Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Nat Commun ; 14(1): 4989, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37591837

ABSTRACT

The estrogen receptor (ER) designated ERα has actions in many cell and tissue types that impact glucose homeostasis. It is unknown if these include mechanisms in endothelial cells, which have the potential to influence relative obesity, and processes in adipose tissue and skeletal muscle that impact glucose control. Here we show that independent of impact on events in adipose tissue, endothelial ERα promotes glucose tolerance by enhancing endothelial insulin transport to skeletal muscle. Endothelial ERα-deficient male mice are glucose intolerant and insulin resistant, and in females the antidiabetogenic actions of estradiol (E2) are absent. The glucose dysregulation is due to impaired skeletal muscle glucose disposal that results from attenuated muscle insulin delivery. Endothelial ERα activation stimulates insulin transcytosis by skeletal muscle microvascular endothelial cells. Mechanistically this involves nuclear ERα-dependent upregulation of vesicular trafficking regulator sorting nexin 5 (SNX5) expression, and PI3 kinase activation that drives plasma membrane recruitment of SNX5. Thus, coupled nuclear and non-nuclear actions of ERα promote endothelial insulin transport to skeletal muscle to foster normal glucose homeostasis.


Subject(s)
Estrogen Receptor alpha , Insulin , Animals , Female , Male , Mice , Endothelial Cells , Glucose , Muscle, Skeletal , Receptors, Estrogen
2.
Nat Commun ; 14(1): 4101, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37491347

ABSTRACT

Hypercholesterolemia and vascular inflammation are key interconnected contributors to the pathogenesis of atherosclerosis. How hypercholesterolemia initiates vascular inflammation is poorly understood. Here we show in male mice that hypercholesterolemia-driven endothelial activation, monocyte recruitment and atherosclerotic lesion formation are promoted by a crosstalk between macrophages and endothelial cells mediated by the cholesterol metabolite 27-hydroxycholesterol (27HC). The pro-atherogenic actions of macrophage-derived 27HC require endothelial estrogen receptor alpha (ERα) and disassociation of the cytoplasmic scaffolding protein septin 11 from ERα, leading to extranuclear ERα- and septin 11-dependent activation of NF-κB. Furthermore, pharmacologic inhibition of cyp27a1, which generates 27HC, affords atheroprotection by reducing endothelial activation and monocyte recruitment. These findings demonstrate cell-to-cell communication by 27HC, and identify a major causal linkage between the hypercholesterolemia and vascular inflammation that partner to promote atherosclerosis. Interventions interrupting this linkage may provide the means to blunt vascular inflammation without impairing host defense to combat the risk of atherosclerotic cardiovascular disease that remains despite lipid-lowering therapies.


Subject(s)
Atherosclerosis , Hypercholesterolemia , Male , Mice , Animals , Estrogen Receptor alpha/metabolism , Hypercholesterolemia/complications , Hypercholesterolemia/metabolism , Endothelial Cells/metabolism , Septins/metabolism , Cholesterol/metabolism , Atherosclerosis/metabolism , Macrophages/metabolism , Signal Transduction , Inflammation/pathology
4.
Sci Immunol ; 6(62)2021 08 27.
Article in English | MEDLINE | ID: mdl-34452924

ABSTRACT

Under normal conditions, the blood-brain barrier effectively regulates the passage of immune cells into the central nervous system (CNS). However, under pathological conditions such as multiple sclerosis (MS), leukocytes, especially monocytes, infiltrate the CNS where they promote inflammatory demyelination, resulting in paralysis. Therapies targeting the immune cells directly and preventing leukocyte infiltration exist for MS but may compromise the immune system. Here, we explore how apolipoprotein E receptor 2 (ApoER2) regulates vascular adhesion and infiltration of monocytes during inflammation. We induced experimental autoimmune encephalitis in ApoER2 knockout mice and in mice carrying a loss-of-function mutation in the ApoER2 cytoplasmic domain. In both models, paralysis and neuroinflammation were largely abolished as a result of greatly diminished monocyte adherence due to reduced expression of adhesion molecules on the endothelial surface. Our findings expand our mechanistic understanding of the vascular barrier, the regulation of inflammation and vascular permeability, and the therapeutic potential of ApoER2-targeted therapies.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Endothelium, Vascular/immunology , LDL-Receptor Related Proteins/immunology , Monocytes/immunology , Animals , Cell Adhesion/immunology , LDL-Receptor Related Proteins/deficiency , Male , Mice , Mice, Knockout
5.
Arterioscler Thromb Vasc Biol ; 41(4): 1309-1318, 2021 04.
Article in English | MEDLINE | ID: mdl-33626909
6.
Sci Transl Med ; 12(556)2020 08 12.
Article in English | MEDLINE | ID: mdl-32801146

ABSTRACT

Neuroinflammation as a result of immune cell recruitment into the central nervous system (CNS) is a key pathogenic mechanism of multiple sclerosis (MS). However, current anti-inflammatory interventions depleting immune cells or directly targeting their trafficking into the CNS can have serious side effects, highlighting a need for better immunomodulatory strategies. We detected increased Reelin concentrations in the serum of patients with MS, resulting in increased endothelial permeability to leukocytes through increased nuclear factor κB-mediated expression of vascular adhesion molecules. We thus investigated the prophylactic and therapeutic potential of Reelin immunodepletion in experimental autoimmune encephalomyelitis (EAE) and further validated the results in Reelin knockout mice. Removal of plasma Reelin by either approach protected against neuroinflammation and largely abolished the neurological consequences by reducing endothelial permeability and immune cell accumulation in the CNS. Our findings suggest Reelin depletion as a therapeutic approach with an inherent good safety margin for the treatment of MS and other diseases where leukocyte extravasation is a major driver of pathogenicity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Encephalomyelitis , Multiple Sclerosis , Animals , Central Nervous System , Humans , Leukocytes , Mice , Mice, Inbred C57BL , Reelin Protein
7.
Circulation ; 140(24): 2005-2018, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31597453

ABSTRACT

BACKGROUND: Obesity-related hypertension is a common disorder, and attempts to combat the underlying obesity are often unsuccessful. We previously revealed that mice globally deficient in the inhibitory immunoglobulin G (IgG) receptor FcγRIIB are protected from obesity-induced hypertension. However, how FcγRIIB participates is unknown. Studies were designed to determine if alterations in IgG contribute to the pathogenesis of obesity-induced hypertension. METHODS: Involvement of IgG was studied using IgG µ heavy chain-null mice deficient in mature B cells and by IgG transfer. Participation of FcγRIIB was interrogated in mice with global or endothelial cell-specific deletion of the receptor. Obesity was induced by high-fat diet (HFD), and blood pressure (BP) was measured by radiotelemetry or tail cuff. The relative sialylation of the Fc glycan on mouse IgG, which influences IgG activation of Fc receptors, was evaluated by Sambucus nigra lectin blotting. Effects of IgG on endothelial NO synthase were assessed in human aortic endothelial cells. IgG Fc glycan sialylation was interrogated in 3442 human participants by mass spectrometry, and the relationship between sialylation and BP was evaluated. Effects of normalizing IgG sialylation were determined in HFD-fed mice administered the sialic acid precursor N-acetyl-D-mannosamine (ManNAc). RESULTS: Mice deficient in B cells were protected from obesity-induced hypertension. Compared with IgG from control chow-fed mice, IgG from HFD-fed mice was hyposialylated, and it raised BP when transferred to recipients lacking IgG; the hypertensive response was absent if recipients were FcγRIIB-deficient. Neuraminidase-treated IgG lacking the Fc glycan terminal sialic acid also raised BP. In cultured endothelial cells, via FcγRIIB, IgG from HFD-fed mice and neuraminidase-treated IgG inhibited vascular endothelial growth factor activation of endothelial NO synthase by altering endothelial NO synthase phosphorylation. In humans, obesity was associated with lower IgG sialylation, and systolic BP was inversely related to IgG sialylation. Mice deficient in FcγRIIB in endothelium were protected from obesity-induced hypertension. Furthermore, in HFD-fed mice, ManNAc normalized IgG sialylation and prevented obesity-induced hypertension. CONCLUSIONS: Hyposialylated IgG and FcγRIIB in endothelium are critically involved in obesity-induced hypertension in mice, and supportive evidence was obtained in humans. Interventions targeting these mechanisms, such as ManNAc supplementation, may provide novel means to break the link between obesity and hypertension.


Subject(s)
Hexosamines/pharmacology , Hypertension/drug therapy , N-Acetylneuraminic Acid/metabolism , Obesity/drug therapy , Animals , Dietary Supplements , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Hypertension/metabolism , Immunoglobulin G/metabolism , Male , Mice, Inbred C57BL , Obesity/metabolism , Receptors, IgG/metabolism
8.
Blood ; 131(19): 2097-2110, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29500169

ABSTRACT

In the antiphospholipid syndrome (APS), antiphospholipid antibody (aPL) recognition of ß2 glycoprotein I promotes thrombosis, and preclinical studies indicate that this is due to endothelial nitric oxide synthase (eNOS) antagonism via apolipoprotein E receptor 2 (apoER2)-dependent processes. How apoER2 molecularly links these events is unknown. Here, we show that, in endothelial cells, the apoER2 cytoplasmic tail serves as a scaffold for aPL-induced assembly and activation of the heterotrimeric protein phosphatase 2A (PP2A). Disabled-2 (Dab2) recruitment to the apoER2 NPXY motif promotes the activating L309 methylation of the PP2A catalytic subunit by leucine methyl transferase-1. Concurrently, Src homology domain-containing transforming protein 1 (SHC1) recruits the PP2A scaffolding subunit to the proline-rich apoER2 C terminus along with 2 distinct regulatory PP2A subunits that mediate inhibitory dephosphorylation of Akt and eNOS. In mice, the coupling of these processes in endothelium is demonstrated to underlie aPL-invoked thrombosis. By elucidating these intricacies in the pathogenesis of APS-related thrombosis, numerous potential new therapeutic targets have been identified.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antibodies, Antiphospholipid/immunology , Autoantibodies/immunology , Endothelium/metabolism , LDL-Receptor Related Proteins/metabolism , Protein Phosphatase 2/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis Regulatory Proteins , Endothelial Cells/metabolism , Endothelium/immunology , Endothelium, Vascular/metabolism , Humans , Male , Mice , Models, Biological , Multiprotein Complexes , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Thrombosis/etiology , Thrombosis/metabolism , Thrombosis/pathology
9.
Semin Thromb Hemost ; 44(5): 475-482, 2018 Jul.
Article in English | MEDLINE | ID: mdl-28129662

ABSTRACT

The antiphospholipid syndrome (APS) is an autoimmune disorder characterized by an elevated risk for arterial and venous thrombosis and pregnancy-related morbidity. Since the discovery of the disease in 1980s, numerous studies in cell culture systems, in animal models, and in patient populations have been reported, leading to a deeper understanding of the pathogenesis of APS. These studies have determined that circulating autoantibodies, collectively called antiphospholipid antibodies (aPL), the majority of which recognize cell surface proteins attached to the plasma membrane phospholipids, play a causal role in the development of the disease. The binding of aPL to the cell surface antigens triggers interaction of the complex with transmembrane receptors to initiate intracellular signaling in critical cell types, including platelets, monocytes, endothelial cells, and trophoblasts. Subsequent alteration of various cell functions results in inflammation, thrombus formation, and pregnancy complications. Apolipoprotein E receptor 2 (apoER2), a lipoprotein receptor family member, has been implicated as a mediator for aPL actions in platelets and endothelial cells. Nitric oxide (NO) is a signaling molecule known to exert potent antithrombotic, anti-inflammatory, and anti-atherogenic effects. NO insufficiency and oxidative stress have been linked to APS pathogenesis. This review will focus on the recent findings on how apoER2 and dysregulation of NO production contribute to aPL-mediated pathologies in APS.


Subject(s)
Antiphospholipid Syndrome/physiopathology , Female , Humans
10.
J Clin Invest ; 128(1): 309-322, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29202472

ABSTRACT

Type 2 diabetes mellitus (T2DM) is a common complication of obesity. Here, we have shown that activation of the IgG receptor FcγRIIB in endothelium by hyposialylated IgG plays an important role in obesity-induced insulin resistance. Despite becoming obese on a high-fat diet (HFD), mice lacking FcγRIIB globally or selectively in endothelium were protected from insulin resistance as a result of the preservation of insulin delivery to skeletal muscle and resulting maintenance of muscle glucose disposal. IgG transfer in IgG-deficient mice implicated IgG as the pathogenetic ligand for endothelial FcγRIIB in obesity-induced insulin resistance. Moreover, IgG transferred from patients with T2DM but not from metabolically healthy subjects caused insulin resistance in IgG-deficient mice via FcγRIIB, indicating that similar processes may be operative in T2DM in humans. Mechanistically, the activation of FcγRIIB by IgG from obese mice impaired endothelial cell insulin transcytosis in culture and in vivo. These effects were attributed to hyposialylation of the Fc glycan, and IgG from T2DM patients was also hyposialylated. In HFD-fed mice, supplementation with the sialic acid precursor N-acetyl-D-mannosamine restored IgG sialylation and preserved insulin sensitivity without affecting weight gain. Thus, IgG sialylation and endothelial FcγRIIB may represent promising therapeutic targets to sever the link between obesity and T2DM.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Immunoglobulin G/metabolism , Insulin Resistance , Obesity/metabolism , Receptors, IgG/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hexosamines/pharmacology , Immunoglobulin G/genetics , Mice , Mice, Knockout , Obesity/genetics , Obesity/pathology , Receptors, IgG/genetics , Transcytosis/drug effects
11.
PLoS One ; 11(7): e0158757, 2016.
Article in English | MEDLINE | ID: mdl-27463336

ABSTRACT

In the antiphospholipid syndrome (APS), patients produce antiphospholipid antibodies (aPL) that promote thrombosis and adverse pregnancy outcomes. Current therapy with anticoagulation is only partially effective and associated with multiple complications. We previously discovered that aPL recognition of cell surface ß2-glycoprotein I (ß2-GPI) initiates apolipoprotein E receptor 2 (apoER2)-dependent signaling in endothelial cells and in placental trophoblasts that ultimately promotes thrombosis and fetal loss, respectively. Here we sought to identify a monoclonal antibody (mAb) to ß2-GPI that negates aPL-induced processes in cell culture and APS disease endpoints in mice. In a screen measuring endothelial NO synthase (eNOS) activity in cultured endothelial cells, we found that whereas aPL inhibit eNOS, the mAb 1N11 does not, and instead 1N11 prevents aPL action. Coimmunoprecipitation studies revealed that 1N11 decreases pathogenic antibody binding to ß2-GPI, and it blocks aPL-induced complex formation between ß2-GPI and apoER2. 1N11 also prevents aPL antagonism of endothelial cell migration, and in mice it reverses the impairment in reendothelialization caused by aPL, which underlies the non-thrombotic vascular occlusion provoked by disease-causing antibodies. In addition, aPL inhibition of trophoblast proliferation and migration is negated by 1N11, and the more than 6-fold increase in fetal resorption caused by aPL in pregnant mice is prevented by 1N11. Furthermore, the promotion of thrombosis by aPL is negated by 1N11. Thus, 1N11 has been identified as an mAb that attenuates APS-related pregnancy complications and thrombosis in mice. 1N11 may provide an efficacious, mechanism-based therapy to combat the often devastating conditions suffered by APS patients.


Subject(s)
Antibodies, Monoclonal/immunology , Antiphospholipid Syndrome/complications , Pregnancy Complications/prevention & control , Thrombosis/complications , Antiphospholipid Syndrome/prevention & control , Cells, Cultured , Endothelium, Vascular/pathology , Female , Fetal Resorption , Humans , Nitric Oxide Synthase Type III/metabolism , Pregnancy , Thrombosis/prevention & control , Trophoblasts/pathology
12.
Diabetes ; 65(7): 1996-2005, 2016 07.
Article in English | MEDLINE | ID: mdl-27207525

ABSTRACT

Modest elevations in C-reactive protein (CRP) are associated with type 2 diabetes. We previously revealed in mice that increased CRP causes insulin resistance and mice globally deficient in the CRP receptor Fcγ receptor IIB (FcγRIIB) were protected from the disorder. FcγRIIB is expressed in numerous cell types including endothelium and B lymphocytes. Here we investigated how endothelial FcγRIIB influences glucose homeostasis, using mice with elevated CRP expressing or lacking endothelial FcγRIIB. Whereas increased CRP caused insulin resistance in mice expressing endothelial FcγRIIB, mice deficient in the endothelial receptor were protected. The insulin resistance with endothelial FcγRIIB activation was due to impaired skeletal muscle glucose uptake caused by attenuated insulin delivery, and it was associated with blunted endothelial nitric oxide synthase (eNOS) activation in skeletal muscle. In culture, CRP suppressed endothelial cell insulin transcytosis via FcγRIIB activation and eNOS antagonism. Furthermore, in knock-in mice harboring constitutively active eNOS, elevated CRP did not invoke insulin resistance. Collectively these findings reveal that by inhibiting eNOS, endothelial FcγRIIB activation by CRP blunts insulin delivery to skeletal muscle to cause insulin resistance. Thus, a series of mechanisms in endothelium that impairs insulin movement has been identified that may contribute to type 2 diabetes pathogenesis.


Subject(s)
Endothelium, Vascular/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Muscle, Skeletal/metabolism , Receptors, IgG/metabolism , Animals , Aorta/cytology , Aorta/metabolism , Biological Transport , C-Reactive Protein/metabolism , Cattle , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Glucose/metabolism , Homeostasis/physiology , Mice , Mice, Knockout , Mice, Transgenic , Nitric Oxide Synthase Type III/metabolism , Signal Transduction/physiology
13.
Arthritis Rheumatol ; 68(3): 730-739, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26474194

ABSTRACT

OBJECTIVE: Pregnancies in women with the antiphospholipid syndrome (APS) are frequently complicated by fetal loss and intrauterine growth restriction (IUGR). How circulating antiphospholipid antibodies (aPL) cause pregnancy complications in APS is poorly understood. We sought to determine whether the low-density lipoprotein receptor family member apolipoprotein E receptor 2 (ApoER2) mediates trophoblast dysfunction and pregnancy complications induced by aPL. METHODS: Placental and trophoblast ApoER2 expression was evaluated by immunohistochemistry and immunoblotting. Normal human IgG and aPL were purified from healthy individuals and APS patients, respectively. The role of ApoER2 in aPL-induced changes in trophoblast proliferation and migration and in kinase activation was assessed using RNA interference in HTR-8/SVneo cells. The participation of ApoER2 in aPL-induced pregnancy loss and IUGR was evaluated in pregnant ApoER2(+/+) and ApoER2(-/-) mice injected with aPL or normal human IgG. RESULTS: We found that ApoER2 is abundant in human and mouse placental trophoblasts and in multiple trophoblast-derived cell lines, including HTR-8/SVneo cells. ApoER2 and its interaction with the cell surface protein ß2 -glycoprotein I were required for aPL-induced inhibition of cultured trophoblast proliferation and migration. In parallel, aPL antagonism of Akt kinase activation by epidermal growth factor in trophoblasts was mediated by ApoER2. Furthermore, in a murine passive-transfer model of pregnancy complications of APS, ApoER2(-/-) mice were protected from both aPL-induced fetal loss and aPL-induced IUGR. CONCLUSION: ApoER2 plays a major role in the attenuation of trophoblast function by aPL, and the receptor mediates aPL-induced pregnancy complications in vivo in mice. ApoER2-directed interventions can now potentially be developed to combat the pregnancy complications associated with APS.


Subject(s)
Antiphospholipid Syndrome/immunology , LDL-Receptor Related Proteins/immunology , Pregnancy Complications/immunology , Trophoblasts/immunology , Abortion, Spontaneous/immunology , Animals , Cell Line , Female , Humans , Immunoblotting , Immunohistochemistry , Keratin-7/immunology , Mice , Placenta/immunology , Pregnancy , RNA Interference , Transfection , Trophoblasts/cytology , beta 2-Glycoprotein I/immunology
14.
Mol Cell ; 58(5): 845-53, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25982116

ABSTRACT

Protein kinase C has been implicated in the phosphorylation of the erythrocyte/brain glucose transporter, GLUT1, without a clear understanding of the site(s) of phosphorylation and the possible effects on glucose transport. Through in vitro kinase assays, mass spectrometry, and phosphospecific antibodies, we identify serine 226 in GLUT1 as a PKC phosphorylation site. Phosphorylation of S226 is required for the rapid increase in glucose uptake and enhanced cell surface localization of GLUT1 induced by the phorbol ester 12-O-tetradecanoyl-phorbol-13-acetate (TPA). Endogenous GLUT1 is phosphorylated on S226 in primary endothelial cells in response to TPA or VEGF. Several naturally occurring, pathogenic mutations that cause GLUT1 deficiency syndrome disrupt this PKC phosphomotif, impair the phosphorylation of S226 in vitro, and block TPA-mediated increases in glucose uptake. We demonstrate that the phosphorylation of GLUT1 on S226 regulates glucose transport and propose that this modification is important in the physiological regulation of glucose transport.


Subject(s)
Carbohydrate Metabolism, Inborn Errors/genetics , Glucose Transporter Type 1/metabolism , Monosaccharide Transport Proteins/deficiency , Protein Kinase C-alpha/physiology , Amino Acid Sequence , Animals , Biological Transport , Carbohydrate Metabolism, Inborn Errors/enzymology , Cell Line , Endothelial Cells/metabolism , Erythrocytes/metabolism , Glucose/metabolism , Glucose Transporter Type 1/genetics , HeLa Cells , Humans , Molecular Sequence Data , Monosaccharide Transport Proteins/genetics , Mutation, Missense , Phosphorylation , Protein Processing, Post-Translational , Rats , Xenopus laevis
15.
PLoS One ; 10(4): e0124494, 2015.
Article in English | MEDLINE | ID: mdl-25886360

ABSTRACT

Scavenger receptor class B, type I (SR-BI) and its adaptor protein PDZK1 mediate responses to HDL cholesterol in endothelium. Whether the receptor-adaptor protein tandem serves functions in other vascular cell types is unknown. The current work determined the roles of SR-BI and PDZK1 in vascular smooth muscle (VSM). To evaluate possible VSM functions of SR-BI and PDZK1 in vivo, neointima formation was assessed 21 days post-ligation in the carotid arteries of wild-type, SR-BI-/- or PDZK1-/- mice. Whereas neointima development was negligible in wild-type and SR-BI-/-, there was marked neointima formation in PDZK1-/- mice. PDZK1 expression was demonstrated in primary mouse VSM cells, and compared to wild-type cells, PDZK1-/- VSM displayed exaggerated proliferation and migration in response to platelet derived growth factor (PDGF). Tandem affinity purification-mass spectrometry revealed that PDZK1 interacts with breakpoint cluster region kinase (Bcr), which contains a C-terminal PDZ binding sequence and is known to enhance responses to PDGF in VSM. PDZK1 interaction with Bcr in VSM was demonstrated by pull-down and by coimmunoprecipitation, and the augmented proliferative response to PDGF in PDZK1-/- VSM was abrogated by Bcr depletion. Furthermore, compared with wild-type Bcr overexpression, the introduction of a Bcr mutant incapable of PDZK1 binding into VSM cells yielded an exaggerated proliferative response to PDGF. Thus, PDZK1 has novel SR-BI-independent function in VSM that affords protection from neointima formation, and this involves PDZK1 suppression of VSM cell proliferation via an inhibitory interaction with Bcr.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Muscle, Smooth, Vascular/enzymology , Proto-Oncogene Proteins c-bcr/antagonists & inhibitors , Tunica Intima/growth & development , Animals , Cell Movement , Cell Proliferation , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Proto-Oncogene Proteins c-bcr/metabolism
16.
J Biol Chem ; 287(9): 6582-91, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22235125

ABSTRACT

ETS-related gene (ERG) is a member of the ETS transcription factor family. Our previous studies have shown that ERG expression is highly enriched in endothelial cells (EC) both in vitro and in vivo. ERG expression is markedly repressed in response to inflammatory stimuli. It has been shown that ERG is a positive regulator of several EC-restricted genes including VE-cadherin, endoglin, and von Willebrand factor, and a negative regulator of other genes such as interleukin (IL)-8 and intercellular adhesion molecule (ICAM)-1. In this study we have identified a novel role for ERG in the regulation of EC barrier function. ERG knockdown results in marked increases in EC permeability. This is associated with a significant increase of stress fiber and gap formation in EC. Furthermore, we identify CLDN5 as a downstream target of ERG in EC. Thus, our results suggest that ERG plays a pivotal role in regulating EC barrier function and that this effect is mediated in part through its regulation of CLDN5 gene expression.


Subject(s)
Capillary Permeability/physiology , Claudins/genetics , Endothelial Cells/metabolism , Trans-Activators/metabolism , Transcriptional Activation/physiology , Adenoviridae/genetics , Capillary Permeability/drug effects , Claudin-5 , Coronary Vessels/cytology , Cytoskeleton/physiology , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Junctions/physiology , Microvessels/cytology , Mutagenesis, Site-Directed , Pulmonary Artery/cytology , Trans-Activators/genetics , Transcriptional Regulator ERG , Tumor Necrosis Factor-alpha/pharmacology
17.
Cells Tissues Organs ; 195(1-2): 122-43, 2012.
Article in English | MEDLINE | ID: mdl-21997121

ABSTRACT

Considerable progress has been made toward a molecular understanding of how cells form lumen and tube structures in three-dimensional (3D) extracellular matrices (ECM). This progress has occurred through work performed with endothelial and epithelial cell models using both in vitro and in vivo approaches. Despite the apparent similarities between endothelial and epithelial cell lumen and tube formation mechanisms, there are clear distinctions that directly relate to their functional differences. This review will focus on endothelial cell (EC) lumen formation mechanisms which control blood vessel formation during development and postnatal life. Of great interest is that an EC lumen signaling complex has been identified which controls human EC lumen and tube formation in 3D matrices and which coordinates integrin-ECM contacts, cell surface proteolysis, cytoskeletal rearrangements, and cell polarity. This complex consists of the collagen-binding integrin α2ß1, the collagen-degrading membrane-type 1 matrix metalloproteinase (MT1-MMP), junction adhesion molecule (Jam)C, JamB, polarity proteins Par3 and Par6b, and the Rho GTPase Cdc42-GTP. These interacting proteins are necessary to stimulate 3D matrix-specific signaling events (including activation of protein kinase cascades that regulate the actin and microtubule cytoskeletons) to control the formation of EC lumens and tube networks. Also, EC lumen formation is directly coupled to the generation of vascular guidance tunnels, enzymatically generated ECM conduits that facilitate EC tube remodeling and maturation. Mural cells such as pericytes are recruited along EC tubes within these tunnel spaces to control ECM remodeling events resulting in vascular basement membrane matrix assembly, a key step in tube maturation and stabilization.


Subject(s)
Blood Vessels/physiology , Endothelial Cells/cytology , Extracellular Matrix/metabolism , Neovascularization, Physiologic , Animals , Blood Vessels/growth & development , Endothelial Cells/metabolism , Humans
18.
Blood ; 118(4): 1145-53, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21628409

ABSTRACT

ERG is a member of the ETS transcription factor family that is highly enriched in endothelial cells (ECs). To further define the role of ERG in regulating EC function, we evaluated the effect of ERG knock-down on EC lumen formation in 3D collagen matrices. Blockade of ERG using siRNA completely interferes with EC lumen formation. Quantitative PCR (QPCR) was used to identify potential downstream gene targets of ERG. In particular, we identified RhoJ as the Rho GTPase family member that is closely related to Cdc42 as a target of ERG. Knockdown of ERG expression in ECs led to a 75% reduction in the expression of RhoJ. Chromatin immunoprecipitation and transactivation studies demonstrated that ERG could bind to functional sites in the proximal promoter of the RhoJ gene. Knock-down of RhoJ similarly resulted in a marked reduction in the ability of ECs to form lumens. Suppression of either ERG or RhoJ during EC lumen formation was associated with a marked increase in RhoA activation and a decrease in Rac1 and Cdc42 activation and their downstream effectors. Finally, in contrast to other Rho GTPases, RhoJ exhibits a highly EC-restricted expression pattern in several different tissues, including the brain, heart, lung, and liver.


Subject(s)
Blood Vessels/growth & development , Endothelial Cells/metabolism , Trans-Activators/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Blotting, Western , Gene Knockdown Techniques , Humans , Immunoprecipitation , Lasers , Mice , Mice, Nude , Microdissection , Morphogenesis , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transcriptional Regulator ERG
19.
J Clin Invest ; 121(5): 1871-81, 2011 May.
Article in English | MEDLINE | ID: mdl-21490399

ABSTRACT

Cerebral cavernous malformations (CCMs) are a common type of vascular malformation in the brain that are a major cause of hemorrhagic stroke. This condition has been independently linked to 3 separate genes: Krev1 interaction trapped (KRIT1), Cerebral cavernous malformation 2 (CCM2), and Programmed cell death 10 (PDCD10). Despite the commonality in disease pathology caused by mutations in these 3 genes, we found that the loss of Pdcd10 results in significantly different developmental, cell biological, and signaling phenotypes from those seen in the absence of Ccm2 and Krit1. PDCD10 bound to germinal center kinase III (GCKIII) family members, a subset of serine-threonine kinases, and facilitated lumen formation by endothelial cells both in vivo and in vitro. These findings suggest that CCM may be a common tissue manifestation of distinct mechanistic pathways. Nevertheless, loss of heterozygosity (LOH) for either Pdcd10 or Ccm2 resulted in CCMs in mice. The murine phenotype induced by loss of either protein reproduced all of the key clinical features observed in human patients with CCM, as determined by direct comparison with genotype-specific human surgical specimens. These results suggest that CCM may be more effectively treated by directing therapies based on the underlying genetic mutation rather than treating the condition as a single clinical entity.


Subject(s)
Hemangioma, Cavernous, Central Nervous System/genetics , Intracellular Signaling Peptides and Proteins/genetics , Models, Genetic , Mutation , Animals , Apoptosis Regulatory Proteins , Brain/embryology , Brain/metabolism , Gene Expression Regulation, Developmental , Genotype , Humans , KRIT1 Protein , Loss of Heterozygosity , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Microtubule-Associated Proteins/genetics , Phenotype , Proto-Oncogene Proteins/genetics , Time Factors
20.
Int Rev Cell Mol Biol ; 288: 101-65, 2011.
Article in English | MEDLINE | ID: mdl-21482411

ABSTRACT

Many studies reveal a fundamental role for extracellular matrix-mediated signaling through integrins and Rho GTPases as well as matrix metalloproteinases (MMPs) in the molecular control of vascular tube morphogenesis in three-dimensional (3D) tissue environments. Recent work has defined an endothelial cell (EC) lumen signaling complex of proteins that controls these vascular morphogenic events. These findings reveal a signaling interdependence between Cdc42 and MT1-MMP to control the 3D matrix-specific process of EC tubulogenesis. The EC tube formation process results in the creation of a network of proteolytically generated vascular guidance tunnels in 3D matrices that are utilized to remodel EC-lined tubes through EC motility and could facilitate processes such as flow-induced remodeling and arteriovenous EC sorting and differentiation. Within vascular guidance tunnels, key dynamic interactions occur between ECs and pericytes to affect vessel remodeling, diameter, and vascular basement membrane matrix assembly, a fundamental process necessary for endothelial tube maturation and stabilization. Thus, the EC lumen and tube formation mechanism coordinates the concomitant establishment of a network of vascular tubes within tunnel spaces to allow for flow responsiveness, EC-mural cell interactions, and vascular extracellular matrix assembly to control the development of the functional microcirculation.


Subject(s)
Endothelial Cells/physiology , Endothelium, Vascular/physiology , Neovascularization, Physiologic/physiology , Animals , Cell Adhesion , Cell Polarity , Cells, Cultured , Coculture Techniques , Endothelial Cells/cytology , Endothelium, Vascular/cytology , Extracellular Matrix/metabolism , Humans , Matrix Metalloproteinase 14 , Matrix Metalloproteinases/metabolism , Morphogenesis , Pericytes/cytology , Pericytes/metabolism , Protein Kinase C-epsilon/metabolism , Signal Transduction/physiology , Tissue Inhibitor of Metalloproteinases/metabolism , cdc42 GTP-Binding Protein/metabolism , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...