Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Sci Rep ; 9(1): 7286, 2019 05 13.
Article in English | MEDLINE | ID: mdl-31086203

ABSTRACT

The cellular origins of vasa vasorum are ill-defined and may involve circulating or local progenitor cells. We previously discovered that murine aortic adventitia contains Sca-1+CD45+ progenitors that produce macrophages. Here we investigated whether they are also vasculogenic. In aortas of C57BL/6 mice, Sca-1+CD45+ cells were localised to adventitia and lacked surface expression of endothelial markers (<1% for CD31, CD144, TIE-2). In contrast, they did show expression of CD31, CD144, TIE-2 and VEGFR2 in atherosclerotic ApoE-/- aortas. Although Sca-1+CD45+ cells from C57BL/6 aorta did not express CD31, they formed CD31+ colonies in endothelial differentiation media and produced interconnecting vascular-like cords in Matrigel that contained both endothelial cells and a small population of macrophages, which were located at branch points. Transfer of aortic Sca-1+CD45+ cells generated endothelial cells and neovessels de novo in a hindlimb model of ischaemia and resulted in a 50% increase in perfusion compared to cell-free control. Similarly, their injection into the carotid adventitia of ApoE-/- mice produced donor-derived adventitial and peri-adventitial microvessels after atherogenic diet, suggestive of newly formed vasa vasorum. These findings show that beyond its content of macrophage progenitors, adventitial Sca-1+CD45+ cells are also vasculogenic and may be a source of vasa vasorum during atherogenesis.


Subject(s)
Atherosclerosis/pathology , Cell Differentiation , Neovascularization, Pathologic/pathology , Stem Cells/physiology , Vasa Vasorum/pathology , Adventitia/cytology , Adventitia/pathology , Animals , Antigens, Ly/metabolism , Aorta/cytology , Aorta/pathology , Atherosclerosis/etiology , Diet, Atherogenic , Disease Models, Animal , Endothelial Cells/physiology , Female , Humans , Leukocyte Common Antigens/metabolism , Macrophages/physiology , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout, ApoE , Neovascularization, Pathologic/etiology , Vasa Vasorum/cytology
2.
PLoS One ; 13(9): e0204045, 2018.
Article in English | MEDLINE | ID: mdl-30265729

ABSTRACT

Changes in extracellular matrix proteins may contribute significantly to the adaptation of vein grafts to the arterial circulation. We examined the production and distribution of versican and hyaluronan in intact human vein rings cultured ex vivo, veins perfused ex vivo, and cultured venous adventitial and smooth muscle cells. Immunohistochemistry revealed higher levels of versican in the intima/media compared to the adventitia, and no differences in hyaluronan. In the vasa vasorum, versican and hyaluronan associated with CD34+ progenitor cells. Culturing the vein rings for 14 days revealed increased versican immunostaining of 30-40% in all layers, with no changes in hyaluronan. Changes in versican accumulation appear to result from increased synthesis in the intima/media and decreased degradation in the adventitia as versican transcripts were increased in the intima/media, but unchanged in the adventitia, and versikine (the ADAMTS-mediated cleavage product of versican) was increased in the intima/media, but decreased in the adventitia. In perfused human veins, versican was specifically increased in the intima/media in the presence of venous pressure, but not with arterial pressure. Unexpectedly, cultured adventitial cells express and accumulate more versican and hyaluronan than smooth muscle cells. These data demonstrate a differential regulation of versican and hyaluronan in human venous adventitia vs. intima/media and suggest distinct functions for these extracellular matrix macromolecules in these venous wall compartments during the adaptive response of vein grafts to the arterial circulation.


Subject(s)
Veins/metabolism , Veins/transplantation , Versicans/metabolism , Adventitia/metabolism , Antigens, CD34/metabolism , Arterial Pressure/physiology , Cells, Cultured , Humans , Hyaluronic Acid/metabolism , Immunohistochemistry , Myocytes, Smooth Muscle/metabolism , Saphenous Vein/cytology , Saphenous Vein/metabolism , Stem Cells/metabolism , Tissue Culture Techniques , Tunica Intima/cytology , Tunica Intima/metabolism , Tunica Media/cytology , Tunica Media/metabolism , Vasa Vasorum/cytology , Vasa Vasorum/metabolism , Veins/cytology , Versicans/genetics
3.
J Theor Biol ; 450: 53-65, 2018 08 07.
Article in English | MEDLINE | ID: mdl-29704490

ABSTRACT

Observational studies have identified angiogenesis from the adventitial vasa vasorum and intraplaque hemorrhage (IPH) as critical factors in atherosclerotic plaque progression and destabilization. Here we propose a mathematical model incorporating intraplaque neovascularization and hemodynamic calculation with plaque destabilization for the quantitative evaluation of the role of neoangiogenesis and IPH in the vulnerable atherosclerotic plaque formation. An angiogenic microvasculature is generated by two-dimensional nine-point discretization of endothelial cell proliferation and migration from the vasa vasorum. Three key cells (endothelial cells, smooth muscle cells and macrophages) and three key chemicals (vascular endothelial growth factors, extracellular matrix and matrix metalloproteinase) are involved in the plaque progression model, and described by the reaction-diffusion partial differential equations. The hemodynamic calculation of the microcirculation on the generated microvessel network is carried out by coupling the intravascular, interstitial and transvascular flow. The plasma concentration in the interstitial domain is defined as the description of IPH area according to the diffusion and convection with the interstitial fluid flow, as well as the extravascular movement across the leaky vessel wall. The simulation results demonstrate a series of pathophysiological phenomena during the vulnerable progression of an atherosclerotic plaque, including the expanding necrotic core, the exacerbated inflammation, the high microvessel density (MVD) region at the shoulder areas, the transvascular flow through the capillary wall and the IPH. The important role of IPH in the plaque destabilization is evidenced by simulations with varied model parameters. It is found that the IPH can significantly speed up the plaque vulnerability by increasing necrotic core and thinning fibrous cap. In addition, the decreased MVD and vessel permeability may slow down the process of plaque destabilization by reducing the IPH dramatically. We envision that the present model and its future advances can serve as a valuable theoretical platform for studying the dynamic changes in the microenvironment during the plaque destabilization.


Subject(s)
Hemorrhage , Models, Theoretical , Neovascularization, Pathologic , Plaque, Atherosclerotic/pathology , Cell Movement , Cell Proliferation , Disease Progression , Endothelial Cells/cytology , Humans , Vasa Vasorum/cytology
4.
Stem Cell Reports ; 9(1): 292-303, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28552602

ABSTRACT

In the microcirculation, pericytes are believed to function as mesenchymal stromal cells (MSCs). We hypothesized that the vasa vasorum harbor progenitor cells within the adventitia of human aorta. Pericytes, endothelial progenitor cells, and other cell subpopulations were detected among freshly isolated adventitial cells using flow cytometry. Purified cultured pericytes were enriched for the MSC markers CD105 and CD73 and depleted of the endothelial markers von Willebrand factor and CD31. Cultured pericytes were capable of smooth muscle lineage progression including inducible expression of smooth muscle myosin heavy chain, calponin, and α-smooth muscle actin, and adopted a spindle shape. Pericytes formed spheroids when cultured on Matrigel substrates and peripherally localized with branching endothelial cells in vitro. Our results indicate that the vasa vasorum form a progenitor cell niche distinct from other previously described progenitor populations in human adventitia. These findings could have important implications for understanding the complex pathophysiology of human aortic disease.


Subject(s)
Aorta/cytology , Endothelial Progenitor Cells/cytology , Pericytes/cytology , Vasa Vasorum/cytology , 5'-Nucleotidase/analysis , Adult , Adventitia/cytology , Aged , Cells, Cultured , Endoglin/analysis , Female , Humans , Male , Middle Aged , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Stem Cell Niche , von Willebrand Factor/analysis
5.
Am J Physiol Cell Physiol ; 312(1): C56-C70, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27856430

ABSTRACT

Angiogenesis is an energy-demanding process; however, the role of cellular energy pathways and their regulation by extracellular stimuli, especially extracellular nucleotides, remain largely unexplored. Using metabolic inhibitors of glycolysis (2-deoxyglucose) and oxidative phosphorylation (OXPHOS) (oligomycin, rotenone, and FCCP), we demonstrate that glycolysis and OXPHOS are both essential for angiogenic responses of vasa vasorum endothelial cell (VVEC). Treatment with P2R agonists, ATP, and 2-methylthioadenosine diphosphate trisodium salt (MeSADP), but not P1 receptor agonist, adenosine, increased glycolytic activity in VVEC (measured by extracellular acidification rate and lactate production). Stimulation of glycolysis was accompanied by increased levels of phospho-phosphofructokinase B3, hexokinase (HK), and GLUT-1, but not lactate dehydrogenase. Moreover, extracellular ATP and MeSADP, and to a lesser extent adenosine, increased basal and maximal oxygen consumption rates in VVEC. These effects were potentiated when the cells were cultured in 20 mM galactose and 5 mM glucose compared with 25 mM glucose. Treatment with P2R agonists decreased phosphorylation of pyruvate dehydrogenase (PDH)-E1α and increased succinate dehydrogenase (SDH), cytochrome oxidase IV, and ß-subunit of F1F0 ATP synthase expression. In addition, P2R stimulation transiently elevated mitochondrial Ca2+ concentration, implying involvement of mitochondria in VVEC angiogenic activation. We also demonstrated a critical role of phosphatidylinositol 3-kinase and Akt pathways in lactate production, PDH-E1α phosphorylation, and the expression of HK, SDH, and GLUT-1 in ATP-stimulated VVEC. Together, our findings suggest that purinergic and metabolic regulation of VVEC energy pathways is essential for VV angiogenesis and may contribute to pathologic vascular remodeling in pulmonary hypertension.


Subject(s)
Endothelial Cells/physiology , Glycolysis/physiology , Neovascularization, Physiologic/physiology , Oxidative Phosphorylation , Vasa Vasorum/cytology , Vasa Vasorum/physiology , Animals , Cattle , Cells, Cultured , Endothelial Cells/cytology , Male , Receptors, Purinergic
6.
Ann Anat ; 205: 22-36, 2016 May.
Article in English | MEDLINE | ID: mdl-26844625

ABSTRACT

Vasa vasorum supply both the tunica adventitia and the tunica media of major arteries with nutrients and oxygen. We estimated the density of von Willebrand factor-positive profiles of vasa vasorum visible in transversal histological sections of 123 tissue samples collected from five anatomical positions in the porcine aortae of growing pigs (n=25). The animals ranged in age from 0 to 230 days. The tunica media of the thoracic aorta had a greater vasa vasorum density, with microvessels penetrating deeper towards the lumen than in the abdominal aorta. The density of vasa vasorum gradually decreased with age in both the media and the adventitia. The relative depth into which the vasa vasorum penetrated and where they branched remained constant during the ageing and growth of the media. The ratio of the tunica media and tunica adventitia thicknesses did not change in the single aortic segments during ageing. The media of older animals received fewer but equally distributed vasa vasorum. A greater density of vasa vasorum in the media was correlated with greater media thickness and a greater elastin fraction (data on elastin taken from another study on the same samples). Immunohistochemical quantification revealed deeper penetration of vasa vasorum towards the adluminal layers of the tunica media that were hitherto reported to be avascular. The complete primary morphometric data, in the form of continuous variables, have been made available as a supplement. Mapping of the vasa vasorum profile density and position has promising illustrative potential for studies on atherosclerotic and inflammatory neovascularization, aortic aneurysms, and drug distribution from arterial stents in experimental porcine models.


Subject(s)
Adventitia/cytology , Aging/pathology , Aorta/cytology , Tunica Media/cytology , Vasa Vasorum/cytology , Adventitia/chemistry , Animals , Animals, Newborn , Aorta/chemistry , Female , Male , Swine , Tissue Distribution , Tunica Media/chemistry , Vasa Vasorum/chemistry , von Willebrand Factor/chemistry
7.
Lab Invest ; 94(12): 1340-54, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25329003

ABSTRACT

Adventitial microvessels, vasa vasorum in the vessel walls, have an active role in the vascular remodeling, although its mechanisms are still unclear. It has been reported that microvascular pericytes (PCs) possess mesenchymal plasticity. Therefore, microvessels would serve as a systemic reservoir of stem cells and contribute to the tissues remodeling. However, most aspects of the biology of multipotent PCs (mPCs), in particular of pathological microvessels are still obscure because of the lack of appropriate methods to detect and isolate these cells. In order to examine the characteristics of mPCs, we established immortalized cells residing in adventitial capillary growing at the injured vascular walls. We recently developed in vivo angiogenesis to observe adventitial microvessels using collagen-coated tube (CCT), which also can be used as an adventitial microvessel-rich tissue. By using the CCT, CD146- or NG2-positive cells were isolated from the adventitial microvessels in the injured arteries of mice harboring a temperature-sensitive SV40 T-antigen gene. Several capillary-derived endothelial cells (cECs) and PCs (cPCs) cell lines were established. cECs and cPCs maintain a number of key endothelial and PC features. Co-incubation of cPCs with cECs formed capillary-like structure in Matrigel. Three out of six cPC lines, termed capillary mPCs demonstrated both mesenchymal stem cell- and neuronal stem cell-like phenotypes, differentiating effectively into adipocytes, osteoblasts, as well as schwann cells. mPCs differentiated to ECs and PCs, and formed capillary-like structure on their own. Transplanted DsRed-expressing mPCs were resident in the capillary and muscle fibers and promoted angiogenesis and myogenesis in damaged skeletal muscle. Adventitial mPCs possess transdifferentiation potential with unique phenotypes, including the reconstitution of capillary-like structures. Their phenotype would contribute to the pathological angiogenesis associated with vascular remodeling. These cell lines also provide a reproducible cellular tool for high-throughput studies on angiogenesis, vascular remodeling, and regeneration as well.


Subject(s)
Capillaries/pathology , Pericytes/physiology , Regeneration/physiology , Vasa Vasorum/cytology , Vascular Remodeling , Animals , Antigens , Cell Differentiation , Cell Separation , Endothelial Cells/physiology , Mice , Mice, SCID , Neovascularization, Physiologic , Proteoglycans , Stem Cells/physiology , Transcriptome
9.
Tissue Eng Part A ; 19(23-24): 2724-34, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23879654

ABSTRACT

Multipotent stem/progenitor cells localize perivascularly in many organs and vessel walls. These tissue-resident stem/progenitor cells differentiate into vascular endothelial cells, pericytes, and other mesenchymal lineages, and participate in physiological maintenance and repair of vasculatures. In this study, we characterized stromal vascular cells obtained through the explant culture method from three different vessel walls in humans: arterial wall (ART; >500 µm in diameter), venous wall (VN; >500 µm in diameter), and small vessels in adipose tissue (SV; arterioles and venules, <100 µm in diameter). These were examined for functionality and compared with adipose-derived stem/stromal cells (ASCs). All stromal vascular cells of different origins presented fibroblast-like morphology and we could not visually discriminate one population from another. Flow cytometry showed that the cultured population heterogeneously expressed a variety of surface antigens associated with stem/progenitor cells, but CD105 was expressed by most cells in all groups, suggesting that the cells generally shared the characteristics of mesenchymal stem cells. Our histological and flow cytometric data suggested that the main population of vessel wall-derived stromal vascular cells were CD34(+)/CD31(-) and came from the tunica adventitia and areola tissue surrounding the adventitia. CD271 (p75NTR) was expressed by the vasa vasorum in the VN adventitia and by a limited population in the adventitia of SV. All three populations differentiated into multiple lineages as did ASCs. ART cells induced the largest quantity of calcium formation in the osteogenic medium, whereas ASCs showed the greatest adipogenic differentiation. SV and VN stromal cells had greater potency for network formation than did ART stromal cells. In conclusion, the three stromal vascular populations exhibited differential functional properties. Our results have clinical implications for vascular diseases such as arterial wall calcification and possible applications to regenerative therapies involving each vessel wall-resident stromal population.


Subject(s)
Adipose Tissue/cytology , Arteries/cytology , Veins/cytology , Adipose Tissue/metabolism , Adventitia/cytology , Adventitia/metabolism , Antigens, CD/biosynthesis , Arteries/metabolism , Cells, Cultured , Humans , Male , Organ Specificity/physiology , Stromal Cells/cytology , Stromal Cells/metabolism , Vasa Vasorum/cytology , Vasa Vasorum/metabolism , Vascular Calcification/metabolism , Veins/metabolism
10.
PLoS One ; 8(4): e59733, 2013.
Article in English | MEDLINE | ID: mdl-23613714

ABSTRACT

BACKGROUND: In a neonatal model of hypoxic pulmonary hypertension, a dramatic pulmonary artery adventitial thickening, accumulation of inflammatory cells in the adventitial compartment, and angiogenic expansion of the vasa vasorum microcirculatory network are observed. These pathophysiological responses suggest that rapidly proliferating vasa vasorum endothelial cells (VVEC) may exhibit increased permeability for circulating blood cells and macromolecules. However, the molecular mechanisms underlying these observations remain unexplored. Some reports implicated extracellular adenosine in the regulation of vascular permeability under hypoxic and inflammatory conditions. Thus, we aimed to determine the role of adenosine in barrier regulation of VVEC isolated from the pulmonary arteries of normoxic (VVEC-Co) or chronically hypoxic (VVEC-Hyp) neonatal calves. PRINCIPAL FINDINGS: We demonstrate via a transendothelial electrical resistance measurement that exogenous adenosine significantly enhanced the barrier function in VVEC-Co and, to a lesser extent, in VVEC-Hyp. Our data from a quantitative reverse transcription polymerase chain reaction show that both VVEC-Co and VVEC-Hyp express all four adenosine receptors (A1, A2A, A2B, and A3), with the highest expression level of A1 receptors (A1Rs). However, A1R expression was significantly lower in VVEC-Hyp compared to VVEC-Co. By using an A1R-specific agonist/antagonist and siRNA, we demonstrate that A1Rs are mostly responsible for adenosine-induced enhancement in barrier function. Adenosine-induced barrier integrity enhancement was attenuated by pretreatment of VVEC with pertussis toxin and GSK690693 or LY294002, suggesting the involvement of Gi proteins and the PI3K-Akt pathway. Moreover, we reveal a critical role of actin cytoskeleton in VVEC barrier regulation by using specific inhibitors of actin and microtubule polymerization. Further, we show that adenosine pretreatment blocked the tumor necrosis factor alpha (TNF-α)-induced permeability in VVEC-Co, validating its anti-inflammatory effects. CONCLUSIONS: We demonstrate for the first time that stimulation of A1Rs enhances the barrier function in VVEC by activation of the Gi/PI3K/Akt pathway and remodeling of actin microfilament.


Subject(s)
Actin Cytoskeleton/metabolism , Endothelial Cells/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Adenosine A1/metabolism , Vasa Vasorum/cytology , Actin Cytoskeleton/drug effects , Animals , Cattle , Chromones/pharmacology , Endothelial Cells/drug effects , Male , Morpholines/pharmacology , Oxadiazoles/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Vasa Vasorum/drug effects , Vasa Vasorum/metabolism
11.
Annu Rev Physiol ; 75: 23-47, 2013.
Article in English | MEDLINE | ID: mdl-23216413

ABSTRACT

The vascular adventitia acts as a biological processing center for the retrieval, integration, storage, and release of key regulators of vessel wall function. It is the most complex compartment of the vessel wall and is composed of a variety of cells, including fibroblasts, immunomodulatory cells (dendritic cells and macrophages), progenitor cells, vasa vasorum endothelial cells and pericytes, and adrenergic nerves. In response to vascular stress or injury, resident adventitial cells are often the first to be activated and reprogrammed to influence the tone and structure of the vessel wall; to initiate and perpetuate chronic vascular inflammation; and to stimulate expansion of the vasa vasorum, which can act as a conduit for continued inflammatory and progenitor cell delivery to the vessel wall. This review presents the current evidence demonstrating that the adventitia acts as a key regulator of vascular wall function and structure from the outside in.


Subject(s)
Adventitia/physiology , Blood Vessels/cytology , Blood Vessels/physiology , Adventitia/cytology , Animals , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Macrophages/cytology , Macrophages/physiology , Stem Cells/cytology , Stem Cells/physiology , Stress, Physiological/physiology , Vasa Vasorum/cytology , Vasa Vasorum/physiology
12.
Histol Histopathol ; 27(9): 1195-202, 2012 09.
Article in English | MEDLINE | ID: mdl-22806906

ABSTRACT

INTRODUCTION: Vasa vasorum (VV) neovasculogenic potential is now widely accepted, and possibly related to the presence of progenitor cells. We studied the morphology of VV in healthy arteries and their immunohistochemical (IHC) expression of Nestin and WT1, two markers of endothelial progenitor cells. MATERIALS AND METHODS: Twenty arteries from 16 multiorgan donors were analyzed; IHC was performed manually (CD34, CD31, Nestin) or automatically (WT1). Microvessel positivity "density" for each antibody was calculated dividing vascular adventitia in 1-mm2 fields with an ocular micrometer. Double immunofluorescence was used to investigate Nestin and WT1 co-localization in VV. RESULTS: The mean positivity "densities" for CD31, CD34, Nestin and WT1 were 13.63, 12.20, 8.90 and 7.98/mm² respectively. Mean Nestin/CD31 and WT1/CD31 ratios were 0.69 and 0.63 respectively. VV <50 µm in diameter showed a higher percentage of Nestin/WT1-positive cells than larger ones, especially in "hot spots", characterized by several small-sized arteriolar VV, often together with nerva vasorum. Immunofluorescence indentified Nestin and WT1 in the same endothelial cells. WT1 nuclear expression was mainly seen in <50 µm VV. DISCUSSION: We describe Nestin and WT1 in adult VV, especially <50 µm and gathered in "hot spots". The nuclear localization of WT1 could express an increasing transcriptional activity in progenitor-committed Nestin-positive cells. The "hot spot" could therefore represent a valid model for the vasculogenic niche in normal arteries and could potentially represent the main source for neovasculogenesis during atherosclerosis.


Subject(s)
Intermediate Filament Proteins/biosynthesis , Neovascularization, Physiologic/physiology , Nerve Tissue Proteins/biosynthesis , Stem Cells/cytology , Vasa Vasorum/cytology , WT1 Proteins/biosynthesis , Arteries/cytology , Arteries/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Immunohistochemistry , Nestin , Stem Cells/metabolism , Vasa Vasorum/metabolism
13.
Angiogenesis ; 14(4): 503-13, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21922294

ABSTRACT

Vascular remodeling plays a pivotal role in a variety of pathophysiological conditions where hypoxia and inflammation are prominent features. Intravascular ATP, ADP and adenosine are known as important regulators of vascular tone, permeability and homeostasis, however contribution of purinergic signalling to endothelial cell growth and angiogenesis remains poorly understood. By using vasa vasorum endothelial cells (VVEC) isolated from pulmonary artery adventitia of control and chronically hypoxic neonatal calves, these studies were aimed to evaluate the effect of hypoxia on biochemical and functional properties of microvascular endothelial network at the sites of angiogenesis. In comparison with normoxic controls, VVEC from hypoxic animals are characterized by (1) drastically impaired nucleoside triphosphate diphosphohydrolase-1 (NTPDase-1/CD39) and ecto-5'-nucleotidase/CD73 activities with respective increases in basal extracellular ATP and ADP levels (2) higher proliferative responses to low micromolar concentrations of ATP and ADP; and (3) enhanced permeability and disordered adenosinergic control of vascular barrier function (measured as a paracellular flux of 70 kDa fluorescein isothiocyanate-dextran). Together, these results suggest that unique pattern of purine-mediated angiogenic activation and enhanced leakiness of VVEC from chronically hypoxic vessels may be defined by disordered endothelial nucleotide homeostasis at sites of active neovascularization.


Subject(s)
5'-Nucleotidase/metabolism , Antigens, CD/metabolism , Apyrase/metabolism , Endothelial Cells/metabolism , Hypoxia/metabolism , Neovascularization, Pathologic/metabolism , Pulmonary Artery/cytology , Vasa Vasorum/cytology , Adenosine/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Analysis of Variance , Animals , Animals, Newborn , Blotting, Western , Capillary Permeability/physiology , Cattle , Cell Proliferation , Cyclic AMP/metabolism , DNA Primers/genetics , Dextrans , Fluorescein-5-isothiocyanate/analogs & derivatives , Regression Analysis , Reverse Transcriptase Polymerase Chain Reaction
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 30(2): 275-7, 2010 Feb.
Article in Chinese | MEDLINE | ID: mdl-20159699

ABSTRACT

OBJECTIVE: To investigate the effect of hyperlipidemia on vasa vasorum and vascular endothelial growth factor (VEGF) and study the role of vasa vasorum in arteriosclerosis. METHODS: Thirty SD rats were randomized into normal control, hyperlipidemic and simvastatin treatment groups (n=10). In simvastatin group, hyperlipidemia was induced by a 4-week administration of atherogenic diet followed by a 16-week treatment with simvastatin at the daily dose of 10 mg/kg, and the rats in hyperlipidemic rats received no treatment. The changes in the aorta and vasa vasorum were examined, and serum lipid concentration and VEGF and NO levels were measured. RESULTS: Compared with the control group, the hyperlipidemic rats showed significantly thickened intima and media aorta and increased vasa vasorum density with lowered NO level, but VEGF underwent no significant changes. Simvastatin treatment significantly reduced the thickness of the intima and media aorta and increased vasa vasorum density in comparison with those in hyperlipidemic group. Simvastatin treatment also significantly increased VEGF and NO levels and a positive correlation was noted between their levels. CONCLUSION: Hyperlipidemia can impair the vasa vasorum and aortic endothelial function. Simvastatin increases VEGF and NO and promotes neogenesis of the vasa vasorum for the benefit of the aortic function.


Subject(s)
Aorta/cytology , Endothelium, Vascular/physiology , Hyperlipidemias/drug therapy , Simvastatin/pharmacology , Vasa Vasorum/cytology , Animals , Arteriosclerosis/pathology , Arteriosclerosis/physiopathology , Hyperlipidemias/pathology , Hyperlipidemias/physiopathology , Hypolipidemic Agents/pharmacology , Male , Nitric Oxide/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factor A/metabolism
15.
Am J Physiol Lung Cell Mol Physiol ; 297(5): L954-64, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19684203

ABSTRACT

We recently reported that vasa vasorum expansion occurs in the pulmonary artery (PA) adventitia of chronically hypoxic animals and that extracellular ATP is a pro-angiogenic factor for isolated vasa vasorum endothelial cells (VVEC). However, the sources of extracellular ATP in the PA vascular wall, as well as the molecular mechanisms underlying its release, remain elusive. Studies were undertaken to explore whether VVEC release ATP in response to hypoxia and to determine signaling pathways involved in this process. We found that hypoxia (1-3% O2) resulted in time- and O2-dependent ATP release from VVEC. Preincubation with the inhibitors of vesicular transport (monensin, brefeldin A, and N-ethylmaleimide) significantly decreased ATP accumulation in the VVEC conditioned media, suggesting that hypoxia-induced ATP release occurs through vesicular exocytosis. Additionally, both hypoxia and exogenously added ATP resulted in the activation of PI3K and accumulation of GTP-bound RhoA in a time-dependent manner. Pharmacological inhibition of PI3K and ROCK or knockout of RhoA by small interfering RNA significantly abolished hypoxia-induced ATP release from VVEC. Moreover, RhoA and ROCK play a critical role in ATP-induced increases in VVEC DNA synthesis, migration, and tube formation, indicating a functional contribution of PI3K, Rho, and ROCK to both the autocrine mechanism of ATP release and ATP-mediated angiogenic activation of VVEC. Taken together, our findings provide novel evidence for the signaling mechanisms that link hypoxia-induced increases in extracellular ATP and vasa vasorum expansion.


Subject(s)
Adenosine Triphosphate/pharmacology , Endothelial Cells/enzymology , Neovascularization, Physiologic/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Vasa Vasorum/cytology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cattle , Cell Hypoxia/drug effects , Cell Movement/drug effects , Collagen/metabolism , DNA/biosynthesis , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Laminin/metabolism , Male , Proteoglycans/metabolism , Pulmonary Artery/cytology , Signal Transduction/drug effects , Time Factors , Transport Vesicles/drug effects , Transport Vesicles/metabolism
16.
Angiogenesis ; 11(2): 169-82, 2008.
Article in English | MEDLINE | ID: mdl-18071915

ABSTRACT

Expansion of the vasa vasorum network has been observed in a variety of systemic and pulmonary vascular diseases. We recently reported that a marked expansion of the vasa vasorum network occurs in the pulmonary artery adventitia of chronically hypoxic calves. Since hypoxia has been shown to stimulate ATP release from both vascular resident as well as circulatory blood cells, these studies were undertaken to determine if extracellular ATP exerts angiogenic effects on isolated vasa vasorum endothelial cells (VVEC) and/or if it augments the effects of other angiogenic factors (VEGF and basic FGF) known to be present in the hypoxic microenvironment. We found that extracellular ATP dramatically increases DNA synthesis, migration, and rearrangement into tube-like networks on Matrigel in VVEC, but not in pulmonary artery (MPAEC) or aortic (AOEC) endothelial cells obtained from the same animals. Extracellular ATP potentiated the effects of both VEGF and bFGF to stimulate DNA synthesis in VVEC but not in MPAEC and AOEC. Analysis of purine and pyrimidine nucleotides revealed that ATP, ADP and MeSADP were the most potent in stimulating mitogenic responses in VVEC, indicating the involvement of the family of P2Y1-like purinergic receptors. Using pharmacological inhibitors, Western blot analysis, and Phosphatidylinositol-3 kinase (PI3K) in vitro kinase assays, we found that PI3K/Akt/mTOR and ERK1/2 play a critical role in mediating the extracellular ATP-induced mitogenic and migratory responses in VVEC. However, PI3K/Akt and mTOR/p70S6K do not significantly contribute to extracellular ATP-induced tube formation on Matrigel. Our studies indicate that VVEC, isolated from the sites of active angiogenesis, exhibit distinct functional responses to ATP, compared to endothelial cells derived from large pulmonary or systemic vessels. Collectively, our data support the idea that extracellular ATP participates in the expansion of the vasa vasorum that can be observed in hypoxic conditions.


Subject(s)
Adenosine Triphosphate/pharmacology , Angiogenesis Inducing Agents/pharmacology , Endothelial Cells/drug effects , Extracellular Space/metabolism , Pulmonary Artery/cytology , Vasa Vasorum/cytology , Vasa Vasorum/drug effects , Animals , Aorta/cytology , Aorta/enzymology , Cattle , Cell Movement/drug effects , Collagen/metabolism , DNA/biosynthesis , Drug Combinations , Endothelial Cells/enzymology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Space/drug effects , Fibroblast Growth Factor 2/pharmacology , Laminin/metabolism , Neovascularization, Physiologic/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Proteoglycans/metabolism , Pulmonary Artery/enzymology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases , Vasa Vasorum/enzymology , Vascular Endothelial Growth Factor A/pharmacology
17.
Plast Reconstr Surg ; 117(6): 1860-75, 2006 May.
Article in English | MEDLINE | ID: mdl-16651959

ABSTRACT

BACKGROUND: The authors previously described induction of spontaneous tissue generation by implanting a collagen matrix and a ligated pedicle (arteriovenous bundle) into a hollow porous chamber in vivo in the rabbit. They hypothesized that increased tissue volume could be obtained by the application of basic fibroblast growth factor (bFGF) and/or by increasing the chamber size and porosity. METHODS: In rabbits, a saphenous arteriovenous pedicle and a collagen sponge were inserted into a porous chamber in the groin. Small-volume pore chambers (experiment 1, n = 7) and larger-volume, wider pore chambers (experiment 2, n = 13) were compared, and each was compared with and without bFGF. An additional three flaps of experiment 2 with bFGF were skin grafted, microsurgically transplanted to the ear, and evaluated at 6 months for stability. RESULTS: All patent chambers grew tissue; chambers with bFGF were almost filled, and those without were only half-filled. Histomorphometric analysis confirmed a significant difference. The larger-volume, larger-pore chambers produced more than twice the volume of tissue as the smaller chambers did, and this was significant. Tissue volume in both the control and bFGF groups of experiment 2 was significantly greater than that in the respective groups of experiment 1. Histology, angiography, and scanning electron microscopy confirmed greater vascularity in the bFGF groups and demonstrated vascular connections penetrating the chamber pores linking with angiogenic sprouts, probably from the vasa vasorum of the pedicle, to contribute to new growth. Transplanted flaps survived and appeared normal 6 months later. CONCLUSIONS: Patent pedicles, bFGF, large pore size, and larger-volume chambers all seemed to contribute to increased tissue growth in this model. The tissue is stable long term.


Subject(s)
Diffusion Chambers, Culture , Fibroblast Growth Factor 2/pharmacology , Skin Transplantation/methods , Skin, Artificial , Surgical Flaps , Tissue Engineering/methods , Animals , Arteries/surgery , Collagen , Ear, External/surgery , Equipment Design , Granulation Tissue/ultrastructure , Groin , Male , Microscopy, Electron, Scanning , Microsurgery , Neovascularization, Physiologic , Rabbits , Saphenous Vein/surgery , Surgical Sponges , Tissue Engineering/instrumentation , Transplantation, Autologous , Vasa Vasorum/cytology , Vasa Vasorum/ultrastructure
18.
Anat Rec A Discov Mol Cell Evol Biol ; 281(2): 1372-82, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15486955

ABSTRACT

The vasa vasorum of skeletonized and nonskeletonized segments of five human great saphenous veins (GSVs), harvested during coronary bypass grafting, were cannulated, rinsed, and injected (casted) with the polymerizing resin Mercox-Cl-2B. After removal of the dry vascular tissue, the casts were examined using scanning electron microscopy. Stereopaired images (tilt angle, 6 degrees ) were taken, imported into a 3D morphometry system, and the 3D architecture of the vasa vasorum (arterial and venous vasa as well as capillaries) was studied qualitatively and quantitatively in terms of vasa diameters, intervascular and interbranching distances, and branching angles. Diameters of parent (d(0)) and large (d(1)) and small (d(2)) daughter vessels of arterial and venous bifurcations served to calculate asymmetry ratios (alpha) and area ratios (beta). Additionally, deviations of bifurcations and branching angles from optimal branches were calculated for selected arterial vasa. The arrangement of the vasa vasorum closely followed the longitudinally oriented connective tissue fibers in the adventitia and the circularly arranged smooth muscle cell layers within the outer layers of the media. Venous vasa by far outnumbered arterial vasa. Vasa vasorum changed their course several times in acute angles and revealed numerous circular constrictions, kinks, and outpouchings. Due to their spatial arrangement, the vasa vasorum are prone to tolerate vessel wall distension generated by acute increases in blood pressure or stretching of the vessel without severe impact on vessel functions. Preliminary comparisons of data from the bifurcations of cast arterial vasa vasorum, with calculated optimal bifurcations, do not yet give clear insights into the optimality principle(s) governing the design of arterial vasa vasorum bifurcations of the human GSVs.


Subject(s)
Corrosion Casting , Vasa Vasorum/anatomy & histology , Vasa Vasorum/ultrastructure , Veins/transplantation , Aged , Capillaries/anatomy & histology , Capillaries/cytology , Humans , Microscopy, Electron, Scanning , Vasa Vasorum/cytology , Veins/anatomy & histology , Veins/cytology
19.
Surg Radiol Anat ; 24(6): 377-81, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12647022

ABSTRACT

The distribution of the vasa vasorum of the human great saphenous vein (GSV) was studied on veins taken both post-mortem and peroperatively. It was found that the stems of feeding vessels approach the venous wall at intervals of 1.5-2.5 cm; their smaller branches first passed the fascial compartments of the GSV and then entered the adventitia at intervals of 0.5-1.5 cm on both the stem and the largest tributaries of the GSV. In the stem regions vasa vasorum arteries and veins ran together but, between neighboring stems, isolated venae vasorum were regularly found which opened individually into terminal segments of the largest tributaries of the GSV. Neither by dissection nor by injection methods were venae vasorum found to open directly into the lumen of the GSV stem. The total thickness of the media ranged between 500 and 1300 micro m, according to the state of constriction of the venous wall before fixation. Two structurally different layers of GSV tunica media were present: an inner loose layer and an outer dense layer, both of similar thickness. The innermost capillaries of the vasa vasorum network were found in all cases on the border between the two layers of media. No lymphatic was found in any of the layers of GSV wall. From the findings the authors recommend extremely careful dissection of the GSV wall during in situ grafting surgery, to ensure the best viability of the venous wall.


Subject(s)
Saphenous Vein/anatomy & histology , Vasa Vasorum/anatomy & histology , Aged , Female , Humans , Male , Microdissection/methods , Middle Aged , Plastic Embedding , Staining and Labeling , Vasa Vasorum/cytology
20.
Cardiovasc Intervent Radiol ; 21(1): 45-9, 1998.
Article in English | MEDLINE | ID: mdl-9473546

ABSTRACT

PURPOSE: To evaluate the effects of bare stents and covered stents on the aortic wall, especially the vasa vasorum. METHODS: Eight bare stents and nine covered stents were placed in the infrarenal aorta of nine dogs. The dogs were euthanized at 4-45 weeks after stent placement. The vasa vasorum was evaluated by microstereoscopy with vascular casting, and the histopathology of the aortic wall was examined by light microscopy. RESULTS: In the unstented normal aorta, vasa vasorum nourished the adventitia and the outer media, and the intima and inner media were avascular. In the stented segment, vascular dilatation and proliferation of vasa vasorum, medial atrophy, and intimal hyperplasia were observed, more prominent for covered stents than for bare stents. CONCLUSION: Intravascular stent placement caused not only medial atrophy and intimal hyperplasia but also proliferation of the vasa vasorum, probably due to hypoxia in the aortic wall.


Subject(s)
Aorta, Abdominal/surgery , Blood Vessel Prosthesis Implantation , Stents , Vasa Vasorum , Animals , Aorta, Abdominal/cytology , Cell Division , Collateral Circulation , Dogs , Follow-Up Studies , Muscle, Smooth, Vascular/blood supply , Muscle, Smooth, Vascular/cytology , Vasa Vasorum/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...