Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
2.
J Biol Chem ; 276(52): 48619-22, 2001 Dec 28.
Article En | MEDLINE | ID: mdl-11689550

The role of endothelial cell caveolae in the uptake and transport of macromolecules from the blood-space to the tissue-space remains controversial. To address this issue directly, we employed caveolin-1 gene knock-out mice that lack caveolin-1 protein expression and caveolae organelles. Here, we show that endothelial cell caveolae are required for the efficient uptake and transport of a known caveolar ligand, i.e. albumin, in vivo. Caveolin-1-null mice were perfused with 5-nm gold-conjugated albumin, and its uptake was followed by transmission electron microscopy. Our results indicate that gold-conjugated albumin is not endocytosed by Cav-1-deficient lung endothelial cells and remains in the blood vessel lumen; in contrast, gold-conjugated albumin was concentrated and internalized by lung endothelial cell caveolae in wild-type mice, as expected. To quantitate this defect in uptake, we next studied the endocytosis of radioiodinated albumin using aortic ring segments from wild-type and Cav-1-null mice. Interestingly, little or no uptake of radioiodinated albumin was observed in the aortic segments from Cav-1-deficient mice, whereas aortic segments from wild-type mice showed robust uptake that was time- and temperature-dependent and competed by unlabeled albumin. We conclude that endothelial cell caveolae are required for the efficient uptake and transport of albumin from the blood to the interstitium.


Albumins/metabolism , Biological Transport/physiology , Caveolae/metabolism , Caveolins/genetics , Endothelium, Vascular/metabolism , Endothelium/metabolism , Animals , Aorta/anatomy & histology , Aorta/metabolism , Caveolin 1 , Caveolins/metabolism , Endothelium/ultrastructure , Endothelium, Vascular/ultrastructure , Gold Colloid/metabolism , Histocytochemistry , In Vitro Techniques , Iodine Radioisotopes/metabolism , Lung , Mice , Mice, Knockout
4.
Trends Mol Med ; 7(10): 435-41, 2001 Oct.
Article En | MEDLINE | ID: mdl-11597517

Caveolae are vesicular invaginations of the plasma membrane, and function as 'message centers' for regulating signal transduction events. Caveolin-3, a muscle-specific caveolin-related protein, is the principal structural protein of caveolar membrane domains in skeletal muscle and in the heart. Several mutations within the coding sequence of the human caveolin-3 gene (located at 3p25) have been identified. Mutations that lead to a loss of approximately 95% of caveolin-3 protein expression are responsible for a novel autosomal dominant form of limb-girdle muscular dystrophy (LGMD-1C) in humans. By contrast, upregulation of the caveolin-3 protein is associated with Duchenne muscular dystrophy (DMD). Thus, tight regulation of caveolin-3 appears essential for maintaining normal muscle health and homeostasis.


Caveolae/metabolism , Caveolins/genetics , Caveolins/metabolism , Muscular Dystrophies/genetics , Animals , Caveolae/chemistry , Caveolin 3 , Disease Models, Animal , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Mutation/genetics , Neoplasms/genetics , Neoplasms/metabolism
5.
Am J Physiol Cell Physiol ; 281(4): C1241-50, 2001 Oct.
Article En | MEDLINE | ID: mdl-11546661

Numerous components of the cAMP-based signaling cascade, namely G-proteins and G- protein coupled receptors, adenylyl cyclase, and protein kinase A (PKA) have been localized to caveolae and shown to be regulated by the caveolar marker proteins, the caveolins. In order to gain mechanistic insights into these processes in vivo, we have assessed the functional interaction of caveolin-1 (Cav-1) with PKA using mutational analysis. As two regions of Cav-1 had previously been implicated in PKA signaling in vitro, we constructed Cav-1 molecules with mutations/deletions in one or both of these domains. Examination of these mutants shows that Cav-1 requires the presence of either the scaffolding domain or the COOH-terminal domain (but not both) to functionally interact with and inhibit PKA. Interestingly, in contrast to the wild-type protein, these Cav-1 mutants are not localized to caveolae microdomains. However, upon coexpression with wild-type Cav-1, a substantial amount of the mutants was recruited to the caveolae membrane fraction. Using the Cav-1 double mutant with both disrupted scaffolding and COOH-terminal domains, we show that wild-type Cav-1's inhibition of PKA signaling can be partially abrogated in a dose-responsive manner; i.e., the mutant acts in a dominant-negative fashion. Thus, this dominant-negative caveolin-1 mutant will be extremely valuable for assessing the functional role of endogenous caveolin-1 in regulating a variety of other signaling cascades.


Caveolins/chemistry , Caveolins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Caveolin 1 , Caveolins/genetics , Cell Line , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , DNA Mutational Analysis , Green Fluorescent Proteins , Indicators and Reagents/metabolism , Luciferases/genetics , Luminescent Proteins/genetics , Mutagenesis/physiology , Protein Structure, Tertiary , Signal Transduction/physiology , Structure-Activity Relationship
6.
Biochem Soc Trans ; 29(Pt 4): 494-9, 2001 Aug.
Article En | MEDLINE | ID: mdl-11498016

Caveolae ('little caves') are plasma membrane specializations of 50-100 nm in diameter, and the caveolins are structural proteins used by cells to form caveolae. We and other investigators have discovered that caveolae organelles may be important both in normal signal transduction and in the pathogenesis of a number of human diseases, such as cancer. Here we describe the functional roles of the caveolin gene family and summarize the evidence that supports a role for caveolae as mediators of a number of cellular signalling processes, including apoptosis.


Caveolins/genetics , Genes, Tumor Suppressor , Amino Acid Sequence , Animals , Apoptosis , Caveolin 1 , Caveolin 2 , Caveolin 3 , Caveolins/chemistry , Chromosome Mapping , Chromosomes, Human, Pair 7 , Humans , Mice , Molecular Sequence Data , Multigene Family , Neoplasms/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction
8.
J Biol Chem ; 276(41): 38121-38, 2001 Oct 12.
Article En | MEDLINE | ID: mdl-11457855

Caveolin-1 is the principal structural protein of caveolae membranes in fibroblasts and endothelia. Recently, we have shown that the human CAV-1 gene is localized to a suspected tumor suppressor locus, and mutations in Cav-1 have been implicated in human cancer. Here, we created a caveolin-1 null (CAV-1 -/-) mouse model, using standard homologous recombination techniques, to assess the role of caveolin-1 in caveolae biogenesis, endocytosis, cell proliferation, and endothelial nitric-oxide synthase (eNOS) signaling. Surprisingly, Cav-1 null mice are viable. We show that these mice lack caveolin-1 protein expression and plasmalemmal caveolae. In addition, analysis of cultured fibroblasts from Cav-1 null embryos reveals the following: (i) a loss of caveolin-2 protein expression; (ii) defects in the endocytosis of a known caveolar ligand, i.e. fluorescein isothiocyanate-albumin; and (iii) a hyperproliferative phenotype. Importantly, these phenotypic changes are reversed by recombinant expression of the caveolin-1 cDNA. Furthermore, examination of the lung parenchyma (an endothelial-rich tissue) shows hypercellularity with thickened alveolar septa and an increase in the number of vascular endothelial growth factor receptor (Flk-1)-positive endothelial cells. As predicted, endothelial cells from Cav-1 null mice lack caveolae membranes. Finally, we examined eNOS signaling by measuring the physiological response of aortic rings to various stimuli. Our results indicate that eNOS activity is up-regulated in Cav-1 null animals, and this activity can be blunted by using a specific NOS inhibitor, nitro-l-arginine methyl ester. These findings are in accordance with previous in vitro studies showing that caveolin-1 is an endogenous inhibitor of eNOS. Thus, caveolin-1 expression is required to stabilize the caveolin-2 protein product, to mediate the caveolar endocytosis of specific ligands, to negatively regulate the proliferation of certain cell types, and to provide tonic inhibition of eNOS activity in endothelial cells.


Caveolins/physiology , Cell Division/genetics , Endothelium, Vascular/metabolism , Albumins/metabolism , Animals , Base Sequence , Caveolin 1 , Caveolins/genetics , Caveolins/metabolism , DNA Primers , Endocytosis , Endothelium, Vascular/enzymology , Gene Targeting , Humans , Hydrolysis , In Vitro Techniques , Lung/cytology , Lung/metabolism , Lung/ultrastructure , Mice , Mice, Knockout , Microscopy, Electron , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Phenotype , Signal Transduction , Transferrin/metabolism
9.
Am J Physiol Cell Physiol ; 280(5): C1204-14, 2001 May.
Article En | MEDLINE | ID: mdl-11287334

Caveolin-1 is a principal structural component of caveolae membranes. These membrane microdomains participate in the regulation of signaling, transcytosis, and cholesterol homeostasis at the plasma membrane. In the present study, we determined the effect of caveolin-1 expression on cellular cholesterol efflux mediated by high-density lipoprotein (HDL). We evaluated this effect in parental NIH/3T3 cells as well as in two transformed NIH/3T3 cell lines in which caveolin-1 protein levels are dramatically downregulated. Compared with parental NIH/3T3 cells, these two transformed cell lines effluxed cholesterol more rapidly to HDL. In addition, NIH/3T3 cells harboring caveolin-1 antisense also effluxed cholesterol more rapidly to HDL. However, this effect was not due to changes in total cellular cholesterol content. We further showed that chronic HDL exposure reduced caveolin-1 protein expression in NIH/3T3 cells. HDL exposure also inhibited caveolin-1 promoter activity, suggesting a direct negative effect of HDL on caveolin-1 gene transcription. Moreover, we showed that HDL-induced downregulation of caveolin-1 prevents the uptake of oxidized low-density lipoprotein in human endothelial cells. These data suggest a novel proatherogenic role for caveolin-1, i.e., regarding the uptake and/or transcytosis of modified lipoproteins.


Caveolins/physiology , Cholesterol/metabolism , Lipoproteins, HDL/pharmacology , Transcription Factors , 3T3 Cells , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Caveolin 1 , Caveolins/genetics , Cell Line, Transformed , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Genes, abl , Genes, ras , Kinetics , Luciferases/genetics , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Promoter Regions, Genetic , Sterol Regulatory Element Binding Protein 1 , Transfection
10.
Biochemistry ; 40(11): 3354-62, 2001 Mar 20.
Article En | MEDLINE | ID: mdl-11258956

The c-Myc oncoprotein contributes to oncogenesis by activating and repressing a repertoire of genes involved in cellular proliferation, metabolism, and apoptosis. Increasing evidence suggests that the repressor function of c-Myc is critical for transformation. Therefore, identifying and characterizing Myc-repressed genes is imperative to understanding the mechanisms of Myc-induced tumorigenesis. Here, we employ NIH 3T3 cell lines harboring c-Myc-ER or N-Myc-ER to dissect the relationship between Myc activation and caveolin-1 expression. In this well-established inducible system, treatment with estrogen like molecules, such as tamoxifen, leads to activation of Myc, but in a tightly controlled fashion. Using this approach, we show that Myc activation induces the repression of caveolin-1 expression at the transcriptional level. We also provide two independent lines of evidence suggesting that caveolin-1 is a direct target of Myc: (i) the effect of Myc activation on caveolin-1 expression is independent of new protein synthesis, as revealed through the use of cycloheximide; and (ii) Myc-mediated repression of the caveolin-1 promoter is dependent on an intact INR sequence. Moreover, we show that expression of caveolin-1, via an adenoviral vector approach, can suppress cell transformation that is mediated by Myc activation. In support of these observations, treatment with an adenoviral vector harboring anti-sense caveolin-1 specifically potentiates transformation induced by Myc activation. Taken together, our results indicate that caveolin-1 is a direct target of Myc repression, and they also provide evidence for an additional mechanism by which Myc repression can elicit a malignant phenotype.


Caveolins/antagonists & inhibitors , Caveolins/genetics , Gene Expression Regulation/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/physiology , Repressor Proteins/physiology , Transcription, Genetic , 3T3 Cells , 5' Untranslated Regions/antagonists & inhibitors , 5' Untranslated Regions/genetics , Animals , Caveolin 1 , Caveolins/biosynthesis , Caveolins/physiology , Cell Transformation, Neoplastic/genetics , Down-Regulation/genetics , Mice , Phenotype , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Isoforms/physiology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Receptors, Estrogen/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transfection
11.
J Biol Chem ; 276(9): 6727-38, 2001 Mar 02.
Article En | MEDLINE | ID: mdl-11102446

Transforming growth factor-beta (TGF-beta) signaling proceeds from the cell membrane to the nucleus through the cooperation of the type I and II serine/threonine kinase receptors and their downstream SMAD effectors. Although various regulatory proteins affecting TGF-beta-mediated events have been described, relatively little is known about receptor interactions at the level of the plasma membrane. Caveolae are cholesterol-rich membrane microdomains that, along with their marker protein caveolin-1 (Cav-1), have been implicated in the compartmentalization and regulation of certain signaling events. Here, we demonstrate that specific components of the TGF-beta cascade are associated with caveolin-1 in caveolae and that Cav-1 interacts with the Type I TGF-beta receptor. Additionally, Cav-1 is able to suppress TGF-beta-mediated phosphorylation of Smad-2 and subsequent downstream events. We localize the Type I TGF-beta receptor interaction to the scaffolding domain of Cav-1 and show that it occurs in a physiologically relevant time frame, acting to rapidly dampen signaling initiated by the TGF-beta receptor complex.


Caveolins/physiology , DNA-Binding Proteins/physiology , Receptors, Transforming Growth Factor beta/physiology , Trans-Activators/physiology , Transforming Growth Factor beta/physiology , 3T3 Cells , Amino Acid Sequence , Animals , Binding Sites , Caveolin 1 , Cell Differentiation , DNA-Binding Proteins/chemistry , Mice , Molecular Sequence Data , Phosphorylation , Receptors, Transforming Growth Factor beta/analysis , Smad2 Protein , Tacrolimus Binding Protein 1A/physiology , Trans-Activators/chemistry
12.
Biochemistry ; 39(45): 13916-24, 2000 Nov 14.
Article En | MEDLINE | ID: mdl-11076533

Squamous cell carcinomas of the lung and cervix arise by neoplastic transformation of their respective tissue epithelia. In the case of cervical carcinomas, an increasing body of evidence implicates the human papillomavirus, HPV (types 16 and 18), as playing a pivotal role in this malignant transformation process. The HPV early genes E6 and E7 are known to inactivate the tumor suppressors p53 and Rb, respectively; this leads to disruption of cell cycle regulation, predisposing cells to a cancerous phenotype. However, the role of caveolin-1 (a putative tumor suppressor) in this process remains unknown. Here, we show that caveolin-1 protein expression is consistently reduced in a panel of lung and cervical cancer derived cell lines and that this reduction is not due to hyperactivation of p42/44 MAP kinase (a known negative regulator of caveolin-1 transcription). Instead, we provide evidence that this down-regulation event is due to expression of the HPV E6 viral oncoprotein, as stable expression of E6 in NIH 3T3 cells is sufficient to dramatically reduce caveolin-1 protein levels. Furthermore, we demonstrate that p53-a tumor suppressor inactivated by E6-is a positive regulator of caveolin-1 gene transcription and protein expression. SiHa cells are derived from a human cervical squamous carcinoma, harbor a fully integrated copy of the HPV 16 genome (including E6), and show dramatically reduced levels of caveolin-1 expression. We show here that adenoviral-mediated gene transfer of the caveolin-1 cDNA to SiHa cells restores caveolin-1 protein expression and abrogates their anchorage-independent growth in soft agar. Taken together, our results suggest that the HPV oncoprotein E6 down-regulates caveolin-1 via inactivation of p53 and that replacement of caveolin-1 expression can partially revert HPV-mediated cell transformation.


Antiviral Agents/physiology , Caveolins/antagonists & inhibitors , Caveolins/biosynthesis , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Viral , Down-Regulation , Genes, p53/physiology , Papillomaviridae/physiology , Repressor Proteins , 3T3 Cells , Animals , Antiviral Agents/antagonists & inhibitors , Antiviral Agents/biosynthesis , Antiviral Agents/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Caveolin 1 , Caveolins/genetics , Caveolins/physiology , Cell Line, Transformed , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cell Transformation, Viral/genetics , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Growth Inhibitors/genetics , Growth Inhibitors/physiology , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Oncogene Proteins, Viral/biosynthesis , Oncogene Proteins, Viral/genetics , Phenotype , Promoter Regions, Genetic , Recombinant Proteins/biosynthesis , Recombinant Proteins/pharmacology , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Up-Regulation/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology
13.
J Cell Sci ; 113 ( Pt 12): 2103-9, 2000 Jun.
Article En | MEDLINE | ID: mdl-10825283

In adult animals and humans, signal transduction maintains homeostasis. When homeostatic mechanisms are interrupted, an illness or disease may ensue. Caveolae are plasma membrane specializations that contain the structural proteins caveolins, and appear to be important for normal signal transduction. The caveolin scaffolding domain interacts with several signaling molecules, sequestering them in the absence of activating signals, and thereby reducing the signal-to-noise ratio. Deletion and mutation of genes that encode caveolins is implicated in the pathogenesis of several human diseases. Down-regulation of caveolin-1 protein expression leads to deregulated signaling and consequently tumorigenesis, whereas naturally occurring dominant-negative caveolin-3 mutations cause muscular dystrophy.


Caveolins , Cell Transformation, Neoplastic , Membrane Proteins/physiology , Muscular Dystrophies , Signal Transduction , Adult , Amino Acid Sequence , Animals , Caveolin 1 , Humans , Molecular Sequence Data
14.
J Biol Chem ; 275(27): 20717-25, 2000 Jul 07.
Article En | MEDLINE | ID: mdl-10748172

Caveolin-1 is a principal component of caveolae membranes that may function as a transformation suppressor. For example, the human caveolin-1 gene is localized to a suspected tumor suppressor locus (D7S522; 7q31.1) that is deleted in human cancers, including mammary carcinomas. However, little is known about the role of caveolins in regulating cell movement, a critical parameter in determining metastatic potential. Here, we examine the role of caveolin-1 in cell movement. For this purpose, we employed an established cellular model, MTLn3, a metastatic rat mammary adenocarcinoma cell line. In this system, epidermal growth factor (EGF) stimulation induces rapid lamellipod extension and cell migration. Interestingly, we find that MTLn3 cells fail to express detectable levels of endogenous caveolin-1. To restore caveolin-1 expression in MTLn3 cells efficiently, we employed an inducible adenoviral gene delivery system to achieve tightly controlled expression of caveolin-1. We show here that caveolin-1 expression in MTLn3 cells inhibits EGF-stimulated lamellipod extension and cell migration and blocks their anchorage-independent growth. Under these conditions, EGF-induced activation of the p42/44 mitogen-activated protein kinase cascade is also blunted. Our results suggest that caveolin-1 expression in motile MTLn3 cells induces a non-motile phenotype.


Caveolins , Epidermal Growth Factor/antagonists & inhibitors , Membrane Proteins/metabolism , Adenocarcinoma , Adenoviridae/genetics , Animals , Caveolin 1 , Caveolin 2 , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Movement/drug effects , Cell Size/drug effects , Epidermal Growth Factor/pharmacology , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal , Membrane Proteins/genetics , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Proteins/metabolism , Rats , Time Factors , Transfection , Tumor Cells, Cultured
15.
J Biol Chem ; 274(45): 32333-41, 1999 Nov 05.
Article En | MEDLINE | ID: mdl-10542274

Caveolin-1 is a principal component of caveolae membranes in vivo. Caveolin-1 mRNA and protein expression are down-regulated in NIH 3T3 cells in response to transformation by activated oncogenes, such as H-Ras(G12V) and v-Abl. The mechanisms governing this down-regulation event remain unknown. Here, we show that caveolin-1 gene expression is directly regulated by activation of the Ras-p42/44 MAP kinase cascade. Down regulation of caveolin-1 protein expression by Ras is independent of (i) the type of activating mutation (G12V versus Q61L) and (ii) the form of activated Ras transfected (H-Ras versus K-Ras versus N-Ras). Treatment of Ras or Raf-transformed NIH 3T3 cells with a well characterized MEK inhibitor (PD 98059) restores caveolin-1 protein expression. In contrast, treatment of v-Src and v-Abl transformed NIH 3T3 cells with PD 98059 does not restore caveolin-1 expression. Thus, there must be at least two pathways for down-regulating caveolin-1 expression: one that is p42/44 MAP kinase-dependent and another that is p42/44 MAP kinase-independent. We focused our efforts on the p42/44 MAP kinase-dependent pathway. The activity of a panel of caveolin-1 promoter constructs was evaluated using transient expression in H-Ras(G12V) transformed NIH 3T3 cells. We show that caveolin-1 promoter activity is up-regulated approximately 5-fold by inhibition of the p42/44 MAP kinase cascade. Using electrophoretic mobility shift assays we provide evidence that the caveolin-1 promoter (from -156 to -561) is differentially bound by transcription factors in normal and H-Ras(G12V)-transformed cells. We also show that activation of protein kinase A (PKA) signaling is sufficient to down-regulate caveolin-1 protein expression and promoter activity. Thus, we have identified two signaling pathways (Ras-p42/44 MAP kinase and PKA) that transcriptionally down-regulate caveolin-1 gene expression.


Caveolins , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation , Gene Expression Regulation , Membrane Proteins/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Promoter Regions, Genetic , Signal Transduction , 3T3 Cells , Animals , Caveolin 1 , Cell Transformation, Viral , Enzyme Activation , Mice , Mitogen-Activated Protein Kinase 3 , Molecular Sequence Data , Transcription Factors/metabolism
16.
J Biol Chem ; 274(37): 26353-60, 1999 Sep 10.
Article En | MEDLINE | ID: mdl-10473592

cAMP-dependent processes are essential for cell growth, differentiation, and homeostasis. The classic components of this system include the serpentine receptors, heterotrimeric G-proteins, adenylyl cyclase, protein kinase A (PKA), and numerous downstream target substrates. Evidence is accumulating that some members of this cascade are concentrated within membrane microdomains, termed caveolae and caveolae-related domains. In addition, the caveolin-1 protein has been shown to interact with some of these components, and this interaction inhibits their enzymatic activity. However, the functional effects of caveolins on cAMP-mediated signaling at the most pivotal step, PKA activation, remain unknown. Here, we show that caveolin-1 can dramatically inhibit cAMP-dependent signaling in vivo. We provide evidence for a direct interaction between caveolin-1 and the catalytic subunit of PKA both in vitro and in vivo. Caveolin-1 binding appears to be mediated both by the caveolin scaffolding domain (residues 82-101) and a portion of the C-terminal domain (residues 135-156). Further functional analysis indicates that caveolin-based peptides derived from these binding regions can inhibit the catalytic activity of purified PKA in vitro. Mutational analysis of the caveolin scaffolding domain reveals that a series of aromatic residues within the caveolin scaffolding domain are critical for mediating inhibition of PKA. In addition, co-expression of caveolin-1 and PKA in cultured cells results in their co-localization as seen by immunofluorescence microscopy. In cells co-expressing caveolin-1 and PKA, PKA assumed a punctate distribution that coincided with the distribution of caveolin-1. In contrast, in cells expressing PKA alone, PKA was localized throughout the cytoplasm and yielded a diffuse staining pattern. Taken together, our results suggest that the direct inhibition of PKA by caveolin-1 is an important and previously unrecognized mechanism for modulating cAMP-mediated signaling.


Caveolins , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/physiology , Membrane Proteins/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Catalytic Domain , Caveolin 1 , Cricetinae , Glutathione Transferase/metabolism , Membrane Proteins/chemistry , Molecular Sequence Data , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Tumor Cells, Cultured
17.
J Biol Chem ; 274(22): 15781-5, 1999 May 28.
Article En | MEDLINE | ID: mdl-10336480

Angiogenesis is the process by which new blood vessels are formed via proliferation of vascular endothelial cells. A variety of angiogenesis inhibitors that antagonize the effects of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) have recently been identified. However, the mechanism by which these diverse angiogenesis inhibitors exert their common effects remains largely unknown. Caveolin-1 and -2 are known to be highly expressed in vascular endothelial cells both in vitro and in vivo. Here, we examine the potential role of caveolins in the angiogenic response. For this purpose, we used the well established human umbilical vein endothelial cell line, ECV 304. Treatment of ECV 304 cells with known angiogenic growth factors (VEGF, bFGF, or hepatocyte growth factor/scatter factor), resulted in a dramatic reduction in the expression of caveolin-1. This down-regulation event was selective for caveolin-1, as caveolin-2 levels remained constant under these conditions of growth factor stimulation. VEGF-induced down-regulation of caveolin-1 expression also resulted in the morphological loss of cell surface caveolae organelles as seen by transmission electron microscopy. A variety of well characterized angiogenesis inhibitors (including angiostatin, fumagillin, 2-methoxy estradiol, transforming growth factor-beta, and thalidomide) effectively blocked VEGF-induced down-regulation of caveolin-1 as seen by immunoblotting and immunofluorescence microscopy. However, treatment with angiogenesis inhibitors alone did not significantly affect the expression of caveolin-1. PD98059, a specific inhibitor of mitogen-activated protein kinase and a known angiogenesis inhibitor, also blocked the observed VEGF-induced down-regulation of caveolin-1. Furthermore, we show that caveolin-1 can function as a negative regulator of VEGF-R (KDR) signal transduction in vivo. Thus, down-regulation of caveolin-1 may be an important step along the pathway toward endothelial cell proliferation.


Caveolins , DNA-Binding Proteins , Endothelial Growth Factors/pharmacology , Endothelium, Vascular/drug effects , Lymphokines/pharmacology , Membrane Proteins/metabolism , Neovascularization, Physiologic/drug effects , Transcription Factors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Caveolin 1 , Cell Division/drug effects , Cell Line , Down-Regulation/drug effects , Fibroblast Growth Factor 2/pharmacology , Hepatocyte Growth Factor/pharmacology , Humans , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , ets-Domain Protein Elk-1
...