Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
2.
Kidney Med ; 5(1): 100567, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36636202

ABSTRACT

Rationale & Objective: Access patency outcomes for arteriovenous fistulas (AVFs) as compared with arteriovenous grafts (AVGs) in patients receiving hemodialysis (HD) who have achieved a functioning permanent access are not fully explored. Study Design: Observational cohort study. Setting & Population: Fee-for-service Medicare beneficiaries aged ≥18 years with kidney failure who were newly using a permanent access for maintenance HD from the United States Renal Data System (2010-2015). Patients using an oral anticoagulant were excluded. Exposure: AVG or AVF. Outcomes: Loss of primary unassisted, primary assisted, and secondary patency. Analytical Approach: Outcomes were characterized using cumulative incidence curves, and HRs adjusted for sociodemographic and clinical factors were estimated for the comparison of AVF versus AVG. Results: The cohort included 60,329 and 17,763 patients newly using an AVF and AVG, respectively, for HD. Over 3 years of follow-up, AVG users, compared to AVF users, had a higher cumulative incidence of loss of primary unassisted patency (87% vs 69%; HR, 1.56; 95% CI, 1.52-1.60), loss of primary assisted patency (69% vs 25%; HR, 3.79; 95% CI, 3.67-3.92), and loss of secondary patency (22% vs 10%; HR, 2.03; 95% CI, 1.92-2.16). Stratified analyses revealed differences by subgroups; in particular, incidence of patency loss was higher among patients who underwent prior interventions to maintain prefunctional access patency and Black patients. Limitations: This analysis focused on outcomes occurring after first successful use of a permanent access and thus does not inform about risk of patency loss during access maturation. Conclusions: Among patients with kidney failure who successfully used a permanent access for HD, patency loss was consistently substantially higher in those using AVGs compared with AVFs. New interventions, such as prophylactic drugs, are needed to improve access longevity and reduce the need for invasive interventions, particularly among patients unable to receive a fistula.

3.
Kidney Med ; 4(6): 100456, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35706716

ABSTRACT

Rationale & Objective: The risks of major bleeding, thrombosis, and cardiovascular events are elevated in patients receiving maintenance hemodialysis (HD). Our objective was to compare the risk of these outcomes in HD according to the permanent vascular access type. Study Design: Observational cohort study. Setting & Participants: Using data from the United States Renal Data System (2010-2015), we included patients with kidney failure who were greater than 18 years, had Medicare as the primary payer, were not using an oral anticoagulant, and were newly using an arteriovenous (AV) access for HD. Exposure: AV graft (AVG) or AV fistula (AVF). Outcomes: Major bleeding, venous thromboembolism, ischemic stroke, myocardial infarction, cardiovascular death, and critical limb ischemia. Analytical Approach: Comparing 17,763 AVG and 60,329 AVF users, we estimated the 3-year incidence rates and incidence rate ratios (IRRs) of each outcome using Poisson regression. IRRs were adjusted for sociodemographic and clinical covariates. Results: The use of an AVG, compared with that of an AVF, was associated with an increased risk of venous thromboembolism (10.8 vs 5.3 events per 100 person-years; adjusted IRR, 1.74; 95% CI, 1.63-1.85) but not with the risk of major bleeding (IRR, 1.04; 95% CI, 0.93-1.17). The use of an AVG was also potentially associated with a slightly increased risk of cardiovascular death (IRR, 1.09; 95% CI, 1.01-1.16). Limitations: This analysis focused on patients with a functioning AV access; adverse events that may occur during access maturation should also be considered when selecting a vascular access. Conclusions: The use of an AVG, relative to an AVF, in HD is associated with an increased risk of venous thromboembolism. Given recent guidelines emphasizing selection of the "right access" for the "right patient," the results of this study should potentially be considered as one additional factor when selecting the optimal access for HD.

5.
J Clin Invest ; 126(4): 1282-99, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26974158

ABSTRACT

Abdominal aortic aneurysm (AAA) is a major cause of morbidity and mortality; however, the mechanisms that are involved in disease initiation and progression are incompletely understood. Extracellular matrix proteins play an integral role in modulating vascular homeostasis in health and disease. Here, we determined that the expression of the matricellular protein CCN3 is strongly reduced in rodent AAA models, including angiotensin II-induced AAA and elastase perfusion-stimulated AAA. CCN3 levels were also reduced in human AAA biopsies compared with those in controls. In murine models of induced AAA, germline deletion of Ccn3 resulted in severe phenotypes characterized by elastin fragmentation, vessel dilation, vascular inflammation, dissection, heightened ROS generation, and smooth muscle cell loss. Conversely, overexpression of CCN3 mitigated both elastase- and angiotensin II-induced AAA formation in mice. BM transplantation experiments suggested that the AAA phenotype of CCN3-deficient mice is intrinsic to the vasculature, as AAA was not exacerbated in WT animals that received CCN3-deficient BM and WT BM did not reduce AAA severity in CCN3-deficient mice. Genetic and pharmacological approaches implicated the ERK1/2 pathway as a critical regulator of CCN3-dependent AAA development. Together, these results demonstrate that CCN3 is a nodal regulator in AAA biology and identify CCN3 as a potential therapeutic target for vascular disease.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , MAP Kinase Signaling System , Nephroblastoma Overexpressed Protein/metabolism , Angiotensin II/adverse effects , Angiotensin II/pharmacology , Animals , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/therapy , Disease Models, Animal , Elastin/metabolism , Gene Deletion , Humans , Mice , Mice, Knockout , Nephroblastoma Overexpressed Protein/genetics , Pancreatic Elastase/toxicity
7.
Nat Commun ; 6: 7231, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26040986

ABSTRACT

Circadian control of nutrient availability is critical to efficiently meet the energetic demands of an organism. Production of bile acids (BAs), which facilitate digestion and absorption of nutrients, is a major regulator of this process. Here we identify a KLF15-Fgf15 signalling axis that regulates circadian BA production. Systemic Klf15 deficiency disrupted circadian expression of key BA synthetic enzymes, tissue BA levels and triglyceride/cholesterol absorption. Studies in liver-specific Klf15-knockout mice suggested a non-hepatic basis for regulation of BA production. Ileal Fgf15 is a potent inhibitor of BA synthesis. Using a combination of biochemical, molecular and functional assays (including ileectomy and bile duct catheterization), we identify KLF15 as the first endogenous negative regulator of circadian Fgf15 expression. Elucidation of this novel pathway controlling circadian BA production has important implications for physiologic control of nutrient availability and metabolic homeostasis.


Subject(s)
Bile Acids and Salts/biosynthesis , Circadian Rhythm , DNA-Binding Proteins/genetics , Fibroblast Growth Factors/genetics , Hepatocytes/metabolism , Ileum/metabolism , Liver/metabolism , RNA, Messenger/metabolism , Transcription Factors/genetics , Animals , Blotting, Western , DNA-Binding Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factors/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Kruppel-Like Transcription Factors , Mice , Mice, Knockout , Receptor, Fibroblast Growth Factor, Type 4/genetics , Transcription Factors/metabolism
8.
J Neuroimmunol ; 274(1-2): 234-9, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25052583

ABSTRACT

Cells of the innate immune system are important mediators of multiple sclerosis (MS). We have previously identified Kruppel-like factor 2 (KLF2) as a critical negative regulator of myeloid activation in the setting of bacterial infection and sepsis, but the role of myeloid KLF2 in MS has not been investigated. In this study, myeloid KLF2 deficient mice exhibited more severe neurological dysfunction and increased spinal cord demyelination and neuroinflammation in experimental autoimmune encephalomyelitis. This study represents the first description of a significant role of myeloid KLF2 in neuroinflammation, identifying KLF2 as a potential target for further investigation in patients with MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Multiple Sclerosis/immunology , Animals , Demyelinating Diseases/genetics , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Macrophages/immunology , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Spinal Cord/immunology , Spinal Cord/pathology
9.
Blood ; 123(24): 3828-31, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24771858

ABSTRACT

Multiple myeloma confers a high risk for vascular thrombosis, a risk that is increased by treatment with immunomodulatory agents. Strikingly, inclusion of the proteasome inhibitor bortezomib reduces thrombotic risk, yet the molecular basis for this observation remains unknown. Here, we show that bortezomib prolongs thrombosis times in the carotid artery photochemical injury assay in normal mice. Cell-based studies show that bortezomib increases expression of the transcription factor Kruppel-like factor 2 (KLF2) in multiple cell types. Global postnatal overexpression of KLF2 (GL-K2-TG) increased time to thrombosis, and global postnatal deletion of KLF2 (GL-K2-KO) conferred an antiparallel effect. Finally, studies in GL-K2-KO mice showed that the thromboprotective effect of bortezomib is KLF2 dependent. These findings identify a transcriptional basis for the antithrombotic effects of bortezomib.


Subject(s)
Boronic Acids/pharmacology , Carotid Artery Thrombosis/prevention & control , Cytoprotection/genetics , Kruppel-Like Transcription Factors/physiology , Pyrazines/pharmacology , Animals , Bortezomib , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/genetics , Carotid Artery Thrombosis/pathology , Cells, Cultured , Coronary Occlusion/genetics , Coronary Occlusion/pathology , Coronary Occlusion/prevention & control , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Whole Blood Coagulation Time
11.
CNS Neurol Disord Drug Targets ; 12(3): 325-37, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23469858

ABSTRACT

Loss of integrity of the blood-brain barrier (BBB) in stroke victims initiates a devastating cascade of events including extravasation of blood-borne molecules, water, and inflammatory cells deep into brain parenchyma. Thus, it is important to identify mechanisms by which BBB integrity can be maintained in the face of ischemic injury in experimental stroke. We previously demonstrated that the phylogenetically conserved small heat shock protein 27 (HSP27) protects against transient middle cerebral artery occlusion (tMCAO). Here we show that HSP27 transgenic overexpression also maintains the integrity of the BBB in mice subjected to tMCAO. Extravasation of endogenous IgG antibodies and exogenous FITC-albumin into the brain following tMCAO was reduced in transgenic mice, as was total brain water content. HSP27 overexpression abolished the appearance of TUNEL-positive profiles in microvessel walls. Transgenics also exhibited less loss of microvessel proteins following tMCAO. Notably, primary endothelial cell cultures were rescued from oxygen-glucose deprivation (OGD) by lentiviral HSP27 overexpression according to four viability assays, supporting a direct effect on this cell type. Finally, HSP27 overexpression reduced the appearance of neutrophils in the brain and inhibited the secretion of five cytokines. These findings reveal a novel role for HSP27 in attenuating ischemia/reperfusion injury - the maintenance of BBB integrity. Endogenous upregulation of HSP27 after ischemia in wild-type animals may exert similar protective functions and warrants further investigation. Exogenous enhancement of HSP27 by rational drug design may lead to future therapies against a host of injuries, including but not limited to a harmful breach in brain vasculature.


Subject(s)
Blood-Brain Barrier/physiology , HSP27 Heat-Shock Proteins/physiology , Infarction, Middle Cerebral Artery/physiopathology , Animals , Brain Edema/genetics , Brain Edema/physiopathology , Cell Survival/physiology , Cytokines/metabolism , Endothelial Cells/physiology , HSP27 Heat-Shock Proteins/genetics , Male , Mice , Mice, Transgenic , Primary Cell Culture , Up-Regulation/physiology
12.
Am J Physiol Heart Circ Physiol ; 304(6): H796-805, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23335794

ABSTRACT

During an ischemic stroke normal brain endothelial function is perturbed, resulting in blood brain barrier (BBB) breakdown with subsequent infiltration of activated inflammatory blood cells, ultimately leading to neuronal cell death. Kruppel-like factor 2 (KLF2) is regulated by flow, is highly expressed in vascular endothelial cells (ECs), and serves as a key molecular switch regulating endothelial function and promoting vascular health. In this study we sought to determine the role of KLF2 in cerebrovascular function and the pathogenesis of ischemic stroke. Transient middle cerebral artery occlusion was performed in KLF2-deficient (KLF2(-/-)), KLF2 overexpressing (KLF2(tg)), and control mice, and stroke volume was analyzed. BBB function was assessed in vivo by real-time neuroimaging using positron emission tomography and Evan's blue dye assay. KLF2(-/-) mice exhibited significantly larger strokes and impairment in BBB function. In contrast, KLF2(tg) mice were protected against ischemic stroke and demonstrated preserved BBB function. In concordance, gain- and loss-of-function studies in primary brain microvascular ECs using transwell assays revealed KLF2 to be BBB protective. Mechanistically, KLF2 was demonstrated, both in vitro and in vivo, to regulate the critical BBB tight junction factor occludin. These data are first to identify endothelial KLF2 as a key regulator of the BBB and a novel neuroprotective factor in ischemic stroke.


Subject(s)
Blood-Brain Barrier/metabolism , Infarction, Middle Cerebral Artery/metabolism , Kruppel-Like Transcription Factors/metabolism , Animals , Blood-Brain Barrier/physiology , Cell Line , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Infarction, Middle Cerebral Artery/diagnostic imaging , Kruppel-Like Transcription Factors/genetics , Mice , Multimodal Imaging , Occludin/genetics , Occludin/metabolism , Positron-Emission Tomography , Tomography, X-Ray Computed
13.
J Clin Invest ; 122(12): 4727-31, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23160196

ABSTRACT

The endothelium regulates vascular homeostasis, and endothelial dysfunction is a proximate event in the pathogenesis of atherothrombosis. Stimulation of the endothelium with proinflammatory cytokines or exposure to hemodynamic-induced disturbed flow leads to a proadhesive and prothrombotic phenotype that promotes atherothrombosis. In contrast, exposure to arterial laminar flow induces a gene program that confers a largely antiadhesive, antithrombotic effect. The molecular basis for this differential effect on endothelial function remains poorly understood. While recent insights implicate Kruppel-like factors (KLFs) as important regulators of vascular homeostasis, the in vivo role of these factors in endothelial biology remains unproven. Here, we show that endothelial KLF4 is an essential determinant of atherogenesis and thrombosis. Using in vivo EC-specific KLF4 overexpression and knockdown murine models, we found that KLF4 induced an antiadhesive, antithrombotic state. Mechanistically, we demonstrated that KLF4 differentially regulated pertinent endothelial targets via competition for the coactivator p300. These observations provide cogent evidence implicating endothelial KLFs as essential in vivo regulators of vascular function in the adult animal.


Subject(s)
Atherosclerosis/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Kruppel-Like Transcription Factors/physiology , Thrombosis/metabolism , Animals , Cells, Cultured , Gene Expression Regulation , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding , Protein Interaction Domains and Motifs , Thrombosis/genetics , Vasculitis/metabolism , p300-CBP Transcription Factors/metabolism
14.
Arterioscler Thromb Vasc Biol ; 32(11): 2640-3, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22995518

ABSTRACT

OBJECTIVE: The endothelial response elicited by the G-protein-coupled receptor pathway involving apelin and APJ predicts an overall vasoprotective effect. As a number of downstream endothelial targets of apelin/APJ signaling are also known to be targeted by statins (3-hydroxy-3-methyl-glutaryl [HMG]-CoA reductase inhibitors) as potential mediators of their known pleiotropic effects, we evaluated for the involvement of apelin/APJ signaling in statin endothelial effects. METHODS AND RESULTS: We found that disruption of apelin/APJ signaling in endothelial cells leads to significantly decreased expression of Kruppel-like factor 2, endothelial nitric oxide synthase, and thrombomodulin. We found that statin-mediated induction of Kruppel-like factor 2, endothelial nitric oxide synthase, and thrombomodulin expression, as well as inhibition of monocyte-endothelial adhesion, was abrogated by concurrent apelin knockdown. Moreover, we found that statins can transcriptionally regulate APJ in a Kruppel-like factor 2-dependent manner, demonstrating the presence of a positive-feedback loop. CONCLUSIONS: Our findings provide a novel mechanism by which the apelin/APJ pathway serves as a critical intermediary that links statin to its pleiotropic effects in regulating endothelial gene targets and function.


Subject(s)
Human Umbilical Vein Endothelial Cells/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Receptors, G-Protein-Coupled/drug effects , Signal Transduction/drug effects , Adipokines , Animals , Apelin , Apelin Receptors , COS Cells , Cell Adhesion/drug effects , Chlorocebus aethiops , Coculture Techniques , Fatty Acids, Monounsaturated/pharmacology , Fluorobenzenes/pharmacology , Fluvastatin , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Indoles/pharmacology , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Knockout , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Pyrimidines/pharmacology , RNA Interference , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Rosuvastatin Calcium , Simvastatin/pharmacology , Sulfonamides/pharmacology , Thrombomodulin/genetics , Thrombomodulin/metabolism , Time Factors , Transfection
15.
Arterioscler Thromb Vasc Biol ; 31(7): 1476-84, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21677290

ABSTRACT

A complex and diverse vascular system is requisite for the survival of higher organisms. The process of vascular development is highly regulated, involving the de novo formation of vessels (vasculogenesis), followed by expansion and remodeling of the primitive vasculature (angiogenesis), culminating in differentiation of endothelial phenotypes, as found in the mature vascular system. Over the last decade, significant advances have been made in understanding the molecular regulation of endothelial cell development and differentiation. Endothelial development, in particular the mechanisms in play during vasculogenesis and angiogenesis, is discussed in a sister review to this article. This review highlights the key pathways governing in endothelial differentiation, with a focus on the major molecular mechanisms of endothelial specification and heterogeneity.


Subject(s)
Cell Differentiation , Cell Lineage , Endothelial Cells/physiology , Neovascularization, Physiologic , Animals , Arteries/embryology , Body Patterning , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation , Gene Expression Regulation, Developmental , Humans , Lymphatic Vessels/embryology , Neovascularization, Physiologic/genetics , Phenotype , Signal Transduction , Veins/embryology
16.
Immunity ; 34(5): 715-28, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21565532

ABSTRACT

Precise control of myeloid cell activation is required for optimal host defense. However, this activation process must be under exquisite control to prevent uncontrolled inflammation. Herein, we identify the Kruppel-like transcription factor 2 (KLF2) as a potent regulator of myeloid cell activation in vivo. Exposure of myeloid cells to hypoxia and/or bacterial products reduced KLF2 expression while inducing hypoxia inducible factor-1α (HIF-1α), findings that were recapitulated in human septic patients. Myeloid KLF2 was found to be a potent inhibitor of nuclear factor-kappaB (NF-κB)-dependent HIF-1α transcription and, consequently, a critical determinant of outcome in models of polymicrobial infection and endotoxemia. Collectively, these observations identify KLF2 as a tonic repressor of myeloid cell activation in vivo and an essential regulator of the innate immune system.


Subject(s)
Bacterial Infections/immunology , Kruppel-Like Transcription Factors/immunology , Shock, Septic/immunology , Animals , Bacterial Infections/microbiology , Cell Line , Female , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Immunity, Innate , Kruppel-Like Transcription Factors/genetics , Lipopolysaccharides/immunology , Male , Mice , Mice, Transgenic , Myeloid Cells/immunology , NF-kappa B/immunology
17.
J Cell Commun Signal ; 4(3): 141-53, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21063504

ABSTRACT

The vascular endothelium plays a fundamental role in the health and disease of the cardiovascular system. The molecular mechanisms regulating endothelial homeostasis, however, remain incompletely understood. CCN3, a member of the CCN (Cyr61, Ctgf, Nov) family of cell growth and differentiation regulators, has been shown to play an important role in numerous cell types. The function of CCN3 in endothelial cells has yet to be elucidated. Immunohistochemical analysis of CCN3 expression in mouse tissues revealed robust immunoreactivity in the endothelium of large arteries, small resistance vessels, and veins. We found that CCN3 expression in human umbilical vein endothelial cells (HUVECs) is transcriptionally induced by laminar shear stress (LSS) and HMG CoA-reductase inhibitors (statins). Promoter analyses identified the transcription factor Kruppel-like factor 2 (KLF2) as a direct regulator of CCN3 expression. In contrast to LSS, proinflammatory cytokines reduced CCN3 expression. Adenoviral overexpression of CCN3 in HUVEC markedly inhibited the cytokine-mediated induction of vascular adhesion molecule-1 (VCAM-1). Consistent with this observation, CCN3 significantly reduced monocyte adhesion. Conversely, CCN3 knockdown in HUVECs resulted in enhancement of cytokine-induced VCAM-1 expression. Concordant effects were observed on monocyte adhesion. Gain and loss-of-function mechanistic studies demonstrated that CCN3 negatively regulates nuclear factor kappaB (NF-κB) activity by reducing its translocation into the nucleus and subsequent binding to the VCAM-1 promoter, suggesting that CCN3's anti-inflammatory effects occur secondary to inhibition of NF-κB nuclear accumulation. This study identifies CCN3 as a novel regulator of endothelial proinflammatory activation.

18.
Arterioscler Thromb Vasc Biol ; 30(10): 1952-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20651277

ABSTRACT

OBJECTIVE: A central function of the endothelium is to serve as a selective barrier that regulates fluid and solute exchange. Although perturbation of barrier function can contribute to numerous disease states, our understanding of the molecular mechanisms regulating this aspect of endothelial biology remains incompletely understood. Accumulating evidence implicates the Kruppel-like factor 2 (KLF2) as a key regulator of endothelial function. However, its role in vascular barrier function is unknown. METHODS AND RESULTS: To assess the role of KLF2 in vascular barrier function in vivo, we measured the leakage of Evans blue dye into interstitial tissues of the mouse ear after treatment with mustard oil. By comparison with KLF2(+/+) mice, KLF2(+/-) mice exhibited a significantly higher degree of vascular leak. In accordance with our in vivo observation, adenoviral overexpression of KLF2 in human umbilical vein endothelial cells strongly attenuated the increase of endothelial leakage by thrombin and H(2)O(2) as measured by fluorescein isothiocyanate dextrans (FITC-dextran) passage. Conversely, KLF2 deficiency in human umbilical vein endothelial cells and primary endothelial cells derived from KLF2(+/-) mice exhibited a marked increase in thrombin and H(2)O(2)-induced permeability. Mechanistically, our studies indicate that KLF2 confers barrier-protection via differential effects on the expression of key junction protein occludin and modification of a signaling molecule (myosin light chain) that regulate endothelial barrier integrity. CONCLUSIONS: These observations identify KLF2 as a novel transcriptional regulator of vascular barrier function.


Subject(s)
Endothelial Cells/physiology , Kruppel-Like Transcription Factors/physiology , Animals , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cells, Cultured , Endothelial Cells/drug effects , Gene Expression , Humans , Hydrogen Peroxide/pharmacology , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Knockout , Myosin Light Chains/physiology , Occludin , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thrombin/pharmacology , Transfection
19.
J Biol Chem ; 284(31): 20522-30, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19491109

ABSTRACT

Hypoxia-inducible factor 1 (HIF-1) is a central regulator of the hypoxic response in many cell types. In endothelial cells, HIF-1 induces the expression of key proangiogenic factors to promote angiogenesis. Recent studies have identified Kruppel-like factor 2 (KLF2) as a potent inhibitor of angiogenesis. However, the role of KLF2 in regulating HIF-1 expression and function has not been evaluated. KLF2 expression was induced acutely by hypoxia in endothelial cells. Adenoviral overexpression of KLF2 inhibited hypoxia-induced expression of HIF-1alpha and its target genes such as interleukin 8, angiopoietin-2, and vascular endothelial growth factor in endothelial cells. Conversely, knockdown of KLF2 increased expression of HIF-1alpha and its targets. Furthermore, KLF2 inhibited hypoxia-induced endothelial tube formation, whereas endothelial cells from mice with haploinsufficiency of KLF2 showed increased tube formation in response to hypoxia. Consistent with this ex vivo observation, KLF2 heterozygous mice showed increased microvessel density in the brain. Mechanistically, KLF2 promoted HIF-1alpha degradation in a von Hippel-Lindau protein-independent but proteasome-dependent manner. Finally, KLF2 disrupted the interaction between HIF-1alpha and its chaperone Hsp90, suggesting that KLF2 promotes degradation of HIF-1alpha by affecting its folding and maturation. These observations identify KLF2 as a novel inhibitor of HIF-1alpha expression and function. Therefore, KLF2 may be a target for modulating the angiogenic response in disease states.


Subject(s)
Endothelium/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kruppel-Like Transcription Factors/metabolism , Animals , Cell Hypoxia , Cell Line , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Neovascularization, Physiologic , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Processing, Post-Translational , Tumor Suppressor Protein p53/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
20.
Circ Res ; 103(7): 690-3, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18757824

ABSTRACT

Krüppel-like factor (KLF)2 is a central regulator of endothelial and monocyte/macrophage gene expression and function in vitro. Although the composite effects of KLF2 in these 2 cell types predict that it likely inhibits vascular inflammation, the role of KLF2 in this process in vivo is uncharacterized. In this study, we provide evidence that hemizygous deficiency of KLF2 increased diet-induced atherosclerosis in apolipoprotein E-deficient mice. Our studies highlight an important role for KLF2 in primary macrophage foam cell formation via the potential regulation of the key lipid binding protein adipocyte protein 2/fatty acid-binding protein 4. These novel observations establish that KLF2 is an atheroprotective factor.


Subject(s)
Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Diet, Atherogenic , Fatty Acid-Binding Proteins/metabolism , Foam Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/chemically induced , Atherosclerosis/genetics , Endothelial Cells/metabolism , Fatty Acid-Binding Proteins/genetics , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Monocytes/metabolism , Vasculitis/chemically induced , Vasculitis/genetics , Vasculitis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...