Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Publication year range
1.
Nature ; 568(7752): 336-343, 2019 04.
Article in English | MEDLINE | ID: mdl-30996318

ABSTRACT

The brains of humans and other mammals are highly vulnerable to interruptions in blood flow and decreases in oxygen levels. Here we describe the restoration and maintenance of microcirculation and molecular and cellular functions of the intact pig brain under ex vivo normothermic conditions up to four hours post-mortem. We have developed an extracorporeal pulsatile-perfusion system and a haemoglobin-based, acellular, non-coagulative, echogenic, and cytoprotective perfusate that promotes recovery from anoxia, reduces reperfusion injury, prevents oedema, and metabolically supports the energy requirements of the brain. With this system, we observed preservation of cytoarchitecture; attenuation of cell death; and restoration of vascular dilatory and glial inflammatory responses, spontaneous synaptic activity, and active cerebral metabolism in the absence of global electrocorticographic activity. These findings demonstrate that under appropriate conditions the isolated, intact large mammalian brain possesses an underappreciated capacity for restoration of microcirculation and molecular and cellular activity after a prolonged post-mortem interval.


Subject(s)
Autopsy , Brain/blood supply , Brain/cytology , Cerebrovascular Circulation , Microcirculation , Swine , Animals , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Caspase 3/metabolism , Cell Survival , Cerebral Arteries/physiology , Disease Models, Animal , Hypoxia, Brain/metabolism , Hypoxia, Brain/pathology , Inflammation/metabolism , Inflammation/pathology , Neuroglia/cytology , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Perfusion , Reperfusion Injury/prevention & control , Swine/blood , Synapses/metabolism , Synapses/pathology , Time Factors , Vasodilation
2.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Article in English | MEDLINE | ID: mdl-34911761

ABSTRACT

Arterial remodeling is an important adaptive mechanism that maintains normal fluid shear stress in a variety of physiologic and pathologic conditions. Inward remodeling, a process that leads to reduction in arterial diameter, plays a critical role in progression of such common diseases as hypertension and atherosclerosis. Yet, despite its pathogenic importance, molecular mechanisms controlling inward remodeling remain undefined. Mitogen-activated protein kinases (MAPKs) perform a number of functions ranging from control of proliferation to migration and cell-fate transitions. While the MAPK ERK1/2 signaling pathway has been extensively examined in the endothelium, less is known about the role of the MEKK3/ERK5 pathway in vascular remodeling. To better define the role played by this signaling cascade, we studied the effect of endothelial-specific deletion of its key upstream MAP3K, MEKK3, in adult mice. The gene's deletion resulted in a gradual inward remodeling of both pulmonary and systematic arteries, leading to spontaneous hypertension in both vascular circuits and accelerated progression of atherosclerosis in hyperlipidemic mice. Molecular analysis revealed activation of TGFß-signaling both in vitro and in vivo. Endothelial-specific TGFßR1 knockout prevented inward arterial remodeling in MEKK3 endothelial knockout mice. These data point to the unexpected participation of endothelial MEKK3 in regulation of TGFßR1-Smad2/3 signaling and inward arterial remodeling in artery diseases.


Subject(s)
Hypertension, Pulmonary/pathology , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 3/metabolism , Transforming Growth Factor beta/metabolism , Vascular Remodeling/physiology , Animals , Gene Deletion , Gene Expression Regulation/drug effects , Genotype , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Humans , Hypertension, Pulmonary/metabolism , Ischemia , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 3/genetics , Mice , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Selective Estrogen Receptor Modulators/toxicity , Signal Transduction , Tamoxifen/toxicity , Transforming Growth Factor beta/genetics
3.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34504019

ABSTRACT

Endothelial cell (EC) sensing of wall fluid shear stress (FSS) from blood flow governs vessel remodeling to maintain FSS at a specific magnitude or set point in healthy vessels. Low FSS triggers inward remodeling to restore normal FSS but the regulatory mechanisms are unknown. In this paper, we describe the signaling network that governs inward artery remodeling. FSS induces Smad2/3 phosphorylation through the type I transforming growth factor (TGF)-ß family receptor Alk5 and the transmembrane protein Neuropilin-1, which together increase sensitivity to circulating bone morphogenetic protein (BMP)-9. Smad2/3 nuclear translocation and target gene expression but not phosphorylation are maximal at low FSS and suppressed at physiological high shear. Reducing flow by carotid ligation in rodents increases Smad2/3 nuclear localization, while the resultant inward remodeling is blocked by the EC-specific deletion of Alk5. The flow-activated MEKK3/Klf2 pathway mediates the suppression of Smad2/3 nuclear translocation at high FSS, mainly through the cyclin-dependent kinase (CDK)-2-dependent phosphosphorylation of the Smad linker region. Thus, low FSS activates Smad2/3, while higher FSS blocks nuclear translocation to induce inward artery remodeling, specifically at low FSS. These results are likely relevant to inward remodeling in atherosclerotic vessels, in which Smad2/3 is activated through TGF-ß signaling.


Subject(s)
Carotid Arteries/physiology , Carotid Artery Diseases/prevention & control , Endothelial Cells/physiology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Stress, Mechanical , Vascular Remodeling , Animals , Carotid Arteries/cytology , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Endothelial Cells/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Signal Transduction , Smad2 Protein/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
4.
J Nucl Cardiol ; 25(6): 2096-2111, 2018 12.
Article in English | MEDLINE | ID: mdl-28695406

ABSTRACT

BACKGROUND: Currently, there is no established non-invasive imaging approach to directly evaluate myocardial microcirculatory function in order to diagnose microvascular disease independent of co-existing epicardial disease. In this work, we developed a methodological framework for quantification of intramyocardial blood volume (IMBV) as a novel index of microcirculatory function with SPECT/CT imaging of 99mTc-labeled red blood cells (RBCs). METHODS: Dual-gated myocardial SPECT/CT equilibrium imaging of 99mTc-RBCs was performed on twelve canines under resting conditions. Five correction schemes were studied: cardiac gating with no other corrections (CG), CG with attenuation correction (CG + AC), CG + AC with scatter correction (CG + AC + SC), dual cardiorespiratory gating with AC + SC (DG + AC + SC), and DG + AC + SC with partial volume correction (DG + AC + SC + PVC). Quantification of IMBV using each approach was evaluated in comparison to those obtained from all corrections. The in vivo SPECT estimates of IMBV values were validated against those obtained from ex vivo microCT imaging of the casted hearts. RESULTS: The estimated IMBV with all corrections was 0.15 ± 0.03 for the end-diastolic phase and 0.11 ± 0.03 for the end-systolic phase. The cycle-dependent change in IMBV (ΔIMBV) with all corrections was 23.9 ± 8.6%. Schemes that applied no correction or partial correction resulted in significant over-estimation of IMBV and significant under-underestimation of ΔIMBV. Estimates of IMBV and ΔIMBV using all corrections were consistent with values reported in the literature using invasive techniques. In vivo SPECT estimates of IMBV strongly correlated (R2 ≥ 0.70) with ex vivo measures for the various correction schemes, while the fully corrected scheme yielded the smallest bias. CONCLUSIONS: Non-invasive quantification of IMBV is feasible using 99mTc-RBCs SPECT/CT imaging, however, requires full compensation of physical degradation factors.


Subject(s)
Blood Volume , Coronary Circulation/physiology , Microcirculation/physiology , Single Photon Emission Computed Tomography Computed Tomography/methods , Animals , Dogs , Erythrocytes , Female , Hemodynamics , Technetium , X-Ray Microtomography
5.
Proc Natl Acad Sci U S A ; 112(41): 12812-7, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26417068

ABSTRACT

The contribution of endothelial-derived miR-17∼92 to ischemia-induced arteriogenesis has not been investigated in an in vivo model. In the present study, we demonstrate a critical role for the endothelial-derived miR-17∼92 cluster in shaping physiological and ischemia-triggered arteriogenesis. Endothelial-specific deletion of miR-17∼92 results in an increase in collateral density limbs and hearts and in ischemic limbs compared with control mice, and consequently improves blood flow recovery. Individual cluster components positively or negatively regulate endothelial cell (EC) functions in vitro, and, remarkably, ECs lacking the cluster spontaneously form cords in a manner rescued by miR-17a, -18a, and -19a. Using both in vitro and in vivo analyses, we identified FZD4 and LRP6 as targets of miR-19a/b. Both of these targets were up-regulated in 17∼92 KO ECs compared with control ECs, and both were shown to be targeted by miR-19 using luciferase assays. We demonstrate that miR-19a negatively regulates FZD4, its coreceptor LRP6, and WNT signaling, and that antagonism of miR-19a/b in aged mice improves blood flow recovery after ischemia and reduces repression of these targets. Collectively, these data provide insights into miRNA regulation of arterialization and highlight the importance of vascular WNT signaling in maintaining arterial blood flow.


Subject(s)
Frizzled Receptors/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , MicroRNAs/metabolism , Multigene Family/physiology , Neovascularization, Physiologic/physiology , Wnt Signaling Pathway/physiology , Animals , Frizzled Receptors/genetics , Ischemia/genetics , Ischemia/metabolism , Ischemia/pathology , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Mice , Mice, Knockout , MicroRNAs/genetics
6.
Nature ; 479(7371): 122-6, 2011 Oct 09.
Article in English | MEDLINE | ID: mdl-21983962

ABSTRACT

PHD2 serves as an oxygen sensor that rescues blood supply by regulating vessel formation and shape in case of oxygen shortage. However, it is unknown whether PHD2 can influence arteriogenesis. Here we studied the role of PHD2 in collateral artery growth by using hindlimb ischaemia as a model, a process that compensates for the lack of blood flow in case of major arterial occlusion. We show that Phd2 (also known as Egln1) haplodeficient (Phd2(+/-)) mice displayed preformed collateral arteries that preserved limb perfusion and prevented tissue necrosis in ischaemia. Improved arteriogenesis in Phd2(+/-) mice was due to an expansion of tissue-resident, M2-like macrophages and their increased release of arteriogenic factors, leading to enhanced smooth muscle cell (SMC) recruitment and growth. Both chronic and acute deletion of one Phd2 allele in macrophages was sufficient to skew their polarization towards a pro-arteriogenic phenotype. Mechanistically, collateral vessel preconditioning relied on the activation of canonical NF-κB pathway in Phd2(+/-) macrophages. These results unravel how PHD2 regulates arteriogenesis and artery homeostasis by controlling a specific differentiation state in macrophages and suggest new treatment options for ischaemic disorders.


Subject(s)
Arteries/growth & development , Extremities/blood supply , Ischemia/prevention & control , Macrophages/metabolism , Procollagen-Proline Dioxygenase/deficiency , Procollagen-Proline Dioxygenase/metabolism , Alleles , Animals , Disease Models, Animal , Extremities/pathology , Female , Heterozygote , Homeostasis , Hypoxia-Inducible Factor-Proline Dioxygenases , Ischemia/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Myocytes, Smooth Muscle/cytology , NF-kappa B/metabolism , Necrosis , Phenotype , Procollagen-Proline Dioxygenase/genetics
7.
Proc Natl Acad Sci U S A ; 111(51): 18309-14, 2014 Dec 23.
Article in English | MEDLINE | ID: mdl-25422430

ABSTRACT

Posttranscriptional RNA regulation is important in determining the plasticity of cellular phenotypes. However, mechanisms of how RNA binding proteins (RBPs) influence cellular behavior are poorly understood. We show here that the RBP embryonic lethal abnormal vision like 1 (ELAVL1, also know as HuR) regulates the alternative splicing of eukaryotic translation initiation factor 4E nuclear import factor 1 (Eif4enif1), which encodes an eukaryotic translation initiation factor 4E transporter (4E-T) protein and suppresses the expression of capped mRNAs. In the absence of ELAVL1, skipping of exon 11 of Eif4enif1 forms the stable, short isoform, 4E-Ts. This alternative splicing event results in the formation of RNA processing bodies (PBs), enhanced turnover of angiogenic mRNAs, and suppressed sprouting behavior of vascular endothelial cells. Further, endothelial-specific Elavl1 knockout mice exhibited reduced revascularization after hind limb ischemia and tumor angiogenesis in oncogene-induced mammary cancer, resulting in attenuated blood flow and tumor growth, respectively. ELAVL1-regulated alternative splicing of Eif4enif1 leading to enhanced formation of PB and mRNA turnover constitutes a novel posttranscriptional mechanism critical for pathological angiogenesis.


Subject(s)
Alternative Splicing/physiology , ELAV Proteins/physiology , Neovascularization, Physiologic/physiology , Animals , ELAV-Like Protein 1 , Exons , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism
8.
Proc Natl Acad Sci U S A ; 111(15): 5514-9, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24706887

ABSTRACT

Vascular endothelial growth factors (VEGFs) signal via their cognate receptor tyrosine kinases designated VEGFR1-3. We report that the docking protein fibroblast growth factor receptor substrate 2 (FRS2α) plays a critical role in cell signaling via these receptors. In vitro FRS2α regulates VEGF-A and VEGF-C-dependent activation of extracellular signal-regulated receptor kinase signaling and blood and lymphatic endothelial cells migration and proliferation. In vivo endothelial-specific deletion of FRS2α results in the profound impairment of postnatal vascular development and adult angiogenesis, lymphangiogenesis, and arteriogenesis. We conclude that FRS2α is a previously unidentified component of VEGF receptors signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Membrane Proteins/metabolism , Receptors, Vascular Endothelial Growth Factor/physiology , Signal Transduction/physiology , Animals , Cell Movement/physiology , DNA Primers/genetics , Endothelial Cells , Gene Expression Profiling , Genetic Vectors , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Laser-Doppler Flowmetry , Lentivirus , Mice , Real-Time Polymerase Chain Reaction , Receptors, Vascular Endothelial Growth Factor/metabolism , X-Ray Microtomography
9.
Development ; 140(8): 1720-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23533173

ABSTRACT

Arteriogenesis requires growth of pre-existing arteriolar collateral networks and determines clinical outcome in arterial occlusive diseases. Factors responsible for the development of arteriolar collateral networks are poorly understood. The Notch ligand Delta-like 4 (Dll4) promotes arterial differentiation and restricts vessel branching. We hypothesized that Dll4 may act as a genetic determinant of collateral arterial networks and functional recovery in stroke and hind limb ischemia models in mice. Genetic loss- and gain-of-function approaches in mice showed that Dll4-Notch signaling restricts pial collateral artery formation by modulating arterial branching morphogenesis during embryogenesis. Adult Dll4(+/-) mice showed increased pial collateral numbers, but stroke volume upon middle cerebral artery occlusion was not reduced compared with wild-type littermates. Likewise, Dll4(+/-) mice showed reduced blood flow conductance after femoral artery occlusion, and, despite markedly increased angiogenesis, tissue ischemia was more severe. In peripheral arteries, loss of Dll4 adversely affected excitation-contraction coupling in arterial smooth muscle in response to vasopressor agents and arterial vessel wall adaption in response to increases in blood flow, collectively contributing to reduced flow reserve. We conclude that Dll4-Notch signaling modulates native collateral formation by acting on vascular branching morphogenesis during embryogenesis. Dll4 furthermore affects tissue perfusion by acting on arterial function and structure. Loss of Dll4 stimulates collateral formation and angiogenesis, but in the context of ischemic diseases such beneficial effects are overruled by adverse functional changes, demonstrating that ischemic recovery is not solely determined by collateral number but rather by vessel functionality.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Ischemia/physiopathology , Membrane Proteins/metabolism , Microvessels/embryology , Morphogenesis/physiology , Neovascularization, Physiologic/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Analysis of Variance , Animals , Calcium-Binding Proteins , Immunohistochemistry , Ischemia/metabolism , Mice , Microvessels/physiology , Real-Time Polymerase Chain Reaction , Regional Blood Flow/physiology , X-Ray Microtomography
10.
Microvasc Res ; 106: 57-66, 2016 07.
Article in English | MEDLINE | ID: mdl-27009591

ABSTRACT

Properly regulated angiogenesis and arteriogenesis are essential for effective wound healing. Tissue injury induces robust new vessel formation and subsequent vessel maturation, which involves vessel regression and remodeling. Although formation of functional vasculature is essential for healing, alterations in vascular structure over the time course of skin wound healing are not well understood. Here, using high-resolution ex vivo X-ray micro-computed tomography (microCT), we describe the vascular network during healing of skin excisional wounds with highly detailed three-dimensional (3D) reconstructed images and associated quantitative analysis. We found that relative vessel volume, surface area and branching number are significantly decreased in wounds from day 7 to days 14 and 21. Segmentation and skeletonization analysis of selected branches from high-resolution images as small as 2.5µm voxel size show that branching orders are decreased in the wound vessels during healing. In histological analysis, we found that the contrast agent fills mainly arterioles, but not small capillaries nor large veins. In summary, high-resolution microCT revealed dynamic alterations of vessel structures during wound healing. This technique may be useful as a key tool in the study of the formation and regression of wound vessels.


Subject(s)
Computed Tomography Angiography/methods , Neovascularization, Physiologic , Skin/blood supply , Skin/diagnostic imaging , Wound Healing , Wounds and Injuries/diagnostic imaging , X-Ray Microtomography , Animals , Arterioles/diagnostic imaging , Arterioles/physiopathology , Disease Models, Animal , Imaging, Three-Dimensional , Male , Mice, Inbred C57BL , Predictive Value of Tests , Radiographic Image Interpretation, Computer-Assisted , Time Factors , Wounds and Injuries/physiopathology
11.
Mol Cell ; 32(1): 140-9, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18851840

ABSTRACT

Mammalian target of rapamycin (mTOR) activity is regulated by assembly of two functionally distinct complexes, mTORC1 and mTORC2. In syndecan-4 (S4) null endothelial cells, mTORC2 activity is reduced, resulting in decreased Akt activation, while mTORC1 activity is increased. Levels of rictor, mLST8, and mSin-1 are unchanged in total cell lysates but decreased in the rafts of S4(-/-) endothelial cells, as is the level of PKCalpha. Expression of myristoylated-PKCalpha in S4(-/-) cells restores rictor, mLST8, and mSin-1 presence in the rafts and rescues Akt phosphorylation. PKCalpha knockdown mimics the effect of S4 deletion on mTORC2 localization and Akt activation. Reduced mTORC2 activity in S4(-/-) endothelial cells results in decreased FoxO1/3a and eNOS phosphorylation, decreased endothelial cell size, and increased arterial blood pressure in S4(-/-) mice. Thus, S4-dependent targeting of PKCalpha to the plasma membrane is required for recruitment of mTORC2 components to the rafts and Akt activation.


Subject(s)
Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Syndecan-4/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Biological Transport, Active , Cell Line , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Activation/drug effects , Growth Substances/pharmacology , Membrane Microdomains/metabolism , Mice , Mice, Knockout , Models, Biological , Syndecan-4/deficiency , Syndecan-4/genetics
12.
J Biol Chem ; 289(1): 510-9, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24235146

ABSTRACT

Angiopoietin-2 (Ang2) is an extracellular protein and one of the principal ligands of Tie2 receptor that is involved in the regulation of vascular integrity, quiescence, and inflammation. The mode of secretion of Ang2 has never been established, however. Here, we provide evidence that Ang2 is secreted from endothelial cells via exosomes and that this process is inhibited by the PI3K/Akt/endothelial nitric oxide synthase (eNOS) signaling pathway, whereas it is positively regulated by the syndecan-4/syntenin pathway. Vascular defects in Akt1 null mice arise, in part, because of excessive Ang2 secretion and can be rescued by the syndecan-4 knock-out that reduces extracellular Ang2 levels. This novel mechanism connects three critical signaling pathways: angiopoietin/Tie2, PI3K/Akt/eNOS, and syndecan/syntenin, which play important roles in vascular growth and stabilization.


Subject(s)
Angiopoietin-2/metabolism , Endothelial Cells/metabolism , Exosomes/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Syndecan-4/metabolism , Syntenins/metabolism , Angiopoietin-2/genetics , Animals , Cells, Cultured , Endothelial Cells/cytology , Exosomes/genetics , Mice , Mice, Knockout , Neovascularization, Physiologic/physiology , Nitric Oxide Synthase Type III/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Syndecan-4/genetics , Syntenins/genetics
13.
Circulation ; 130(11): 902-9, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-24982127

ABSTRACT

BACKGROUND: Regulation of vascular endothelial growth factor receptor-2 (VEGFR2) signaling is a control point that determines the extent of vascular tree formation. Recent studies demonstrated an important role played by VEGFR2 endothelial trafficking in control of its activity and suggested the involvement of a phosphotyrosine phosphatase 1b (PTP1b) in this process. This study was designed to define the role of PTP1b in endothelial VEGFR2 signaling and its role in regulation of angiogenesis and arteriogenesis. METHODS AND RESULTS: We generated mice carrying an endothelial-specific deletion of PTP1b and examined the effect of this knockout on VEGF signaling, angiogenesis, and arteriogenesis in vitro and in vivo. PTP1b knockout endothelial cells had increased VEGF-dependent activation of extracellular signal-regulated kinase signaling, sprouting, migration, and proliferation compared with controls. Endothelial PTP1b null mice had increased retinal and Matrigel implant angiogenesis and accelerated wound healing, pointing to enhanced angiogenesis. Increased arteriogenesis was demonstrated by observations of faster recovery of arterial blood flow and large numbers of newly formed arterioles in the hindlimb ischemia mouse model. PTP1b endothelial knockout also rescued impaired blood flow recovery after common femoral artery ligation in synectin null mice. CONCLUSIONS: PTP1b is a key regulator of endothelial VEGFR2 signaling and plays an important role in regulation of the extent of vascular tree formation.


Subject(s)
Endothelial Cells/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Aorta/cytology , Cell Movement/physiology , Cell Proliferation , Disease Models, Animal , Endothelial Cells/cytology , Female , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Ischemia/metabolism , Ischemia/physiopathology , Male , Mice , Mice, Mutant Strains , Neovascularization, Physiologic/physiology , Primary Cell Culture , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , RNA, Small Interfering/genetics , Vascular Endothelial Growth Factor A/metabolism
14.
Blood ; 121(19): 3988-96, S1-9, 2013 May 09.
Article in English | MEDLINE | ID: mdl-23529931

ABSTRACT

Arterial morphogenesis is one of the most critical events during embryonic vascular development. Although arterial fate specification is mainly controlled by the Notch signaling pathway, arterial-venous patterning is modulated by a number of guidance factors. How these pathways are regulated is still largely unknown. Here, we demonstrate that endothelial activation of RAF1/extracellular signal-regulated kinase (ERK) pathway regulates arterial morphogenesis and arterial-venous patterning via Δ/Notch and semaphorin signaling. Introduction of a single amino acid RAF1 mutant (RAF1 Ser259Ala), which renders it resistant to inhibition by phosphorylation, into endothelial cells in vitro induced expression of virtually the entire embryonic arteriogenic program and activated semaphorin 6A-dependent endothelial cell-cell repulsion. In vivo, endothelial-specific expression of RAF1(S259A) during development induced extensive arterial morphogenesis both in the yolk sac and the embryo proper and disrupted arterial-venous patterning. Our results suggest that endothelial ERK signaling is critical for both arteriogenesis and arterial-venous patterning and that RAF1 Ser(259) phosphorylation plays a critical role in preventing unopposed ERK activation.


Subject(s)
Arteries/embryology , Endothelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Morphogenesis , Proto-Oncogene Proteins c-raf/metabolism , Animals , Arteries/metabolism , Cells, Cultured , Embryo, Mammalian , Enzyme Activation/genetics , Enzyme Activation/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Female , Gene Expression Regulation, Developmental , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Morphogenesis/genetics , Morphogenesis/physiology , Pregnancy , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/physiology , Semaphorins/genetics
15.
Circ Res ; 113(9): 1076-86, 2013 Oct 12.
Article in English | MEDLINE | ID: mdl-23897694

ABSTRACT

RATIONALE: Arteriogenesis is the process of formation of arterial conduits. Its promotion is an attractive therapeutic strategy in occlusive atherosclerotic diseases. Despite the functional and clinical importance of arteriogenesis, the biology of the process is poorly understood. Synectin, a gene previously implicated in the regulation of vascular endothelial cell growth factor signaling, offers a unique opportunity to determine relative contributions of various cell types to arteriogenesis. OBJECTIVE: We investigated the cell-autonomous effects of a synectin knockout in arterial morphogenesis. METHODS AND RESULTS: A floxed synectin knockin mouse line was crossbred with endothelial-specific (Tie2, Cdh5, Pdgfb) and smooth muscle myosin heavy chain-specific Cre driver mouse lines to produce cell type-specific deletions. Ablation of synectin expression in endothelial, but not smooth muscle cells resulted in the presence of developmental arterial morphogenetic defects (smaller size of the arterial tree, reduced number of arterial branches and collaterals) and impaired arteriogenesis in adult mice. CONCLUSIONS: Synectin modulates developmental and adult arteriogenesis in an endothelial cell-autonomous fashion. These findings show for the first time that endothelial cells are central to both developmental and adult arteriogenesis and provide a model for future studies of factors involved in this process.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Arteries/metabolism , Endothelial Cells/metabolism , Neovascularization, Physiologic , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , Genotype , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phenotype , Time Factors
16.
J Craniofac Surg ; 25(3): 766-71, 2014 May.
Article in English | MEDLINE | ID: mdl-24820707

ABSTRACT

Transport distraction osteogenesis (DO) can be used to autologously reconstitute calvarial defects. The purpose of this study is to histomorphologically interrogate osteogenic formation during cranial transport distraction using a novel device. We also evaluate the effect of fat grafting on the regenerate and soft-tissue stability during distraction. This study was approved by Yale IACUC. Ten male New Zealand white rabbits (3 mo; 3.5 kg) were used (8 treatment, 2 control). A 16 × 16 mm defect was created abutted by a 10 × 16 mm transport disc. The device was fixated anterioposteriorly. Four animals were fat-grafted using 2 mL of subdermal intrascapular fat deposited along the distraction site. Latency (1 d), active distraction (12-14 d) (1.5 mm/d), and consolidation (4 wk) followed. Calcein and xylene orange fluorochromes were injected subcutaneously during and post-distraction to mark sites of bone formation. Following sacrifice, osteogenesis was assessed using microCT, histology, and fluorescence. Treatment animals demonstrated regenerate bone between distracted segments on microCT. MicroCT analysis of non-fat-grafted and fat-grafted animals revealed a mean density of 2271.95 mgHA/ccm and 2254.27 mgHA/ccm (P = 0.967), respectively, and defect bone versus total volume (BV/TV) of 0.0999 and 0.0766 (P = 0.5979), respectively. Controls had minimal reossification. Histologically, mean densities measured 43.63% and 8.19%, respectively. Fluorescence revealed ossification from the callus as well as from dura and periosteum in the cranial defect. Transport distraction is effective to reconstruct critically sized rabbit calvarial defects. Regenerate bone arises predominantly from the callus with contribution from surrounding dura and periosteum. Adipose grafting is well tolerated but does not enhance osseous regeneration.


Subject(s)
Adipose Tissue/transplantation , Osteogenesis, Distraction/methods , Plastic Surgery Procedures/methods , Skull/surgery , Animals , Bone Density/physiology , Bone Diseases/surgery , Bone Regeneration/physiology , Bony Callus/pathology , Dura Mater/pathology , Fluoresceins , Fluorescent Dyes , Image Processing, Computer-Assisted/methods , Male , Osteogenesis/physiology , Osteogenesis, Distraction/instrumentation , Periosteum/pathology , Rabbits , Random Allocation , Plastic Surgery Procedures/instrumentation , X-Ray Microtomography/methods
17.
Circulation ; 126(22): 2589-600, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-23091063

ABSTRACT

BACKGROUND: Arteriogenesis and collateral formation are complex processes requiring integration of multiple inputs to coordinate vessel branching, growth, maturation, and network size. Factors regulating these processes have not been determined. METHODS AND RESULTS: We used an inhibitor of NFκB activation (IκBαSR) under control of an endothelial-specific inducible promoter to selectively suppress endothelial nuclear factor-κB activation during development, in the adult vasculature, or in vitro. Inhibition of nuclear factor-κB activation resulted in formation of an excessively branched arterial network that was composed of immature vessels and provided poor distal tissue perfusion. Molecular analysis demonstrated reduced adhesion molecule expression leading to decreased monocyte influx, reduced hypoxia-inducible factor-1α levels, and a marked decrease in δ-like ligand 4 expression with a consequent decrease in Notch signaling. The latter was the principal cause of increased vascular branching as treatment with Jagged-1 peptide reduced the size of the arterial network to baseline levels. CONCLUSIONS: These findings identify nuclear factor-κB as a key regulator of adult and developmental arteriogenesis and collateral formation. Nuclear factor-κB achieves this by regulating hypoxia-inducible factor-1α-dependent expression of vascular endothelial growth factor-A and platelet-derived growth factor-BB, which are necessary for the development and maturation of the arterial collateral network, and by regulating δ-like ligand 4 expression, which in turn determines the size and complexity of the network.


Subject(s)
Endothelial Cells/metabolism , Ischemia/physiopathology , NF-kappa B p50 Subunit/metabolism , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Animals , Animals, Newborn , Becaplermin , Brain/metabolism , Disease Models, Animal , Hindlimb/blood supply , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/metabolism , Mice , Mice, Transgenic , NF-kappa B p50 Subunit/genetics , Neovascularization, Pathologic/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Retina/metabolism , Vascular Endothelial Growth Factor A/metabolism
18.
Angiogenesis ; 15(3): 469-80, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22562363

ABSTRACT

Vascular endothelial growth factor (VEGF)-A regulates angiogenesis, vascular morphology and permeability by signaling through its receptor VEGFR-2. The Shb adapter protein has previously been found to relay certain VEGFR-2 dependent signals and consequently vascular physiology and structure was assessed in Shb knockout mice. X-ray computed tomography of vessels larger than 24 µm diameter (micro-CT) after contrast injection revealed an increased frequency of 48-96 µm arterioles in the hindlimb calf muscle in Shb knockout mice. Intravital microscopy of the cremaster muscle demonstrated a less regular vasculature with fewer branch points and increased vessel tortuosity, changes that led to an increased blood flow velocity. Reduced in vivo angiogenesis was observed in Shb knockout Matrigel™ plugs. Unlike the wild-type situation, VEGF-A did not provoke a dissociation of VE-cadherin from adherens junctions in Shb knockout venules. The reduced angiogenesis and altered properties of junctions had consequences for two patho-physiological responses to arterial occlusion: vascular permeability was reduced in the Shb knockout cremaster muscle after ligation of one supplying artery and heat-induced blood flow determined by Laser-Doppler measurements was decreased in the hindlimb after ligation of the femoral artery. Consequently, the Shb knockout mouse exhibited structural and functional (angiogenesis and vascular permeability) vascular abnormalities that have implications for understanding the function of VEGF-A under physiological conditions.


Subject(s)
Adaptation, Physiological , Endothelium, Vascular/physiology , Proto-Oncogene Proteins/physiology , Animals , Capillary Permeability , Hindlimb/blood supply , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Scanning , Proto-Oncogene Proteins/genetics , Tomography, X-Ray Computed , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
Arterioscler Thromb Vasc Biol ; 31(5): 1059-65, 2011 May.
Article in English | MEDLINE | ID: mdl-21330605

ABSTRACT

OBJECTIVE: Notch has been implicated in neointima formation as reflected by increased Notch/Jagged expression on vascular injury and the promigratory effect of Notch signaling on smooth muscle cells. Soluble Jagged-1 (sJag1) has been shown to inhibit Notch signaling in vitro; however, its capacity to suppress neointima formation remains unknown. METHODS AND RESULTS: Balloon injury of rat carotid arteries induced Notch1, Notch3, and Jagged-1 expression at days 3 and 14 postinjury. Notch signaling was activated as shown by increased expression of the Notch target gene Herp2. Adenoviral sJag1 (Ad-sJag1) transfection reduced neointima formation in carotid artery and enhanced reendothelialization, whereas adenoviral full-length Jagged-1 (Ad-Fl-Jag1) or LacZ had no effect. Injury-induced Herp2 expression was absent in vessels treated with Ad-sJag1. Consistently, Herp2 expression was reduced in Ad-sJag1-infected or recombinant sJag1 -treated coronary artery smooth muscle cells (CASMCs). Ad-sJag1 had no effect on human umbilical endothelial cell behavior, but it significantly reduced proliferation and migration of CASMCs. Overexpression of Herp2 in sJag1-treated CASMCs rescued the migratory and proliferative capacity in vitro. CONCLUSIONS: Our results demonstrate that sJag1 can inhibit neointima formation after balloon injury by decreasing smooth muscle cell proliferation and migration through interference with Notch-Herp2 signaling.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Calcium-Binding Proteins/metabolism , Carotid Arteries/metabolism , Carotid Artery Injuries/prevention & control , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptor, Notch1/metabolism , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Signal Transduction , Tunica Intima/metabolism , Analysis of Variance , Animals , Calcium-Binding Proteins/genetics , Carotid Arteries/pathology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Cell Movement , Cell Proliferation , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Hyperplasia , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Membrane Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Rats , Rats, Sprague-Dawley , Receptor, Notch3 , Serrate-Jagged Proteins , Time Factors , Transfection , Tunica Intima/pathology
20.
Am J Cancer Res ; 12(8): 3679-3692, 2022.
Article in English | MEDLINE | ID: mdl-36119846

ABSTRACT

Epigenomic-wide DNA methylation profiling holds the potential to reflect both electronic cigarette exposure-associated risks and individual poor health outcomes. However, a systemic study in animals or humans is still lacking. Using the Infinium Mouse Methylation BeadChip, we examined the DNA methylation status of white blood cells in male ApoE-/- mice after 14 weeks of electronic cigarette exposure with the InExpose system (2 hr/day, 5 days/week, 50% PG and 50% VG) with low (6 mg/ml) and high (36 mg/ml) nicotine concentrations. Our results indicate that electronic cigarette aerosol inhalation induces significant alteration of 8,985 CpGs in a dose-dependent manner (FDR<0.05); 7,389 (82.2%) of the CpG sites are annotated with known genes. Among the top 6 significant CpG sites (P-value<1e-8), 4 CpG sites are located in the known genes, and most (3/5) of these genes have been related to cigarette smoking. The other two CpGs are close to/associated with the Phc2 gene that was recently linked to smoking in a transcriptome-wide associations study. Furthermore, the gene set enrichment analysis highlights the activation of MAPK and 4 cardiomyocyte/cardiomyopathy-related signaling pathways (including adrenergic signaling in cardiomyocytes and arrhythmogenic right ventricular cardiomyopathy) following repeated electronic cigarette use. The MAPK pathway activation correlates well with our finding of increased cytokine mRNA expression after electronic cigarette exposure in the same batch of mice. Interestingly, two pathways related to mitochondrial activities, namely mitochondrial gene expression and mitochondrial translation, are also activated after electronic cigarette exposure. Elucidating the relationship between these pathways and the increased circulating mitochondrial DNA observed here will provide further insight into the cell-damaging effects of prolonged inhalation of e-cigarette aerosols.

SELECTION OF CITATIONS
SEARCH DETAIL