Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 199
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 16(4): 386-96, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25665101

ABSTRACT

In the lymphatic sinuses of draining lymph nodes, soluble lymph-borne antigens enter the reticular conduits in a size-selective manner and lymphocytes transmigrate to the parenchyma. The molecular mechanisms that control these processes are unknown. Here we unexpectedly found that PLVAP, a prototypic endothelial protein of blood vessels, was synthesized in the sinus-lining lymphatic endothelial cells covering the distal conduits. In PLVAP-deficient mice, both small antigens and large antigens entered the conduit system, and the transmigration of lymphocytes through the sinus floor was augmented. Mechanistically, the filtering function of the lymphatic sinus endothelium was dependent on diaphragms formed by PLVAP fibrils in transendothelial channels. Thus, in the lymphatic sinus, PLVAP forms a physical sieve that regulates the parenchymal entry of lymphocytes and soluble antigens.


Subject(s)
Carrier Proteins/immunology , Endothelial Cells/immunology , Lymph Nodes/immunology , Lymphocytes/immunology , Membrane Proteins/immunology , Animals , Antigens/immunology , Antigens, CD/genetics , Antigens, CD/metabolism , Carrier Proteins/genetics , Caveolin 1/deficiency , Caveolin 1/genetics , Caveolin 1/immunology , Endothelial Cells/cytology , Endothelium, Lymphatic/cytology , Endothelium, Lymphatic/immunology , Female , Gene Expression Regulation , Lymph Nodes/cytology , Lymphatic Vessels/cytology , Lymphatic Vessels/immunology , Lymphocytes/cytology , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Transendothelial and Transepithelial Migration/immunology
2.
Cardiovasc Diabetol ; 23(1): 138, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664801

ABSTRACT

BACKGROUND: Neutral cholesterol ester hydrolase 1 (NCEH1) plays a critical role in the regulation of cholesterol ester metabolism. Deficiency of NCHE1 accelerated atherosclerotic lesion formation in mice. Nonetheless, the role of NCEH1 in endothelial dysfunction associated with diabetes has not been explored. The present study sought to investigate whether NCEH1 improved endothelial function in diabetes, and the underlying mechanisms were explored. METHODS: The expression and activity of NCEH1 were determined in obese mice with high-fat diet (HFD) feeding, high glucose (HG)-induced mouse aortae or primary endothelial cells (ECs). Endothelium-dependent relaxation (EDR) in aortae response to acetylcholine (Ach) was measured. RESULTS: Results showed that the expression and activity of NCEH1 were lower in HFD-induced mouse aortae, HG-exposed mouse aortae ex vivo, and HG-incubated primary ECs. HG exposure reduced EDR in mouse aortae, which was exaggerated by endothelial-specific deficiency of NCEH1, whereas NCEH1 overexpression restored the impaired EDR. Similar results were observed in HFD mice. Mechanically, NCEH1 ameliorated the disrupted EDR by dissociating endothelial nitric oxide synthase (eNOS) from caveolin-1 (Cav-1), leading to eNOS activation and nitric oxide (NO) release. Moreover, interaction of NCEH1 with the E3 ubiquitin-protein ligase ZNRF1 led to the degradation of Cav-1 through the ubiquitination pathway. Silencing Cav-1 and upregulating ZNRF1 were sufficient to improve EDR of diabetic aortas, while overexpression of Cav-1 and downregulation of ZNRF1 abolished the effects of NCEH1 on endothelial function in diabetes. Thus, NCEH1 preserves endothelial function through increasing NO bioavailability secondary to the disruption of the Cav-1/eNOS complex in the endothelium of diabetic mice, depending on ZNRF1-induced ubiquitination of Cav-1. CONCLUSIONS: NCEH1 may be a promising candidate for the prevention and treatment of vascular complications of diabetes.


Subject(s)
Caveolin 1 , Diet, High-Fat , Endothelial Cells , Endothelium, Vascular , Mice, Inbred C57BL , Nitric Oxide Synthase Type III , Vasodilation , Animals , Male , Mice , Aorta/enzymology , Aorta/physiopathology , Aorta/metabolism , Aorta/drug effects , Aorta/pathology , Caveolin 1/metabolism , Caveolin 1/deficiency , Caveolin 1/genetics , Cells, Cultured , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/physiopathology , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Endothelium, Vascular/physiopathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/enzymology , Endothelium, Vascular/drug effects , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Obesity/enzymology , Obesity/physiopathology , Obesity/metabolism , Signal Transduction , Sterol Esterase/metabolism , Sterol Esterase/genetics , Ubiquitination , Vasodilation/drug effects
3.
Nature ; 555(7698): 673-677, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29562231

ABSTRACT

Obesity-induced metabolic disease involves functional integration among several organs via circulating factors, but little is known about crosstalk between liver and visceral adipose tissue (VAT). In obesity, VAT becomes populated with inflammatory adipose tissue macrophages (ATMs). In obese humans, there is a close correlation between adipose tissue inflammation and insulin resistance, and in obese mice, blocking systemic or ATM inflammation improves insulin sensitivity. However, processes that promote pathological adipose tissue inflammation in obesity are incompletely understood. Here we show that obesity in mice stimulates hepatocytes to synthesize and secrete dipeptidyl peptidase 4 (DPP4), which acts with plasma factor Xa to inflame ATMs. Silencing expression of DPP4 in hepatocytes suppresses inflammation of VAT and insulin resistance; however, a similar effect is not seen with the orally administered DPP4 inhibitor sitagliptin. Inflammation and insulin resistance are also suppressed by silencing expression of caveolin-1 or PAR2 in ATMs; these proteins mediate the actions of DPP4 and factor Xa, respectively. Thus, hepatocyte DPP4 promotes VAT inflammation and insulin resistance in obesity, and targeting this pathway may have metabolic benefits that are distinct from those observed with oral DPP4 inhibitors.


Subject(s)
Dipeptidyl Peptidase 4/metabolism , Hepatocytes/metabolism , Inflammation/enzymology , Insulin Resistance , Intra-Abdominal Fat/pathology , Obesity/enzymology , Administration, Oral , Animals , Caveolin 1/deficiency , Caveolin 1/genetics , Caveolin 1/metabolism , Dipeptidyl Peptidase 4/deficiency , Dipeptidyl Peptidase 4/genetics , Factor Xa/metabolism , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Inflammation/genetics , Inflammation/metabolism , Insulin Resistance/genetics , Intra-Abdominal Fat/metabolism , Macrophages/metabolism , Male , Mice , Mice, Obese , Obesity/genetics , Obesity/metabolism , Receptor, PAR-2/deficiency , Receptor, PAR-2/genetics , Receptor, PAR-2/metabolism , Sitagliptin Phosphate/administration & dosage , Sitagliptin Phosphate/pharmacology
4.
Arterioscler Thromb Vasc Biol ; 40(6): 1510-1522, 2020 06.
Article in English | MEDLINE | ID: mdl-32349535

ABSTRACT

OBJECTIVE: Endothelial Cav-1 (caveolin-1) expression plays a relevant role during atherogenesis by controlling NO production, vascular inflammation, LDL (low-density lipoprotein) transcytosis, and extracellular matrix remodeling. Additional studies have identified cholesterol-rich membrane domains as important regulators of autophagy by recruiting ATGs (autophagy-related proteins) to the plasma membrane. Here, we investigate how the expression of Cav-1 in the aortic endothelium influences autophagy and whether enhanced autophagy contributes to the atheroprotective phenotype observed in Cav-1-deficient mice. Approach and Results: To analyze the impact of Cav-1 deficiency on regulation of autophagy in the aortic endothelium during the progression of atherosclerosis, we fed Ldlr-/- and Cav-1-/-Ldlr-/- mice a Western diet and assessed autophagy in the vasculature. We observe that the absence of Cav-1 promotes autophagy activation in athero-prone areas of the aortic endothelium by enhancing autophagic flux. Mechanistically, we found that Cav-1 interacts with the ATG5-ATG12 complex and influences the cellular localization of autophagosome components in lipid rafts, which controls the autophagosome formation and autophagic flux. Pharmacological inhibition of autophagy attenuates the atheroprotection observed in Cav-1-/- mice by increasing endothelial inflammation and macrophage recruitment, identifying a novel molecular mechanism by which Cav-1 deficiency protects against the progression of atherosclerosis. CONCLUSIONS: These results identify Cav-1 as a relevant regulator of autophagy in the aortic endothelium and demonstrate that pharmacological suppression of autophagic flux in Cav-1-deficient mice attenuates the atheroprotection observed in Cav-1-/- mice. Additionally, these findings suggest that activation of endothelial autophagy by blocking Cav-1 might provide a potential therapeutic strategy for cardiovascular diseases including atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Autophagy/physiology , Caveolin 1/deficiency , Endothelium, Vascular/physiopathology , Vasculitis/prevention & control , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Aorta/pathology , Aorta/physiopathology , Aorta/ultrastructure , Atherosclerosis/etiology , Autophagy/drug effects , Caveolin 1/analysis , Caveolin 1/physiology , Diet, Western , Endothelial Cells/chemistry , Endothelial Cells/physiology , Endothelial Cells/ultrastructure , Endothelium, Vascular/chemistry , Endothelium, Vascular/ultrastructure , Female , Humans , Male , Membrane Microdomains/chemistry , Membrane Microdomains/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Receptors, LDL/deficiency
5.
J Cardiovasc Pharmacol ; 78(2): 175-183, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34554674

ABSTRACT

ABSTRACT: Atrial fibrillation (AF) is a common arrhythmia in the clinic. Ablation failure and recurrence after cardioversion have become medical problems worldwide. An important pathological feature of AF is atrial fibrosis, which increases susceptibility to AF. As an important target of fibrosis signal integration, the signal transducer and activator of transcription 3 (STAT3) signaling pathway plays an important role in fibrosis. Caveolin-1 (CAV1), a cell membrane protein, is involved in a variety of the biological functions of cells. However, the role of CAV1 in atrial fibrosis remains unclear. In this study, Masson's trichrome staining was used to detect the degree of atrial fibrosis, and the expression of CAV1 in the human atrium was evaluated by immunohistochemistry. To further study the role of CAV1, its expression in cultured rat atrial fibroblasts was silenced using siRNAs. Atrial fibroblasts were treated with angiotensin II to observe the effects on CAV1 and the transforming growth factor-ß1 and STAT3 signaling pathways. We also detected the effects of CAV1 scaffolding domain (CSD) peptide on fibrosis through the addition of exogenous CSD peptide. The results showed that CAV1 expression decreased with the aggravation of atrial fibrosis and that this effect increased the incidence of AF. The depletion of CAV1 induced excessive extracellular matrix deposition by activating the STAT3 and transforming growth factor-ß1/SMAD2 signaling pathways, and this effect was exacerbated by stimulation with angiotensin II and improved by CSD peptide. These data suggested that CAV1 not only plays a critical role in fibrosis progression but also provides a target for the treatment of atrial fibrosis and AF.


Subject(s)
Atrial Fibrillation/metabolism , Atrial Remodeling , Caveolin 1/deficiency , Fibroblasts/metabolism , Heart Atria/metabolism , Heart Rate , STAT3 Transcription Factor/metabolism , Adult , Aged , Animals , Atrial Fibrillation/genetics , Atrial Fibrillation/pathology , Atrial Fibrillation/physiopathology , Caveolin 1/genetics , Cells, Cultured , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Female , Fibroblasts/pathology , Fibrosis , Heart Atria/pathology , Heart Atria/physiopathology , Humans , Male , Middle Aged , Myofibroblasts/metabolism , Myofibroblasts/pathology , Rats, Sprague-Dawley , STAT3 Transcription Factor/genetics , Signal Transduction
6.
Am J Physiol Cell Physiol ; 319(5): C933-C944, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32936699

ABSTRACT

Caveolin-1 (Cav-1) is a scaffolding protein and a major component of caveolae/lipid rafts. Previous reports have shown that endothelial dysfunction in Cav-1-deficient (Cav-1-/-) mice is mediated by elevated oxidative stress through endothelial nitric oxide synthase (eNOS) uncoupling and increased NADPH oxidase. Oxidant stress is the net balance of oxidant generation and scavenging, and the role of Cav-1 as a regulator of antioxidant enzymes in vascular tissue is poorly understood. Extracellular SOD (SOD3) is a copper (Cu)-containing enzyme that is secreted from vascular smooth muscle cells/fibroblasts and subsequently binds to the endothelial cells surface, where it scavenges extracellular [Formula: see text] and preserves endothelial function. SOD3 activity is dependent on Cu, supplied by the Cu transporter ATP7A, but whether Cav-1 regulates the ATP7A-SOD3 axis and its role in oxidative stress-mediated vascular dysfunction has not been studied. Here we show that the activity of SOD3, but not SOD1, was significantly decreased in Cav-1-/- vessels, which was rescued by re-expression of Cav-1 or Cu supplementation. Loss of Cav-1 reduced ATP7A protein, but not mRNA, and this was mediated by ubiquitination of ATP7A and proteasomal degradation. ATP7A bound to Cav-1 and was colocalized with SOD3 in caveolae/lipid rafts or perinucleus in vascular tissues or cells. Impaired endothelium-dependent vasorelaxation in Cav-1-/- mice was rescued by gene transfer of SOD3 or by ATP7A-overexpressing transgenic mice. These data reveal an unexpected role of Cav-1 in stabilizing ATP7A protein expression by preventing its ubiquitination and proteasomal degradation, thereby increasing SOD3 activity, which in turn protects against vascular oxidative stress-mediated endothelial dysfunction.


Subject(s)
Caveolin 1/genetics , Copper-Transporting ATPases/genetics , Endothelial Cells/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase/genetics , Animals , Aorta/cytology , Aorta/metabolism , Caveolin 1/deficiency , Copper/pharmacology , Copper Transport Proteins/genetics , Copper Transport Proteins/metabolism , Copper-Transporting ATPases/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation , Male , Mesenteric Arteries/cytology , Mesenteric Arteries/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Oxidative Stress , Primary Cell Culture , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Signal Transduction , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/metabolism , Ubiquitination/drug effects , Vasodilation/drug effects
7.
J Biol Chem ; 294(27): 10544-10552, 2019 07 05.
Article in English | MEDLINE | ID: mdl-31126986

ABSTRACT

Cavin-1/polymerase I and transcript release factor (PTRF) is a requisite component of caveolae, small plasma membrane invaginations that are highly abundant in adipocytes. Cavin-1 is a dynamic molecule whose dissociation from caveolae plays an important role in mechanoprotection and rRNA synthesis. In the former situation, the acute dissociation of cavin-1 from caveolae allows cell membrane expansion that occurs upon insulin-aided lipid uptake into the fat cells. Cavin-1 dissociation from caveolae and membrane flattening alters the cytoskeleton and the interaction of plasma membrane proteins with the extracellular matrix through interactions with focal adhesion structures. Here, using cavin-1 knockout mice, subcellular fractionation, and immunoblotting methods, we addressed the relationship of cavin-1 with focal adhesion complexes following nutritional stimulation. We found that cavin-1 is acutely translocated to focal complex compartments upon insulin stimulation, where it regulates focal complex formation through an interaction with paxillin. We found that loss of cavin-1 impairs focal complex remodeling and focal adhesion formation and causes a mechanical stress response, concomitant with activation of proinflammatory and senescence/apoptosis pathways. We conclude that cavin-1 plays key roles in dynamic remodeling of focal complexes upon metabolic stimulation. This mechanism also underlies the crucial role of caveolae in the long-term healthy expansion of the adipocyte.


Subject(s)
Caveolin 1/metabolism , Diet, High-Fat , Focal Adhesions/drug effects , Inflammation/metabolism , Insulin/pharmacology , 3T3-L1 Cells , Animals , Caveolae/metabolism , Caveolin 1/deficiency , Caveolin 1/genetics , Focal Adhesions/metabolism , Inflammation/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Paxillin/metabolism , Protein Binding , Signal Transduction , Stress, Mechanical
8.
Arterioscler Thromb Vasc Biol ; 39(4): 754-764, 2019 04.
Article in English | MEDLINE | ID: mdl-30786746

ABSTRACT

Objective- Arteriovenous fistulae (AVF) are the most common access created for hemodialysis; however, many AVF fail to mature and require repeated intervention, suggesting a need to improve AVF maturation. Eph-B4 (ephrin type-B receptor 4) is the embryonic venous determinant that is functional in adult veins and can regulate AVF maturation. Cav-1 (caveolin-1) is the major scaffolding protein of caveolae-a distinct microdomain that serves as a mechanosensor at the endothelial cell membrane. We hypothesized that Cav-1 function is critical for Eph-B4-mediated AVF maturation. Approach and Results- In a mouse aortocaval fistula model, both Cav-1 mRNA and protein were increased in the AVF compared with control veins. Cav-1 KO (knockout) mice showed increased fistula wall thickening ( P=0.0005) and outward remodeling ( P<0.0001), with increased eNOS (endothelial NO synthase) activity compared with WT (wild type) mice. Ephrin-B2/Fc inhibited AVF outward remodeling in WT mice but not in Cav-1 KO mice and was maintained in Cav-1 RC (Cav-1 endothelial reconstituted) mice (WT, P=0.0001; Cav-1 KO, P=0.7552; Cav-1 RC, P=0.0002). Cavtratin-a Cav-1 scaffolding domain peptide-decreased AVF wall thickness in WT mice and in Eph-B4 het mice compared with vehicle alone (WT, P=0.0235; Eph-B4 het, P=0.0431); cavtratin also increased AVF patency (day 42) in WT mice ( P=0.0275). Conclusions- Endothelial Cav-1 mediates Eph-B4-mediated AVF maturation. The Eph-B4-Cav-1 axis regulates adaptive remodeling during venous adaptation to the fistula environment. Manipulation of Cav-1 function may be a translational strategy to enhance AVF patency.


Subject(s)
Arteriovenous Shunt, Surgical , Caveolin 1/physiology , Receptor, EphB4/physiology , Signal Transduction/physiology , Vena Cava, Inferior/physiology , Animals , Aorta, Abdominal/surgery , Caveolae/metabolism , Caveolin 1/biosynthesis , Caveolin 1/deficiency , Caveolin 1/genetics , Caveolin 1/pharmacology , Cells, Cultured , Drug Evaluation, Preclinical , Hemorheology , Humans , Lung/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/physiology , Peptide Fragments/pharmacology , Vascular Remodeling/physiology , Vena Cava, Inferior/surgery
9.
Arterioscler Thromb Vasc Biol ; 39(6): 1191-1202, 2019 06.
Article in English | MEDLINE | ID: mdl-30943774

ABSTRACT

Objective- To determine whether pulmonary arterial hypertension is associated with endothelial cell (EC)-Cav-1 (caveolin-1) depletion, EC-derived extracellular vesicle cross talk with macrophages, and proliferation of Cav-1 depleted ECs via TGF-ß (transforming growth factor-ß) signaling. Approach and Results- Pulmonary vascular disease was induced in Sprague-Dawley rats by exposure to a single injection of VEGFRII (vascular endothelial growth factor receptor II) antagonist SU5416 (Su) followed by hypoxia (Hx) plus normoxia (4 weeks each-HxSu model) and in WT (wild type; Tie2.Cre-; Cav1 lox/lox) and EC- Cav1-/- (Tie2.Cre+; Cav1 fl/fl) mice (Hx: 4 weeks). We observed reduced lung Cav-1 expression in the HxSu rat model in association with increased Cav-1+ extracellular vesicle shedding into the circulation. Whereas WT mice exposed to hypoxia exhibited increased right ventricular systolic pressure and pulmonary microvascular thickening compared with the group maintained in normoxia, the remodeling was further increased in EC- Cav1-/- mice indicating EC Cav-1 expression protects against hypoxia-induced pulmonary hypertension. Depletion of EC Cav-1 was associated with reduced BMPRII (bone morphogenetic protein receptor II) expression, increased macrophage-dependent TGF-ß production, and activation of pSMAD2/3 signaling in the lung. In vitro, in the absence of Cav-1, eNOS (endothelial NO synthase) dysfunction was implicated in the mechanism of EC phenotype switching. Finally, reduced expression of EC Cav-1 in lung histological sections from human pulmonary arterial hypertension donors was associated with increased plasma concentration of Cav-1, extracellular vesicles, and TGF-ß, indicating Cav-1 may be a plasma biomarker of vascular injury and key determinant of TGF-ß-induced pulmonary vascular remodeling. Conclusions- EC Cav-1 depletion occurs, in part, via Cav-1+ extracellular vesicle shedding into the circulation, which contributes to increased TGF-ß signaling, EC proliferation, vascular remodeling, and pulmonary arterial hypertension.


Subject(s)
Caveolin 1/deficiency , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Pulmonary Arterial Hypertension/metabolism , Transforming Growth Factor beta/metabolism , Vascular Remodeling , Adolescent , Adult , Aged , Animals , Bone Morphogenetic Protein Receptors, Type II/metabolism , Case-Control Studies , Caveolin 1/genetics , Cell Proliferation , Disease Models, Animal , Endothelial Cells/pathology , Extracellular Vesicles/pathology , Female , Humans , Hypoxia/complications , Indoles , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Pulmonary Arterial Hypertension/etiology , Pulmonary Arterial Hypertension/pathology , Pyrroles , Rats, Sprague-Dawley , Signal Transduction , Smad Proteins/metabolism , Young Adult
10.
Int J Mol Sci ; 21(15)2020 Jul 23.
Article in English | MEDLINE | ID: mdl-32718046

ABSTRACT

Elevated free fatty acids (FFAs) impair beta cell function and reduce beta cell mass as a consequence of the lipotoxicity that occurs in type 2 diabetes (T2D). We previously reported that the membrane protein caveolin-1 (CAV1) sensitizes to palmitate-induced apoptosis in the beta pancreatic cell line MIN6. Thus, our hypothesis was that CAV1 knock-out (CAV1 KO) mice subjected to a high fat diet (HFD) should suffer less damage to beta cells than wild type (WT) mice. Here, we evaluated the in vivo response of beta cells in the pancreatic islets of 8-week-old C57Bl/6J CAV1 KO mice subjected to a control diet (CD, 14% kcal fat) or a HFD (60% kcal fat) for 12 weeks. We observed that CAV1 KO mice were resistant to weight gain when on HFD, although they had high serum cholesterol and FFA levels, impaired glucose tolerance and were insulin resistant. Some of these alterations were also observed in mice on CD. Interestingly, KO mice fed with HFD showed an adaptive response of the pancreatic beta cells and exhibited a significant decrease in beta cell apoptosis in their islets compared to WT mice. These in vivo results suggest that although the CAV1 KO mice are metabolically unhealthy, they adapt better to a HFD than WT mice. To shed light on the possible signaling pathway(s) involved, MIN6 murine beta cells expressing (MIN6 CAV) or not expressing (MIN6 Mock) CAV1 were incubated with the saturated fatty acid palmitate in the presence of mitogen-activated protein kinase inhibitors. Western blot analysis revealed that CAV1 enhanced palmitate-induced JNK, p38 and ERK phosphorylation in MIN6 CAV1 cells. Moreover, all the MAPK inhibitors partially restored MIN6 viability, but the effect was most notable with the ERK inhibitor. In conclusion, our results suggest that CAV1 KO mice adapted better to a HFD despite their altered metabolic state and that this may at least in part be due to reduced beta cell damage. Moreover, they indicate that the ability of CAV1 to increase sensitivity to FFAs may be mediated by MAPK and particularly ERK activation.


Subject(s)
Caveolin 1/deficiency , Diet, High-Fat/adverse effects , Insulin Resistance , Insulin-Secreting Cells/metabolism , MAP Kinase Signaling System , Animals , Caveolin 1/metabolism , Cell Death/drug effects , Cell Death/genetics , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Knockout
11.
Am J Pathol ; 188(10): 2207-2222, 2018 10.
Article in English | MEDLINE | ID: mdl-30253845

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a debilitating, incurable, and life-threatening disease. A cardinal feature of the pathogenesis of IPF is excessive extracellular matrix deposition attributable to proliferation of activated fibrotic lung fibroblasts (fLfs). To assess the underlying mechanism, we analyzed the status of the tumor suppressor protein p53 in fLfs from the lungs of IPF patients or mice with bleomycin-induced established PF. We report that basal expression of p53 is markedly reduced in fLfs. Forced expression of caveolin-1 in fLfs increased basal p53 and reduced profibrogenic proteins, including collagen-1. Transduction of fLfs with adenovirus expressing p53 reduced expression of these proteins. Conversely, inhibition of baseline p53 in control lung fibroblasts from lung tissues increased profibrogenic protein expression. Lung transduction of adenovirus expressing p53 reduced bleomycin-induced PF in wild-type or caveolin-1-deficient mice. Furthermore, treatment of fLfs or fibrotic lung tissues with caveolin-1 scaffolding domain peptide (CSP) or its fragment, CSP7, restored p53 and reduced profibrogenic proteins. Treatment of wild-type mice with i.p. CSP or CSP7 resolved bleomycin-induced PF. These peptides failed to resolve PF in inducible conditional knockout mice lacking p53 in fLfs, indicating the induction of baseline fLf p53 as the basis of the antifibrotic effects.


Subject(s)
Airway Remodeling/physiology , Fibroblasts/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Caveolin 1/deficiency , Caveolin 1/metabolism , Caveolin 1/pharmacology , Humans , Idiopathic Pulmonary Fibrosis/physiopathology , Mice, Inbred C57BL , Peptide Fragments/pharmacology , Transduction, Genetic , Tumor Suppressor Protein p53/antagonists & inhibitors
12.
J Immunol ; 199(3): 874-884, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28637901

ABSTRACT

TCR stimulation by peptide-MHC complexes on APCs requires precise reorganization of molecules into the area of cellular contact to form an immunological synapse from where T cell signaling is initiated. Caveolin (Cav)1, a widely expressed transmembrane protein, is involved in the regulation of membrane composition, cellular polarity and trafficking, and the organization of signal transduction pathways. The presence of Cav1 protein in T cells was identified only recently, and its function in this context is not well understood. We show that Cav1-knockout CD8 T cells have a reduction in membrane cholesterol and sphingomyelin, and upon TCR triggering they exhibit altered morphology and polarity, with reduced effector function compared with Cav1 wild-type CD8 T cells. In particular, redistribution of the ß2 integrin LFA-1 to the immunological synapse is compromised in Cav1-knockout T cells, as is the ability of LFA-1 to form high-avidity interactions with ICAM-1. Our results identify a role for Cav1 in membrane organization and ß2 integrin function in primary CD8 T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Caveolin 1/metabolism , Immunological Synapses/metabolism , Lymphocyte Activation , Lymphocyte Function-Associated Antigen-1/metabolism , Receptors, Antigen, T-Cell/immunology , Animals , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/metabolism , Caveolin 1/deficiency , Cell Membrane/chemistry , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Polarity/immunology , Cholesterol/analysis , Immunological Synapses/chemistry , Immunological Synapses/immunology , Intercellular Adhesion Molecule-1/metabolism , Mice , Receptors, Antigen, T-Cell/chemistry , Signal Transduction , Sphingomyelins/analysis
13.
Arterioscler Thromb Vasc Biol ; 37(6): 1180-1193, 2017 06.
Article in English | MEDLINE | ID: mdl-28473444

ABSTRACT

OBJECTIVE: A disintegrin and metalloproteinase ADAM17 (tumor necrosis factor-α [TNF]-converting enzyme) regulates soluble TNF levels. We tested the hypothesis that aging-induced activation in adipose tissue (AT)-expressed ADAM17 contributes to the development of remote coronary microvascular dysfunction in obesity. APPROACH AND RESULTS: Coronary arterioles (CAs, ≈90 µm) from right atrial appendages and mediastinal AT were examined in patients (aged: 69±11 years, BMI: 30.2±5.6 kg/m2) who underwent open heart surgery. CA and AT were also studied in 6-month and 24-month lean and obese mice fed a normal or high-fat diet. We found that obesity elicited impaired endothelium-dependent CA dilations only in older patients and in aged high-fat diet mice. Transplantation of AT from aged obese, but not from young or aged, mice increased serum cytokine levels, including TNF, and impaired CA dilation in the young recipient mice. In patients and mice, obesity was accompanied by age-related activation of ADAM17, which was attributed to vascular endothelium-expressed ADAM17. Excess, ADAM17-shed TNF from AT arteries in older obese patients was sufficient to impair CA dilation in a bioassay in which the AT artery was serially connected to a CA. Moreover, we found that the increased activity of endothelial ADAM17 is mediated by a diminished inhibitory interaction with caveolin-1, owing to age-related decline in caveolin-1 expression in obese patients and mice or to genetic deletion of caveolin-1. CONCLUSIONS: The present study indicates that aging and obesity cooperatively reduce caveolin-1 expression and increase vascular endothelial ADAM17 activity and soluble TNF release in AT, which may contribute to the development of remote coronary microvascular dysfunction in older obese patients.


Subject(s)
ADAM17 Protein/metabolism , Adipose Tissue/enzymology , Aging/metabolism , Arterioles/enzymology , Coronary Artery Disease/enzymology , Coronary Vessels/enzymology , Vasodilation , ADAM17 Protein/genetics , Adipose Tissue/transplantation , Adult , Age Factors , Aged , Aged, 80 and over , Aging/genetics , Animals , Arterioles/physiopathology , Caveolin 1/deficiency , Caveolin 1/genetics , Caveolin 1/metabolism , Cells, Cultured , Coronary Artery Disease/genetics , Coronary Artery Disease/physiopathology , Coronary Vessels/physiopathology , Diet, High-Fat , Disease Models, Animal , Endothelial Cells/enzymology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Obesity/enzymology , Obesity/genetics , Obesity/physiopathology , RNA Interference , Risk Factors , Signal Transduction , Transfection , Tumor Necrosis Factor-alpha/metabolism
14.
J Cardiovasc Pharmacol ; 71(2): 113-126, 2018 02.
Article in English | MEDLINE | ID: mdl-29419573

ABSTRACT

AIMS: Nitric oxide (NO) and endothelium-dependent hyperpolarization (EDH) play important roles in maintaining cardiovascular homeostasis. We have previously demonstrated that endothelial NO synthase (eNOS) plays diverse roles depending on vessel size, as a NO generating system in conduit arteries and an EDH-mediated system in resistance arteries, for which caveolin-1 (Cav-1) is involved. However, the physiological role of endothelial Cav-1 in microvessels remains to be elucidated. METHODS AND RESULTS: We newly generated endothelium-specific Cav-1-knockout (eCav-1-KO) mice. eCav-1-KO mice showed loss of endothelial Cav-1/eNOS complex and had cardiac hypertrophy despite normal blood pressure. In eCav-1-KO mice, as compared to wild-type controls, the extent of eNOS phosphorylation at inhibitory Thr495 was significantly reduced in mesenteric arteries and the heart. Isometric tension and Langendorff-perfused heart experiments showed that NO-mediated responses were enhanced, whereas EDH-mediated responses were reduced in coronary microcirculation in eCav-1-KO mice. Immunohistochemistry showed increased level of 8-nitroguanosine 3',5'-cyclic monophosphate (8-nitro-cGMP), a marker of nitrative stress, in the heart from eCav-1-KO mice. S-guanylation of cardiac H-Ras in eCav-1-KO mice was also significantly increased compared with wild-type controls. CONCLUSIONS: These results suggest that eCav-1 is involved in the protective role of EDH against nitrative stress caused by excessive NO to maintain cardiac microvascular homeostasis.


Subject(s)
Biological Factors/pharmacology , Cardiomegaly/metabolism , Caveolin 1/metabolism , Coronary Vessels/drug effects , Endothelial Cells/drug effects , Mesenteric Arteries/drug effects , Microvessels/drug effects , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , Nitrosative Stress , Vasodilator Agents/pharmacology , Animals , Biological Factors/metabolism , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Caveolin 1/deficiency , Caveolin 1/genetics , Coronary Vessels/metabolism , Coronary Vessels/physiopathology , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Endothelial Cells/metabolism , Guanosine/analogs & derivatives , Guanosine/metabolism , Isolated Heart Preparation , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Microvessels/metabolism , Microvessels/physiopathology , Nitric Oxide Donors/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitro Compounds/metabolism , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/drug effects
15.
Int J Mol Sci ; 19(6)2018 06 11.
Article in English | MEDLINE | ID: mdl-29891777

ABSTRACT

Liver fibrosis is the common pathological process characterized by activation of hepatic stellate cells (HSCs) and overproduction of extracellular matrix (ECM). Caveolin-1 (Cav1), the principal component of caveolae, is regarded as an important inhibitor of multiple signaling molecules including transforming growth factor ß1(TGF-ß1) signaling. To evaluate the role of Cav1 in liver fibrosis, Cav1 deficient (Cav1−/−) and wild type (WT) mice were subjected to liver fibrosis induced by carbon tetrachloride (CCl4). Results indicated no significant difference between Cav1−/− and WT mice in inflammation or collagen content before CCl4 treatment. After CCl4 administration, Cav1−/− mice showed enhanced TGF-ß1 signaling, as reflected by a significantly greater amount of phosphorylation of Smad2 and collagen deposition in livers over WT animals. Qualitative and quantitative analysis indicated that inflammatory injury to the liver was markedly aggravated, accompanied by increased degeneration and necrosis of hepatocytes, higher alanine aminotransferase (ALT)/aspartate aminotransferase (AST), TGF-α and IL-1ß levels in Cav1−/− animals. The mRNA and protein levels of α-smooth muscle actin (α-SMA), Collagen α1(I), and Collagen α1(III) were further enhanced in Cav1−/− animals. We also observed a significant decrease in collagen content in Cav1−/− and WT animals administrated with Cav1 scaffolding domain peptides (CSD). In vitro study indicated that phosphorylation of Smad2 was inhibited after CSD treatment, accompanied by decreased protein levels of α-SMA, Collagen α1(I), and Collagen α1(III) in HSCs. We conclude that Cav1 is an important inhibitor of TGF-ß1/Smad signaling in HSCs activation and collagen production, which might make it a promising target for therapy of liver fibrosis.


Subject(s)
Caveolin 1/chemistry , Caveolin 1/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Peptides/therapeutic use , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Carbon Tetrachloride , Caveolin 1/deficiency , Cells, Cultured , Collagen Type I/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Inflammation/pathology , Liver Cirrhosis/pathology , Male , Mice , Mice, Knockout , Peptides/pharmacology , Phosphorylation/drug effects , Protein Domains
16.
Arterioscler Thromb Vasc Biol ; 36(1): 97-107, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26543099

ABSTRACT

OBJECTIVE: Endothelium-derived nitric oxide (NO) and endothelium-dependent hyperpolarization (EDH) play important roles in modulating vascular tone in a distinct vessel size-dependent manner; NO plays a dominant role in conduit arteries and EDH in resistance vessels. We have recently demonstrated that endothelial NO synthase (eNOS) is functionally suppressed in resistance vessels through caveolin-1 (Cav-1)-dependent mechanism, switching its function from NO to EDH/hydrogen peroxide generation in mice. Here, we examined the possible importance of the physiological balance between NO and EDH in cardiovascular homeostasis. APPROACH AND RESULTS: We used 2 genotypes of mice in which eNOS activity is genetically upregulated; Cav-1-knockout (Cav-1-KO) and endothelium-specific eNOS transgenic (eNOS-Tg) mice. Isometric tension recordings and Langendorff experiments with isolated perfused hearts showed that NO-mediated relaxations were significantly enhanced, whereas EDH-mediated relaxations were markedly reduced in microcirculations. Importantly, impaired EDH-mediated relaxations of small mesenteric arteries from Cav-1-KO mice were completely rescued by crossing the mice with those with endothelium-specific overexpression of Cav-1. Furthermore, both genotypes showed altered cardiovascular phenotypes, including cardiac hypertrophy in Cav-1-KO mice and hypotension in eNOS-Tg mice. Finally, we examined cardiac responses to chronic pressure overload by transverse aortic constriction in vivo. When compared with wild-type mice, both Cav-1-KO and eNOS-Tg mice exhibited reduced survival after transverse aortic constriction associated with accelerated left ventricular systolic dysfunction, reduced coronary flow reserve, and enhanced myocardial hypoxia. CONCLUSIONS: These results indicate that excessive endothelium-derived NO with reduced EDH impairs cardiovascular homeostasis in mice in vivo.


Subject(s)
Biological Factors/metabolism , Endothelium, Vascular/enzymology , Nitric Oxide/metabolism , Vasodilation , Ventricular Function, Left , Animals , Cardiomegaly/enzymology , Cardiomegaly/genetics , Cardiomegaly/pathology , Caveolin 1/deficiency , Caveolin 1/genetics , Cell Hypoxia , Coronary Circulation , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Enzyme Inhibitors/pharmacology , Genotype , Homeostasis , Hypotension/enzymology , Hypotension/genetics , Hypotension/physiopathology , Isolated Heart Preparation , Male , Membrane Potentials , Mice, Inbred C57BL , Mice, Knockout , Microcirculation , Myocardium/metabolism , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , Phenotype , Signal Transduction , Systole , Time Factors , Up-Regulation , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology
17.
Arterioscler Thromb Vasc Biol ; 36(6): 1220-9, 2016 06.
Article in English | MEDLINE | ID: mdl-27079882

ABSTRACT

OBJECTIVE: Left ventricular (LV) remodeling after acute myocardial infarction still remains an important issue in cardiovascular medicine. We have recently demonstrated that low-intensity pulsed ultrasound (LIPUS) therapy improves myocardial ischemia in a pig model of chronic myocardial ischemia through enhanced myocardial angiogenesis. In the present study, we aimed to demonstrate whether LIPUS also ameliorates LV remodeling after acute myocardial infarction and if so, to elucidate the underlying molecular mechanisms involved in the beneficial effects of LIPUS. APPROACH AND RESULTS: We examined the effects of LIPUS on LV remodeling in a mouse model of acute myocardial infarction, where the heart was treated with either LIPUS or no-LIPUS 3 times in the first week (days 1, 3, and 5). The LIPUS improved mortality and ameliorated post-myocardial infarction LV remodeling in mice. The LIPUS upregulated the expression of vascular endothelial growth factor, endothelial nitric oxide synthase, phosphorylated ERK, and phosphorylated Akt in the infarcted area early after acute myocardial infarction, leading to enhanced angiogenesis. Microarray analysis in cultured human endothelial cells showed that a total of 1050 genes, including those of the vascular endothelial growth factor signaling and focal adhesion pathways, were significantly altered by the LIPUS. Knockdown with small interfering RNA of either ß1-integrin or caveolin-1, both of which are known to play key roles in mechanotransduction, suppressed the LIPUS-induced upregulation of vascular endothelial growth factor. Finally, in caveolin-1-deficient mice, the beneficial effects of LIPUS on mortality and post-myocardial infarction LV remodeling were absent. CONCLUSIONS: These results indicate that the LIPUS therapy ameliorates post-myocardial infarction LV remodeling in mice in vivo, for which mechanotransduction and its downstream pathways may be involved.


Subject(s)
Myocardial Infarction/therapy , Myocardium/metabolism , Neovascularization, Physiologic , Ultrasonic Waves , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left , Ventricular Remodeling , Aged , Animals , Autopsy , Case-Control Studies , Caveolin 1/deficiency , Caveolin 1/genetics , Caveolin 1/metabolism , Cells, Cultured , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Genotype , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Integrin beta1/genetics , Integrin beta1/metabolism , Male , Mechanotransduction, Cellular , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Nitric Oxide Synthase Type III/metabolism , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Time Factors , Transfection , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
18.
Am J Physiol Cell Physiol ; 310(11): C993-C1000, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27122158

ABSTRACT

The expression of caveolin-1 (Cav1) in corneal epithelium is associated with regeneration potency. We used Cav1(-/-) mice to study the role of Cav1 in modulating corneal wound healing. Western blot and whole cell patch clamp were employed to study the effect of Cav1 deletion on Kir4.1 current density in corneas. We found that Ba(2+)-sensitive K(+) currents in primary cultured murine corneal epithelial cells (pMCE) from Cav1(-/-) were dramatically reduced (602 pA) compared with those from wild type (WT; 1,300 pA). As a consequence, membrane potential was elevated in pMCE from Cav1(-/-) compared with that from WT (-43 ± 7.5 vs. -58 ± 4.0 mV, respectively). Western blot showed that either inhibition of Cav1 expression or Ba(2+) incubation stimulated phosphorylation of the EGFR. The transwell migration assay showed that Cav1 genetic inactivation accelerated cell migration. The regrowth efficiency of human corneal epithelial cells (HCE) transfected with siRNA-Cav1 or negative control was evaluated by scrape injury assay. With the presence of mitomycin C (10 µg/ml) to avoid the influence of cell proliferation, Cav1 inhibition with siRNA significantly increased migration compared with control siRNA in HCE. This promoting effect by siRNA-Cav1 could not be further enhanced by cotransfection with siRNA-Kcnj10. By using corneal debridement, we found that wound healing was significantly accelerated in Cav1(-/-) compared with WT mice (70 ± 10 vs. 36 ± 3%, P < 0.01). Our findings imply that the mechanism by which Cav-1 knockout promotes corneal regrowth is, at least partially, due to the inhibition of Kir4.1 which stimulates EGFR signaling.


Subject(s)
Caveolin 1/metabolism , Corneal Injuries/metabolism , Epithelium, Corneal/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Potassium/metabolism , Wound Healing , Animals , Caveolin 1/deficiency , Caveolin 1/genetics , Cell Line , Cell Movement , Corneal Injuries/genetics , Corneal Injuries/pathology , Disease Models, Animal , Epithelium, Corneal/injuries , Epithelium, Corneal/pathology , ErbB Receptors/metabolism , Genotype , Humans , Membrane Potentials , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphorylation , Potassium Channels, Inwardly Rectifying/genetics , Primary Cell Culture , RNA Interference , Signal Transduction , Transfection
19.
J Cell Physiol ; 231(6): 1334-42, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26529275

ABSTRACT

Increased vascular smooth muscle cell (VSMC) proliferation is a factor in atherosclerosis and injury-induced arterial (re) stenosis. Inhibition of polyamine synthesis by α-difluoro-methylornithine (DFMO), an irreversible inhibitor of ornithine decarboxylase, attenuates VSMC proliferation with high sensitivity and specificity. However, cells can escape polyamine synthesis blockade by importing polyamines from the environment. To address this issue, polyamine transport inhibitors (PTIs) have been developed. We investigated the effects of the novel trimer44NMe (PTI-1) alone and in combination with DFMO on VSMC polyamine uptake, proliferation and phenotype regulation. PTI-1 efficiently inhibited polyamine uptake in primary mouse aortic and human coronary VSMCs in the absence as well as in the presence of DFMO. Interestingly, culture with DFMO for 2 days substantially (>95%) reduced putrescine (Put) and spermidine (Spd) contents without any effect on proliferation. Culture with PTI-1 alone had no effect on either polyamine levels or proliferation rate, but the combination of both treatments reduced Put and Spd levels below the detection limit and inhibited proliferation. Treatment with DFMO for a longer time period (4 days) reduced Put and Spd below their detection limits and reduced proliferation, showing that only a small pool of polyamines is needed to sustain VSMC proliferation. Inhibited proliferation by polyamine depletion was associated with maintained expression of contractile smooth marker genes. In cultured intact mouse aorta, PTI-1 potentiated the DFMO-induced inhibition of cell proliferation. The combination of endogenous polyamine synthesis inhibition with uptake blockade is thus a viable approach for targeting unwanted vascular cell proliferation in vivo, including vascular restenosis.


Subject(s)
Biogenic Polyamines/biosynthesis , Cell Proliferation/drug effects , Eflornithine/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Ornithine Decarboxylase Inhibitors/pharmacology , Polyamines/pharmacology , Vasoconstriction/drug effects , Animals , Biological Transport , Caveolin 1/deficiency , Caveolin 1/genetics , Cells, Cultured , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Regulation , Humans , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phenotype , Putrescine/metabolism , Spermidine/metabolism , Time Factors , Tissue Culture Techniques
20.
Adv Exp Med Biol ; 854: 411-8, 2016.
Article in English | MEDLINE | ID: mdl-26427439

ABSTRACT

Caveolin-1 (Cav-1), the scaffolding protein of caveolae, is expressed in several retinal cell types and is associated with ocular pathologies. Cav-1 modulates neuroinflammatory/neuroprotective responses to central nervous system injury. We have shown that loss of Cav-1 results in a blunted cytokine response in retinas challenged with inflammatory stimuli. As neuroinflammatory and neuroprotective signaling overlap in their cytokine production and downstream signaling pathways, we hypothesized that loss of Cav-1 may also suppress neuroprotective signaling in the retina. To test this, we subjected mice in which Cav-1 was deleted specifically in the retina to a neurodegenerative insult induced by sodium iodate (NaIO3) and measured STAT3 activation, a measure of neuroprotective signaling. Our results show that Cav-1 ablation blunts STAT3 activation induced by NaIO3. STAT3 activation in response to intravitreal administration of the IL-6 family cytokine, leukemia inhibitory factor (LIF), was not affected by Cav-1 deletion indicating a competent gp130 receptor response. Thus, Cav-1 modulates neuroprotective signaling by regulating the endogenous production of neuroprotective factors.


Subject(s)
Caveolin 1/genetics , Neuroprotection/genetics , Retina/metabolism , Signal Transduction/genetics , Animals , Blotting, Western , Caveolin 1/deficiency , Female , Immunohistochemistry , Injections, Intraperitoneal , Iodates/administration & dosage , Iodates/pharmacology , Leukemia Inhibitory Factor/administration & dosage , Leukemia Inhibitory Factor/pharmacology , Male , Mice , Mice, Knockout , Neuroprotection/drug effects , Retina/drug effects , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL