxib P ≤ 0.05) than that of the apo E-/- mice fed the Western diet alone, atorvastatin alone, or celecoxib alone. The decreased extent of atherosclerosis observed in the apo E-/- mice fed the combination of drugs was associated with reduced levels of prostaglandin (PG) E(2,) decreased protein expression of metalloproteinase (MMP)-9, macrophage chemotactic protein (MCP-1), and COX 2, and decreased staining for MMP-9, F4-80 (a marker for macrophages), and vascular cell adhesion molecule (VCAM)., Conclusion: This study indicates that using statins with a COX-2 inhibitor reduced the extent of atherosclerosis and inflammatory/cell adhesion molecule levels in the apo E-/- mouse model., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
27. The autophagic tumor stroma model of cancer: Role of oxidative stress and ketone production in fueling tumor cell metabolism.
- Author
-
Pavlides S, Tsirigos A, Migneco G, Whitaker-Menezes D, Chiavarina B, Flomenberg N, Frank PG, Casimiro MC, Wang C, Pestell RG, Martinez-Outschoorn UE, Howell A, Sotgia F, and Lisanti MP
- Subjects
- Animals, Arginine metabolism, Biomarkers analysis, Biomarkers blood, Breast Neoplasms diagnosis, Caveolin 1 genetics, Caveolin 1 metabolism, Female, Gene Expression Profiling, Humans, Mice, MicroRNAs metabolism, Models, Biological, Stromal Cells metabolism, 3-Hydroxybutyric Acid metabolism, Arginine analogs & derivatives, Autophagy, Breast Neoplasms metabolism, Oxidative Stress
- Abstract
A loss of stromal Cav-1 in the tumor fibroblast compartment is associated with early tumor recurrence, lymph-node metastasis, and tamoxifen-resistance, resulting in poor clinical outcome in breast cancer patients. Here, we have used Cav-1 (-/-) null mice as a pre-clinical model for this "lethal tumor micro-environment." Metabolic profiling of Cav-1 (-/-) mammary fat pads revealed the upregulation of numerous metabolites (nearly 100), indicative of a major catabolic phenotype. Our results are consistent with the induction of oxidative stress, mitochondrial dysfunction, and autophagy/mitophagy. The two most prominent metabolites that emerged from this analysis were ADMA (asymmetric dimethyl arginine) and BHB (beta-hydroxybutyrate; a ketone body), which are markers of oxidative stress and mitochondrial dysfunction, respectively. Transcriptional profiling of Cav-1 (-/-) stromal cells and human tumor stroma from breast cancer patients directly supported an association with oxidative stress, mitochondrial dysfunction, and autophagy/mitophagy, as well as ADMA and ketone production. MircoRNA profiling of Cav-1 (-/-) stromal cells revealed the upregulation of two key cancer-related miR's, namely miR-31 and miR-34c. Consistent with our metabolic findings, these miR's are associated with oxidative stress (miR-34c) or activation of the hypoxic response/HIF1a (miR-31), which is sufficient to drive authophagy/mitophagy. Thus, via an unbiased comprehensive analysis of a lethal tumor micro-environment, we have identified a number of candidate biomarkers (ADMA, ketones, and miR-31/34c) that could be used to identify high-risk cancer patients at diagnosis, for treatment stratification and/or for evaluating therapeutic efficacy during anti-cancer therapy. We propose that the levels of these key biomarkers (ADMA, ketones/BHB, miR-31, and miR-34c) could be (1) assayed using serum or plasma from cancer patients, or (2) performed directly on excised tumor tissue. Importantly, induction of oxidative stress and autophagy/mitophagy in the tumor stromal compartment provides a means by which epithelial cancer cells can directly "feed off" of stromal-derived essential nutrients, chemical building blocks (amino acids, nucleotides), and energy-rich metabolites (glutamine, pyruvate, ketones/BHB), driving tumor progression and metastasis. Essentially, aggressive cancer cells are "eating" the cancer-associated fibroblasts via autophagy/mitophagy in the tumor micro-environment. Lastly, we discuss that this "Autophagic Tumor Stroma Model of Cancer Metabolism" provides a viable solution to the "Autophagy Paradox" in cancer etiology and chemo-therapy.
- Published
- 2010
- Full Text
- View/download PDF
28. Evolutionarily conserved role of calcineurin in phosphodegron-dependent degradation of phosphodiesterase 4D.
- Author
-
Zhu H, Suk HY, Yu RY, Brancho D, Olabisi O, Yang TT, Yang X, Zhang J, Moussaif M, Durand JL, Jelicks LA, Kim JY, Scherer PE, Frank PG, Lisanti MP, Calvert JW, Duranski MR, Lefer DJ, Huston E, Baillie GS, Houslay MD, Molkentin JD, Jin J, and Chow CW
- Subjects
- Amino Acid Motifs, Animals, Caenorhabditis elegans physiology, Calcineurin Inhibitors, Cell Line, Cyclic AMP metabolism, Cyclic Nucleotide Phosphodiesterases, Type 4 genetics, Cyclosporine metabolism, Enzyme Inhibitors metabolism, Gene Expression Regulation, Enzymologic, Humans, Mice, Mice, Knockout, Proteasome Endopeptidase Complex metabolism, Ubiquitin metabolism, Caenorhabditis elegans Proteins genetics, Caenorhabditis elegans Proteins metabolism, Calcineurin genetics, Calcineurin metabolism, Cyclic Nucleotide Phosphodiesterases, Type 4 metabolism, Evolution, Molecular, Second Messenger Systems physiology
- Abstract
Calcineurin is a widely expressed and highly conserved Ser/Thr phosphatase. Calcineurin is inhibited by the immunosuppressant drug cyclosporine A (CsA) or tacrolimus (FK506). The critical role of CsA/FK506 as an immunosuppressant following transplantation surgery provides a strong incentive to understand the phosphatase calcineurin. Here we uncover a novel regulatory pathway for cyclic AMP (cAMP) signaling by the phosphatase calcineurin which is also evolutionarily conserved in Caenorhabditis elegans. We found that calcineurin binds directly to and inhibits the proteosomal degradation of cAMP-hydrolyzing phosphodiesterase 4D (PDE4D). We show that ubiquitin conjugation and proteosomal degradation of PDE4D are controlled by a cullin 1-containing E(3) ubiquitin ligase complex upon dual phosphorylation by casein kinase 1 (CK1) and glycogen synthase kinase 3beta (GSK3beta) in a phosphodegron motif. Our findings identify a novel signaling process governing G-protein-coupled cAMP signal transduction-opposing actions of the phosphatase calcineurin and the CK1/GSK3beta protein kinases on the phosphodegron-dependent degradation of PDE4D. This novel signaling system also provides unique functional insights into the complications elicited by CsA in transplant patients.
- Published
- 2010
- Full Text
- View/download PDF
29. Ketones and lactate "fuel" tumor growth and metastasis: Evidence that epithelial cancer cells use oxidative mitochondrial metabolism.
- Author
-
Bonuccelli G, Tsirigos A, Whitaker-Menezes D, Pavlides S, Pestell RG, Chiavarina B, Frank PG, Flomenberg N, Howell A, Martinez-Outschoorn UE, Sotgia F, and Lisanti MP
- Subjects
- 3-Hydroxybutyric Acid metabolism, Animals, Autophagy, Breast Neoplasms pathology, Cell Line, Tumor, Cell Movement, Female, Glycolysis, Humans, Lung Neoplasms pathology, Lung Neoplasms secondary, Mice, Oxidative Phosphorylation, Stromal Cells metabolism, Transplantation, Heterologous, Up-Regulation, 3-Hydroxybutyric Acid pharmacology, Breast Neoplasms metabolism, Lactic Acid pharmacology, Mitochondria metabolism
- Abstract
Previously, we proposed a new model for understanding the "Warburg effect" in tumor metabolism. In this scheme, cancer-associated fibroblasts undergo aerobic glycolysis and the resulting energy-rich metabolites are then transferred to epithelial cancer cells, where they enter the TCA cycle, resulting in high ATP production via oxidative phosphorylation. We have termed this new paradigm "The Reverse Warburg Effect." Here, we directly evaluate whether the end-products of aerobic glycolysis (3-hydroxy-butyrate and L-lactate) can stimulate tumor growth and metastasis, using MDA-MB-231 breast cancer xenografts as a model system. More specifically, we show that administration of 3-hydroxy-butyrate (a ketone body) increases tumor growth by ∼2.5-fold, without any measurable increases in tumor vascularization/angiogenesis. Both 3-hydroxy-butyrate and L-lactate functioned as chemo-attractants, stimulating the migration of epithelial cancer cells. Although L-lactate did not increase primary tumor growth, it stimulated the formation of lung metastases by ∼10-fold. Thus, we conclude that ketones and lactate fuel tumor growth and metastasis, providing functional evidence to support the "Reverse Warburg Effect". Moreover, we discuss the possibility that it may be unwise to use lactate-containing i.v. solutions (such as Lactated Ringer's or Hartmann's solution) in cancer patients, given the dramatic metastasis-promoting properties of L-lactate. Also, we provide evidence for the up-regulation of oxidative mitochondrial metabolism and the TCA cycle in human breast cancer cells in vivo, via an informatics analysis of the existing raw transcriptional profiles of epithelial breast cancer cells and adjacent stromal cells. Lastly, our findings may explain why diabetic patients have an increased incidence of cancer, due to increased ketone production, and a tendency towards autophagy/mitophagy in their adipose tissue.
- Published
- 2010
- Full Text
- View/download PDF
30. Endothelial caveolae and caveolin-1 as key regulators of atherosclerosis.
- Author
-
Frank PG
- Subjects
- Animals, Caveolin 1 genetics, Humans, Mice, Mice, Knockout, Atherosclerosis metabolism, Atherosclerosis pathology, Caveolae metabolism, Caveolin 1 metabolism, Endothelial Cells metabolism, Endothelial Cells ultrastructure
- Abstract
This commentary discusses the role of caveolin-1 in atherosclerosis.
- Published
- 2010
- Full Text
- View/download PDF
31. Loss of stromal caveolin-1 leads to oxidative stress, mimics hypoxia and drives inflammation in the tumor microenvironment, conferring the "reverse Warburg effect": a transcriptional informatics analysis with validation.
- Author
-
Pavlides S, Tsirigos A, Vera I, Flomenberg N, Frank PG, Casimiro MC, Wang C, Fortina P, Addya S, Pestell RG, Martinez-Outschoorn UE, Sotgia F, and Lisanti MP
- Subjects
- Animals, Caveolin 1 antagonists & inhibitors, Caveolin 1 genetics, Cell Hypoxia, Electron Transport Complex I metabolism, Glycolysis, Hypoxia-Inducible Factor 1 metabolism, Mice, Mice, Knockout, Models, Biological, NF-kappa B metabolism, Nitric Oxide metabolism, RNA Interference, RNA, Small Interfering metabolism, Stromal Cells cytology, Caveolin 1 metabolism, Gene Expression Profiling, Inflammation Mediators metabolism, Oxidative Stress, Stromal Cells metabolism, Tumor Microenvironment immunology
- Abstract
Cav-1 (-/-) deficient stromal cells are a new genetic model for myofibroblasts and cancer-associated fibroblasts. Using an unbiased informatics analysis of the transcriptional profile of Cav-1 (-/-) deficient mesenchymal stromal cells, we have now identified many of the major signaling pathways that are activated by a loss of Cav-1, under conditions of metabolic restriction (with low glucose media). Our informatics analysis suggests that a loss of Cav-1 induces oxidative stress, which mimics a constitutive pseudo-hypoxic state, leading to (1) aerobic glycolysis and (2) inflammation in the tumor stromal microenvironment. This occurs via the activation of two major transcription factors, namely HIF (aerobic glycolysis) and NFκB (inflammation) in Cav-1 (-/-) stromal fibroblastic cells. Experimentally, we show that Cav-1 deficient stromal cells may possess defective mitochondria, due to the over-production of nitric oxide (NO), resulting in the tyrosine nitration of the mitochondrial respiratory chain components (such as complex I). Elevated levels of nitro-tyrosine were observed both in Cav-1 (-/-) stromal cells, and via acute knock-down with siRNA targeting Cav-1. Finally, metabolic restriction with mitochondrial (complex I) and glycolysis inhibitors was synthetically lethal with a Cav-1 (-/-) deficiency in mice. As such, Cav-1 deficient mice show a dramatically reduced mitochondrial reserve capacity. Thus, a mitochondrial defect in Cav-1 deficient stromal cells could drive oxidative stress, leading to aerobic glycolysis, and inflammation, in the tumor microenvironment. These stromal alterations may underlie the molecular basis of the "reverse Warburg effect", and could provide the key to targeted anti-cancer therapies using metabolic inhibitors. In direct support of these findings, the transcriptional profile of Cav-1 (-/-) stromal cells overlaps significantly with Alzheimer disease, which is characterized by oxidative stress, NO over-production (peroxynitrite formation), inflammation, hypoxia and mitochondrial dysfunction. We conclude that Cav-1 (-/-) deficient mice are a new whole-body animal model for an activated lethal tumor microenvironment, i.e., "tumor stroma" without the tumor. Since Cav-1 (-/-) mice are also an established animal model for profibrotic disease, our current results may have implications for understanding the pathogenesis of scleroderma (systemic sclerosis) and pulmonary fibrosis, which are also related to abnormal mesenchymal stem cell function.
- Published
- 2010
- Full Text
- View/download PDF
32. The reverse Warburg effect: glycolysis inhibitors prevent the tumor promoting effects of caveolin-1 deficient cancer associated fibroblasts.
- Author
-
Bonuccelli G, Whitaker-Menezes D, Castello-Cros R, Pavlides S, Pestell RG, Fatatis A, Witkiewicz AK, Vander Heiden MG, Migneco G, Chiavarina B, Frank PG, Capozza F, Flomenberg N, Martinez-Outschoorn UE, Sotgia F, and Lisanti MP
- Subjects
- 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, Caveolin 1 deficiency, Fibroblasts metabolism, Glycolysis drug effects, Lactate Dehydrogenases metabolism, Pyruvate Kinase metabolism
- 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
- Full Text
- View/download PDF
33. Transcriptional evidence for the "Reverse Warburg Effect" in human breast cancer tumor stroma and metastasis: similarities with oxidative stress, inflammation, Alzheimer's disease, and "Neuron-Glia Metabolic Coupling".
- Author
-
Pavlides S, Tsirigos A, Vera I, Flomenberg N, Frank PG, Casimiro MC, Wang C, Pestell RG, Martinez-Outschoorn UE, Howell A, Sotgia F, and Lisanti MP
- Subjects
- Alzheimer Disease genetics, Alzheimer Disease metabolism, Animals, Caveolin 1 deficiency, Caveolin 1 genetics, Female, Gene Expression Profiling, Humans, Lymphatic Metastasis genetics, Lymphatic Metastasis physiopathology, Mice, Mice, Knockout, Neuroglia metabolism, Neurons metabolism, Oxidative Stress, Proteomics, Stromal Cells metabolism, Breast Neoplasms genetics, Breast Neoplasms metabolism, Models, Biological
- Abstract
Caveolin-1 (-/-) null stromal cells are a novel genetic model for cancer-associated fibroblasts and myofibroblasts. Here, we used an unbiased informatics analysis of transcriptional gene profiling to show that Cav-1 (-/-) bone-marrow derived stromal cells bear a striking resemblance to the activated tumor stroma of human breast cancers. More specifically, the transcriptional profiles of Cav-1 (-/-) stromal cells were most closely related to the primary tumor stroma of breast cancer patients that had undergone lymph-node (LN) metastasis. This is consistent with previous morphological data demonstrating that a loss of stromal Cav-1 protein (by immuno-histochemical staining in the fibroblast compartment) is significantly associated with increased LN-metastasis. We also provide evidence that the tumor stroma of human breast cancers shows a transcriptional shift towards oxidative stress, DNA damage/repair, inflammation, hypoxia, and aerobic glycolysis, consistent with the "Reverse Warburg Effect". Finally, the tumor stroma of "metastasis-prone" breast cancer patients was most closely related to the transcriptional profiles derived from the brains of patients with Alzheimer's disease. This suggests that certain fundamental biological processes are common to both an activated tumor stroma and neuro-degenerative stress. These processes may include oxidative stress, NO over-production (peroxynitrite formation), inflammation, hypoxia, and mitochondrial dysfunction, which are thought to occur in Alzheimer?s disease pathology. Thus, a loss of Cav-1 expression in cancer-associated myofibroblasts may be a protein biomarker for oxidative stress, aerobic glycolysis, and inflammation, driving the "Reverse Warburg Effect" in the tumor micro-environment and cancer cell metastasis.
- Published
- 2010
- Full Text
- View/download PDF
34. Endothelial cells isolated from caveolin-2 knockout mice display higher proliferation rate and cell cycle progression relative to their wild-type counterparts.
- Author
-
Xie L, Frank PG, Lisanti MP, and Sowa G
- Subjects
- Animals, Caveolin 2 genetics, Cells, Cultured, Cyclin A metabolism, Cyclin B1 metabolism, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Cyclin-Dependent Kinase Inhibitor p27, Flow Cytometry, Intracellular Signaling Peptides and Proteins metabolism, Lung blood supply, Mice, Mice, Knockout, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, Phosphorylation, Retinoblastoma Protein metabolism, Signal Transduction, Time Factors, Caveolin 2 deficiency, Cell Cycle, Cell Proliferation, Endothelial Cells metabolism
- Abstract
The goal of this study was to determine whether caveolin-2 (Cav-2) is capable of controlling endothelial cell (EC) proliferation in vitro. To realize this goal, we have directly compared proliferation rates and cell cycle-associated signaling proteins between lung ECs isolated from wild-type (WT) and Cav-2 knockout (KO) mice. Using three independent proliferation assays, we have determined that Cav-2 KO ECs proliferate by ca. 2-fold faster than their WT counterparts. Cell cycle analysis by flow cytometry of propidium iodide-stained cells showed a relatively higher percentage of Cav-2 KO ECs in S and G(2)/M and lower percentage in G(o)/G(1) phases of cell cycle relative to their WT counterparts. Furthermore, an over 2-fold increase in the percentage of S phase-associated Cav-2 KO relative to WT ECs was independently determined with bromodeoxyuridine incorporation assay. Mechanistically, the increase in proliferation/cell cycle progression of Cav-2 KO ECs correlated well with elevated expression levels of predominantly S phase- and G(2)/M phase-associated cyclin A and B1, respectively. Further mechanistic analysis of molecular events controlling cell cycle progression revealed increased level of hyperphosphorylated (inactive) form of G(1) to S phase transition inhibitor, the retinoblastoma protein in hyperproliferating Cav-2 KO ECs. Conversely, the expression level of the two cyclin-dependent kinase inhibitors p16(INK4) and p27(Kip1) was reduced in Cav-2 KO ECs. Finally, increased phosphorylation (activation) of proproliferative extracellular signal-regulated kinase 1/2 was observed in hyperproliferating Cav-2 KO ECs. Overall, our data suggest that Cav-2 negatively regulates lung EC proliferation and cell cycle progression.
- Published
- 2010
- Full Text
- View/download PDF
35. The reverse Warburg effect: aerobic glycolysis in cancer associated fibroblasts and the tumor stroma.
- Author
-
Pavlides S, Whitaker-Menezes D, Castello-Cros R, Flomenberg N, Witkiewicz AK, Frank PG, Casimiro MC, Wang C, Fortina P, Addya S, Pestell RG, Martinez-Outschoorn UE, Sotgia F, and Lisanti MP
- Subjects
- Animals, Annexins metabolism, Caveolin 1 deficiency, Caveolin 1 genetics, Caveolin 1 metabolism, Female, Humans, L-Lactate Dehydrogenase metabolism, Mice, Mice, Knockout, Proteomics, Pyruvate Kinase metabolism, Stromal Cells metabolism, Tenascin metabolism, Up-Regulation, Breast Neoplasms metabolism, Fibroblasts metabolism, Glycolysis
- Abstract
Here, we propose a new model for understanding the Warburg effect in tumor metabolism. Our hypothesis is that epithelial cancer cells induce the Warburg effect (aerobic glycolysis) in neighboring stromal fibroblasts. These cancer-associated fibroblasts, then undergo myo-fibroblastic differentiation, and secrete lactate and pyruvate (energy metabolites resulting from aerobic glycolysis). Epithelial cancer cells could then take up these energy-rich metabolites and use them in the mitochondrial TCA cycle, thereby promoting efficient energy production (ATP generation via oxidative phosphorylation), resulting in a higher proliferative capacity. In this alternative model of tumorigenesis, the epithelial cancer cells instruct the normal stroma to transform into a wound-healing stroma, providing the necessary energy-rich micro-environment for facilitating tumor growth and angiogenesis. In essence, the fibroblastic tumor stroma would directly feed the epithelial cancer cells, in a type of host-parasite relationship. We have termed this new idea the "Reverse Warburg Effect." In this scenario, the epithelial tumor cells "corrupt" the normal stroma, turning it into a factory for the production of energy-rich metabolites. This alternative model is still consistent with Warburg's original observation that tumors show a metabolic shift towards aerobic glycolysis. In support of this idea, unbiased proteomic analysis and transcriptional profiling of a new model of cancer-associated fibroblasts (caveolin-1 (Cav-1) deficient stromal cells), shows the upregulation of both (1) myo-fibroblast markers and (2) glycolytic enzymes, under normoxic conditions. We validated the expression of these proteins in the fibroblastic stroma of human breast cancer tissues that lack stromal Cav-1. Importantly, a loss of stromal Cav-1 in human breast cancers is associated with tumor recurrence, metastasis, and poor clinical outcome. Thus, an absence of stromal Cav-1 may be a biomarker for the "Reverse Warburg Effect," explaining its powerful predictive value.
- Published
- 2009
- Full Text
- View/download PDF
36. Clinical and translational implications of the caveolin gene family: lessons from mouse models and human genetic disorders.
- Author
-
Mercier I, Jasmin JF, Pavlides S, Minetti C, Flomenberg N, Pestell RG, Frank PG, Sotgia F, and Lisanti MP
- Subjects
- Adult Stem Cells metabolism, Animals, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Cardiomyopathies metabolism, Caveolae metabolism, Caveolins biosynthesis, Caveolins genetics, Diabetes Mellitus drug therapy, Diabetes Mellitus metabolism, Humans, Hypertension, Pulmonary drug therapy, Hypertension, Pulmonary metabolism, Male, Mice, Muscular Dystrophies drug therapy, Muscular Dystrophies metabolism, Mutation, Peptides therapeutic use, Prostatic Neoplasms drug therapy, Prostatic Neoplasms metabolism, Signal Transduction, Caveolins physiology
- Abstract
Here we review the clinical and translational implications of the caveolin gene family for understanding the pathogenesis of human diseases, including breast and prostate cancers, pulmonary hypertension, cardiomyopathy, diabetes, and muscular dystrophy. Detailed phenotypic analysis of caveolin knockout mice has served to highlight the crucial role of a caveolin deficiency in the pathogenesis of many human disease processes. Mutations in the human caveolin genes are associated with a number of established genetic disorders (such as breast cancer, lipodystrophy, muscular dystrophy, and cardiomyopathy), making the caveolins important and novel targets for drug development. The implementation of new strategies for caveolin replacement therapy-including caveolin mimetic peptides-is ongoing.
- Published
- 2009
- Full Text
- View/download PDF
37. Towards a new "stromal-based" classification system for human breast cancer prognosis and therapy.
- Author
-
Witkiewicz AK, Casimiro MC, Dasgupta A, Mercier I, Wang C, Bonuccelli G, Jasmin JF, Frank PG, Pestell RG, Kleer CG, Sotgia F, and Lisanti MP
- Subjects
- Angiogenesis Inhibitors therapeutic use, Breast Neoplasms diagnosis, Breast Neoplasms therapy, Female, Fibroblasts metabolism, Fibroblasts physiology, Humans, Prognosis, Risk, Stromal Cells metabolism, Survival Analysis, Up-Regulation, Breast Neoplasms classification, Caveolin 1 metabolism
- Abstract
Here, we discuss recent evidence that an absence of stromal Cav-1 expression in human breast cancers is a powerful single independent predictor of early disease recurrence, metastasis and poor clinical outcome. These findings have now been validated in two independent patient populations. Importantly, the predictive value of stromal Cav-1 is independent of epithelial marker status, making stromal Cav-1 a new "universal" or "widely-applicable" breast cancer prognostic marker. We propose based on the expression of stromal Cav-1, that breast cancer patients could be stratified into high-risk and low-risk groups. High-risk patients showing an absence of stromal Cav-1 should be offered more aggressive therapies, such as anti-angiogenic approaches, in addition to the standard therapy regimens. Mechanistically, loss of stromal Cav-1 is a surrogate biomarker for increased cell cycle progression, growth factor secretion, "stemness", and angiogenic potential in the tumor microenvironment. Since almost all cancers develop within the context of a stromal microenvironment, this new stromal classification system may be broadly applicable to other epithelial and non-epithelial cancer subtypes, as well as "pre-malignant" lesions (carcinoma in situ).
- Published
- 2009
- Full Text
- View/download PDF
38. Caveolin-1 (P132L), a common breast cancer mutation, confers mammary cell invasiveness and defines a novel stem cell/metastasis-associated gene signature.
- Author
-
Bonuccelli G, Casimiro MC, Sotgia F, Wang C, Liu M, Katiyar S, Zhou J, Dew E, Capozza F, Daumer KM, Minetti C, Milliman JN, Alpy F, Rio MC, Tomasetto C, Mercier I, Flomenberg N, Frank PG, Pestell RG, and Lisanti MP
- Subjects
- 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, Biomarkers, Tumor genetics, Caveolin 1 genetics, Mammary Neoplasms, Animal genetics, Mammary Neoplasms, Animal pathology, Mutation genetics, Neoplastic Stem Cells pathology
- 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.
- Published
- 2009
- Full Text
- View/download PDF
39. Genetic ablation of caveolin-1 drives estrogen-hypersensitivity and the development of DCIS-like mammary lesions.
- Author
-
Mercier I, Casimiro MC, Zhou J, Wang C, Plymire C, Bryant KG, Daumer KM, Sotgia F, Bonuccelli G, Witkiewicz AK, Lin J, Tran TH, Milliman J, Frank PG, Jasmin JF, Rui H, Pestell RG, and Lisanti MP
- Subjects
- Animals, Carcinoma, Intraductal, Noninfiltrating metabolism, Carcinoma, Intraductal, Noninfiltrating pathology, Caveolin 1 deficiency, Cell Transformation, Neoplastic genetics, Estrogen Receptor alpha genetics, Estrogen Receptor alpha metabolism, Female, Hepatocyte Nuclear Factor 3-alpha genetics, Hepatocyte Nuclear Factor 3-alpha metabolism, Humans, Immunohistochemistry, Mammary Neoplasms, Experimental metabolism, Mammary Neoplasms, Experimental pathology, Mice, Mice, Knockout, Oligonucleotide Array Sequence Analysis, Ovariectomy, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, Receptors, Progesterone genetics, Receptors, Progesterone metabolism, Tissue Array Analysis, Trans-Activators genetics, Trans-Activators metabolism, Carcinoma, Intraductal, Noninfiltrating genetics, Caveolin 1 genetics, Estrogens pharmacology, Gene Expression Profiling, Mammary Neoplasms, Experimental genetics
- Abstract
Caveolin-1 (Cav-1) loss-of-function mutations are exclusively associated with estrogen receptor-positive (ER(+)) human breast cancers. To dissect the role of Cav-1 loss-of-function in the pathogenesis of human breast cancers, we used Cav-1(-/-) null mice as a model system. First, we demonstrated that Cav-1(-/-) mammary epithelia overexpress two well-established ER co-activator genes, CAPER and Foxa1, in addition to ER-alpha. Thus, the functional loss of Cav-1 may be sufficient to confer estrogen-hypersensitivity in the mammary gland. To test this hypothesis directly, we subjected Cav-1(-/-) mice to ovariectomy and estrogen supplementation. As predicted, Cav-1(-/-) mammary glands were hyper-responsive to estrogen and developed dysplastic mammary lesions with adjacent stromal angiogenesis that resemble human ductal carcinoma in situ. Based on an extensive biomarker analysis, these Cav-1(-/-) mammary lesions contain cells that are hyperproliferative and stain positively with nucleolar (B23/nucleophosmin) and stem/progenitor cell markers (SPRR1A and beta-catenin). Genome-wide transcriptional profiling identified many estrogen-related genes that were over-expressed in Cav-1(-/-) mammary glands, including CAPER--an ER co-activator gene and putative stem/progenitor cell marker. Analysis of human breast cancer samples revealed that CAPER is overexpressed and undergoes a cytoplasmic-to-nuclear shift during the transition from pre-malignancy to ductal carcinoma in situ. Thus, Cav-1(-/-) null mice are a new preclinical model for studying the molecular paradigm of estrogen hypersensitivity and the development of estrogen-dependent ductal carcinoma in situ lesions.
- Published
- 2009
- Full Text
- View/download PDF
40. Caveolin-1-/- null mammary stromal fibroblasts share characteristics with human breast cancer-associated fibroblasts.
- Author
-
Sotgia F, Del Galdo F, Casimiro MC, Bonuccelli G, Mercier I, Whitaker-Menezes D, Daumer KM, Zhou J, Wang C, Katiyar S, Xu H, Bosco E, Quong AA, Aronow B, Witkiewicz AK, Minetti C, Frank PG, Jimenez SA, Knudsen ES, Pestell RG, and Lisanti MP
- Subjects
- Blotting, Western, Breast cytology, Breast physiology, Breast Neoplasms mortality, Cell Culture Techniques, Cell Division, Disease Progression, Disease-Free Survival, Epithelial Cells cytology, Epithelial Cells pathology, Epithelial Cells physiology, Female, Fibroblasts cytology, Fibroblasts physiology, Humans, Polymerase Chain Reaction, RNA, Messenger genetics, RNA, Neoplasm genetics, Stromal Cells cytology, Stromal Cells physiology, Survival Analysis, Breast Neoplasms genetics, Breast Neoplasms pathology, Caveolin 1 deficiency, Caveolin 1 genetics, Fibroblasts pathology, Stromal Cells pathology
- Abstract
Recently, we reported that human breast cancer-associated fibroblasts show functional inactivation of the retinoblastoma (RB) tumor suppressor and down-regulation of caveolin-1 (Cav-1) protein expression. However, it remains unknown whether loss of Cav-1 is sufficient to confer functional RB inactivation in mammary fibroblasts. To establish a direct cause-and-effect relationship, mammary stromal fibroblasts (MSFs) were prepared from Cav-1(-/-) null mice and subjected to phenotypic analysis. Here, we provide evidence that Cav-1(-/-) MSFs share many characteristics with human cancer-associated fibroblasts. The Cav-1(-/-) MSF transcriptome significantly overlaps with human cancer-associated fibroblasts; both show a nearly identical profile of RB/E2F-regulated genes that are up-regulated, which is consistent with RB inactivation. This Cav-1(-/-) MSF gene signature is predictive of poor clinical outcome in breast cancer patients treated with tamoxifen. Consistent with these findings, Cav-1(-/-) MSFs show RB hyperphosphorylation and the up-regulation of estrogen receptor co-activator genes. We also evaluated the paracrine effects of "conditioned media" prepared from Cav-1(-/-) MSFs on wild-type mammary epithelia. Our results indicate that Cav-1(-/-) MSF "conditioned media" is sufficient to induce an epithelial-mesenchymal transition, indicative of an invasive phenotype. Proteomic analysis of this "conditioned media" reveals increased levels of proliferative/angiogenic growth factors. Consistent with these findings, Cav-1(-/-) MSFs are able to undergo endothelial-like transdifferentiation. Thus, these results have important implications for understanding the role of cancer-associated fibroblasts and RB inactivation in promoting tumor angiogenesis.
- Published
- 2009
- Full Text
- View/download PDF
41. Loss of caveolin-3 induces a lactogenic microenvironment that is protective against mammary tumor formation.
- Author
-
Sotgia F, Casimiro MC, Bonuccelli G, Liu M, Whitaker-Menezes D, Er O, Daumer KM, Mercier I, Witkiewicz AK, Minetti C, Capozza F, Gormley M, Quong AA, Rui H, Frank PG, Milliman JN, Knudsen ES, Zhou J, Wang C, Pestell RG, and Lisanti MP
- Subjects
- 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, Caveolin 3 genetics, Caveolin 3 metabolism, Gene Expression, Lactation physiology, Mammary Neoplasms, Experimental genetics
- 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
- Full Text
- View/download PDF
42. Caveolae and transcytosis in endothelial cells: role in atherosclerosis.
- Author
-
Frank PG, Pavlides S, and Lisanti MP
- Subjects
- Animals, Atherosclerosis pathology, Biological Transport, Caveolae pathology, Endothelial Cells pathology, Humans, Transport Vesicles pathology, Atherosclerosis metabolism, Caveolae metabolism, Caveolin 1 metabolism, Endothelial Cells metabolism, Transport Vesicles metabolism
- Abstract
The endothelium plays an important role in the regulation of molecular exchanges between the blood and peripheral tissues. The transport of molecules between tissues must be tightly controlled in order to maintain homeostasis between the different organs of the body. The endothelial transcytosis pathway has been shown to direct the transfer of proteins and solutes and therefore to act as a filtering system. This transport mode has been demonstrated to involve plasma-membrane vesicles that may be transferred with their cargo components from the apical to the basal side of endothelial cells. Among the vesicles implicated in the regulation of transcytosis, caveolae, which are 50 to 100-nm plasma-membrane invaginations, have been reported to play an essential part. In this paper, we review the function of caveolae and their major protein component (i.e., caveolin-1) in the regulation of endothelial transcytosis. The roles of caveolae in vascular diseases, such as atherosclerosis, are discussed.
- Published
- 2009
- Full Text
- View/download PDF
43. ICAM-1: role in inflammation and in the regulation of vascular permeability.
- Author
-
Frank PG and Lisanti MP
- Subjects
- Animals, Atherosclerosis physiopathology, Humans, Leukocytes physiology, Capillary Permeability physiology, Inflammation physiopathology, Intercellular Adhesion Molecule-1 physiology
- Published
- 2008
- Full Text
- View/download PDF
44. Role of caveolin-1 in the regulation of lipoprotein metabolism.
- Author
-
Frank PG, Pavlides S, Cheung MW, Daumer K, and Lisanti MP
- Subjects
- Animals, Aorta metabolism, Biological Transport, Active, Caveolin 1 genetics, Cell Membrane metabolism, Cholesterol metabolism, Endothelial Cells metabolism, Female, Lipoproteins blood, Lipoproteins, HDL metabolism, Lipoproteins, VLDL metabolism, Liver metabolism, Mice, Mice, Knockout, Triglycerides blood, Triglycerides metabolism, Caveolin 1 physiology, Lipoproteins metabolism
- Abstract
Lipoprotein metabolism plays an important role in the development of several human diseases, including coronary artery disease and the metabolic syndrome. A good comprehension of the factors that regulate the metabolism of the various lipoproteins is therefore key to better understanding the variables associated with the development of these diseases. Among the players identified are regulators such as caveolins and caveolae. Caveolae are small plasma membrane invaginations that are observed in terminally differentiated cells. Their most important protein marker, caveolin-1, has been shown to play a key role in the regulation of several cellular signaling pathways and in the regulation of plasma lipoprotein metabolism. In the present paper, we have examined the role of caveolin-1 in lipoprotein metabolism using caveolin-1-deficient (Cav-1(-/-)) mice. Our data show that, while Cav-1(-/-) mice show increased plasma triglyceride levels, they also display reduced hepatic very low-density lipoprotein (VLDL) secretion. Additionally, we also found that a caveolin-1 deficiency is associated with an increase in high-density lipoprotein (HDL), and these HDL particles are enriched in cholesteryl ester in Cav-1(-/-) mice when compared with HDL obtained from wild-type mice. Finally, our data suggest that a caveolin-1 deficiency prevents the transcytosis of LDL across endothelial cells, and therefore, that caveolin-1 may be implicated in the regulation of plasma LDL levels. Taken together, our studies suggest that caveolin-1 plays an important role in the regulation of lipoprotein metabolism by controlling their plasma levels as well as their lipid composition. Thus caveolin-1 may also play an important role in the development of atherosclerosis.
- Published
- 2008
- Full Text
- View/download PDF
45. Altered emotionality, spatial memory and cholinergic function in caveolin-1 knock-out mice.
- Author
-
Gioiosa L, Raggi C, Ricceri L, Jasmin JF, Frank PG, Capozza F, Lisanti MP, Alleva E, Sargiacomo M, and Laviola G
- Subjects
- 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, Acetylcholine physiology, Caveolin 1 deficiency, Emotions physiology, Memory Disorders genetics, Memory, Short-Term physiology
- 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
- 2008
- Full Text
- View/download PDF
46. Caveolin-1 is required for the upregulation of fatty acid synthase (FASN), a tumor promoter, during prostate cancer progression.
- Author
-
Di Vizio D, Sotgia F, Williams TM, Hassan GS, Capozza F, Frank PG, Pestell RG, Loda M, Freeman MR, and Lisanti MP
- Subjects
- Animals, Caveolin 1 genetics, Disease Progression, Fatty Acid Synthases genetics, Male, Mice, Mice, Transgenic, Prostatic Neoplasms enzymology, Prostatic Neoplasms genetics, Up-Regulation, Caveolin 1 physiology, Fatty Acid Synthases metabolism, Prostatic Neoplasms pathology
- 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
- Full Text
- View/download PDF
47. Caveolin-1 and liver regeneration: role in proliferation and lipogenesis.
- Author
-
Frank PG and Lisanti MP
- Subjects
- Animals, Humans, Liver metabolism, Liver physiology, Triglycerides metabolism, Triglycerides physiology, Caveolin 1 physiology, Cell Proliferation, Lipogenesis physiology, Liver cytology, Liver Regeneration physiology
- Abstract
Caveolin-1 is an essential component of caveolae, which form 50-100 nm cell surface plasma membrane invaginations. Caveolin-1 has been previously implicated in the regulation of cellular proliferation and lipogenesis. Current data now suggest that caveolin-1 plays an essential role in the regulation of liver regeneration and that it is implicated in the regulation of triglyceride accumulation, which is an essential process that plays a critical role in the regulation of liver regeneration. These findings are consistent with the importance of a lipogenesis program in the regulation of liver regeneration.
- Published
- 2007
- Full Text
- View/download PDF
48. Caveolae and caveolin-1: novel potential targets for the treatment of cardiovascular disease.
- Author
-
Frank PG, Hassan GS, Rodriguez-Feo JA, and Lisanti MP
- Subjects
- Animals, Arteries drug effects, Arteries pathology, Arteries physiopathology, Atherosclerosis drug therapy, Atherosclerosis pathology, Atherosclerosis physiopathology, Cardiovascular Agents therapeutic use, Caveolae drug effects, Caveolin 1 genetics, Endothelium, Vascular drug effects, Endothelium, Vascular metabolism, Humans, Macrophages drug effects, Macrophages metabolism, Mice, Mice, Knockout, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular metabolism, Thrombosis drug therapy, Thrombosis pathology, Thrombosis physiopathology, Arteries metabolism, Atherosclerosis metabolism, Cardiovascular Agents pharmacology, Caveolae metabolism, Caveolin 1 metabolism, Thrombosis metabolism
- Abstract
Caveolae are 50-100 nm cell surface plasma membrane invaginations that are highly enriched in cholesterol and sphingolipids and are characterized by the protein marker caveolin-1. Caveolin-1 is highly expressed in terminally differentiated cells. Among these cells, endothelial cells, smooth muscle cells, and macrophages have all been shown to play key roles in the development of vascular disease. Atherosclerosis and neointimal formation are two major processes that have been associated with arterial occlusion. In both cases, caveolin-1 has been shown to play an important role. However, depending on the cell type and the metabolic pathways regulated by this protein, caveolin-1 may positively or negatively influence the development of vascular disease. Both of these aspects will be discussed in this review.
- Published
- 2007
- Full Text
- View/download PDF
49. Zebrafish as a novel model system to study the function of caveolae and caveolin-1 in organismal biology.
- Author
-
Frank PG and Lisanti MP
- Subjects
- Amino Acid Sequence, Animals, Molecular Sequence Data, Protein Isoforms, Sequence Homology, Amino Acid, Signal Transduction, Tissue Distribution, Zebrafish embryology, Zebrafish metabolism, Caveolae physiology, Caveolin 1 physiology, Models, Animal, Zebrafish physiology
- Published
- 2006
- Full Text
- View/download PDF
50. Caveolin-1-deficient mice show defects in innate immunity and inflammatory immune response during Salmonella enterica serovar Typhimurium infection.
- Author
-
Medina FA, de Almeida CJ, Dew E, Li J, Bonuccelli G, Williams TM, Cohen AW, Pestell RG, Frank PG, Tanowitz HB, and Lisanti MP
- Subjects
- Animals, Caveolin 1 deficiency, Caveolin 1 genetics, Cytokines genetics, Cytokines metabolism, Granuloma immunology, Granuloma microbiology, Granuloma pathology, Lipopolysaccharides immunology, Liver immunology, Liver pathology, Macrophages microbiology, Mice, Mice, Knockout, Neutrophils immunology, Nitric Oxide metabolism, STAT3 Transcription Factor metabolism, Salmonella Infections genetics, Salmonella Infections pathology, Spleen immunology, Spleen pathology, Caveolin 1 physiology, Immunity, Innate genetics, Macrophages immunology, Salmonella Infections immunology, Salmonella typhimurium
- Abstract
A number of studies have shown an association of pathogens with caveolae. To this date, however, there are no studies showing a role for caveolin-1 in modulating immune responses against pathogens. Interestingly, expression of caveolin-1 has been shown to occur in a regulated manner in immune cells in response to lipopolysaccharide (LPS). Here, we sought to determine the role of caveolin-1 (Cav-1) expression in Salmonella pathogenesis. Cav-1(-/-) mice displayed a significant decrease in survival when challenged with Salmonella enterica serovar Typhimurium. Spleen and tissue burdens were significantly higher in Cav-1(-/-) mice. However, infection of Cav-1(-/-) macrophages with serovar Typhimurium did not result in differences in bacterial invasion. In addition, Cav-1(-/-) mice displayed increased production of inflammatory cytokines, chemokines, and nitric oxide. Regardless of this, Cav-1(-/-) mice were unable to control the systemic infection of Salmonella. The increased chemokine production in Cav-1(-/-) mice resulted in greater infiltration of neutrophils into granulomas but did not alter the number of granulomas present. This was accompanied by increased necrosis in the liver. However, Cav-1(-/-) macrophages displayed increased inflammatory responses and increased nitric oxide production in vitro in response to Salmonella LPS. These results show that caveolin-1 plays a key role in regulating anti-inflammatory responses in macrophages. Taken together, these data suggest that the increased production of toxic mediators from macrophages lacking caveolin-1 is likely to be responsible for the marked susceptibility of caveolin-1-deficient mice to S. enterica serovar Typhimurium.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.