Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
Braz J Cardiovasc Surg ; 38(6): e20230045, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37797088

ABSTRACT

This short article discusses selected scanning electron microscope and transmission electron microscope features of vasa vasorum including pericytes and basement membrane of the human saphenous vein (SV) harvested with either conventional (CON) or no-touch (NT) technique for coronary artery bypass grafting. Scanning electron microscope data shows the general damage to vasa vasorum of CON-SV, while the transmission electron microscope data presents ultrastructural features of the vasa in more detail. Hence there are some features suggesting pericyte involvement in the contraction of vasa blood vessels, particularly in CON-SV. Other features associated with the vasa vasorum of both CON-SV and NT-SV preparations include thickened and/or multiplied layers of the basement membrane. In some cases, multiple layers of basement membrane embrace both pericyte and vasa microvessel making an impression of a "unit" made by basement membrane-pericyte-endothelium/microvessel. It can be speculated that this structural arrangement has an effect on the contractile and/or relaxing properties of the vessels involved. Endothelial colocalization of immunoreactive inducible nitric oxide synthase and endothelin-1 can be observed (with laser confocal microscope) in some of the vasa microvessels. It can be speculated that this phenomenon, particularly of the expression of inducible nitric oxide synthase, might be related to structurally changed vasa vessels, e.g., with expanded basement membrane. Fine physiological relationships between vasa vasorum endothelium, basement membrane, pericyte, and perivascular nerves have yet to be uncovered in the detail needed for better understanding of the cells'specific effects in SV preparations for coronary artery bypass grafting.


Subject(s)
Saphenous Vein , Vasa Vasorum , Humans , Saphenous Vein/transplantation , Nitric Oxide Synthase Type II/metabolism , Vasa Vasorum/metabolism , Vasa Vasorum/ultrastructure , Coronary Artery Bypass/methods , Endothelium, Vascular
2.
Int J Mol Sci ; 24(3)2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36768296

ABSTRACT

Current techniques for the detection of vasa vasorum (VV) in vascular pathology include staining for endothelial cell (EC) markers such as CD31 or VE-cadherin. However, this approach does not permit an objective assessment of vascular geometry upon vasospasm and the clinical relevance of endothelial specification markers found in developmental biology studies remains unclear. Here, we performed a combined immunostaining of rat abdominal aorta (rAA) and human saphenous vein (hSV) for various EC or vascular smooth muscle cell (VSMC) markers and found that the latter (e.g., alpha smooth muscle actin (α-SMA) or smooth muscle myosin heavy chain (SM-MHC)) ensure a several-fold higher signal-to-noise ratio irrespective of the primary antibody origin, fluorophore, or VV type (arterioles, venules, or capillaries). Further, α-SMA or SM-MHC staining allowed unbiased evaluation of the VV area under vasospasm. Screening of the molecular markers of endothelial heterogeneity (mechanosensitive transcription factors KLF2 and KLF4, arterial transcription factors HES1, HEY1, and ERG, venous transcription factor NR2F2, and venous/lymphatic markers PROX1, LYVE1, VEGFR3, and NRP2) have not revealed specific markers of any lineage in hSV (although KLF2 and PROX1 were restricted to venous endothelium in rAA), suggesting the need in high-throughput searches for the clinically relevant signatures of arterial, venous, lymphatic, or capillary differentiation.


Subject(s)
Endothelial Cells , Endothelium, Vascular , Muscle, Smooth, Vascular , Transcription Factors , Vasa Vasorum , Animals , Humans , Rats , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Kruppel-Like Transcription Factors/metabolism , Saphenous Vein , Transcription Factors/metabolism , Vasa Vasorum/metabolism , Vasa Vasorum/pathology
3.
Microvasc Res ; 147: 104479, 2023 05.
Article in English | MEDLINE | ID: mdl-36690271

ABSTRACT

Isolated endothelial cells are valuable in vitro model for vascular research. At present, investigation of disease-relevant changes in vascular endothelium at the molecular level requires established endothelial cell cultures, preserving vascular bed-specific phenotypic characteristics. Vasa vasorum (VV) form a microvascular network around large blood vessels, in both the pulmonary and systemic circulations, that are critically important for maintaining the integrity and oxygen supply of the vascular wall. However, despite the pathophysiological significance of the VV, methods for the isolation and culture of vasa vasorum endothelial cells (VVEC) have not yet been reported. In our prior studies, we demonstrated the presence of hypoxia-induced angiogenic expansion of the VV in the pulmonary artery (PA) of neonatal calves; an observation which has been followed by a series of in vitro studies on isolated PA VVEC. Here we present a detailed protocol for reproducible isolation, purification, and culture of PA VVEC. We show these cells to express generic endothelial markers, (vWF, eNOS, VEGFR2, Tie1, and CD31), as well as progenitor markers (CD34 and CD133), bind lectin Lycopersicon Esculentum, and incorporate acetylated low-density lipoproteins labeled with acetylated LDL (DiI-Ac-LDL). qPCR analysis additionally revealed the expression of CD105, VCAM-1, ICAM-1, MCAM, and NCAM. Ultrastructural electron microscopy and immunofluorescence staining demonstrated that VVEC are morphologically characterized by a developed actin and microtubular cytoskeleton, mitochondrial network, abundant intracellular vacuolar/secretory system, and cell-surface filopodia. VVEC exhibit exponential growth in culture and can be mitogenically activated by multiple growth factors. Thus, our protocol provides the opportunity for VVEC isolation from the PA, and potentially from other large vessels, enabling advances in VV research.


Subject(s)
Adventitia , Vasa Vasorum , Animals , Cattle , Vasa Vasorum/metabolism , Pulmonary Artery/metabolism , Endothelial Cells/metabolism , Biology
4.
J Biomed Mater Res A ; 110(12): 1912-1920, 2022 12.
Article in English | MEDLINE | ID: mdl-35770946

ABSTRACT

Pericytes are essential components of small blood vessels and are found in human aortic vasa vasorum. Prior work uncovered lower vasa vasorum density and decreased levels of pro-angiogenic growth factors in adventitial specimens of human ascending thoracic aortic aneurysm. We hypothesized that adventitial extracellular matrix (ECM) from normal aorta promotes pericyte function by increasing pericyte contractile function through mechanisms deficient in ECM derived from aneurysmal aortic adventitia. ECM biomaterials were prepared as lyophilized particulates from decellularized adventitial specimens of human and porcine aorta. Immortalized human aortic adventitia-derived pericytes were cultured within Type I collagen gels in the presence or absence of human or porcine adventitial ECMs. Cell contractility index was quantified by measuring the gel area immediately following gelation and after 48 h of culture. Normal human and porcine adventitial ECM increased contractility of pericytes when compared with pericytes cultured in absence of adventitial ECM. In contrast, aneurysm-derived human adventitial ECM failed to promote pericyte contractility. Pharmacological inhibition of TGFßR1 and antibody blockade of α2 ß1 integrin independently decreased porcine adventitial ECM-induced pericyte contractility. By increasing pericyte contractility, adventitial ECM may improve microvascular function and thus represents a candidate biomaterial for less invasive and preventative treatment of human ascending aortic disease.


Subject(s)
Adventitia , Vasa Vasorum , Adventitia/metabolism , Animals , Biocompatible Materials/metabolism , Collagen Type I/metabolism , Extracellular Matrix , Humans , Hydrogels/metabolism , Hydrogels/pharmacology , Integrins/metabolism , Pericytes , Swine , Transforming Growth Factor beta/metabolism , Vasa Vasorum/metabolism
5.
Am J Physiol Heart Circ Physiol ; 320(6): H2438-H2447, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33961504

ABSTRACT

Adventitial abnormalities including enhanced vasa vasorum malformation are associated with development and vulnerability of atherosclerotic plaque. However, the mechanisms of vasa vasorum malformation and its role in vascular remodeling have not been fully clarified. We recently reported that ninjurin-1 (Ninj1) is a crucial adhesion molecule for pericytes to form matured neovessels. The purpose is to examine if Ninj1 regulates adventitial angiogenesis and affects the vascular remodeling of injured vessels using pericyte-specific Ninj1 deletion mouse model. Mouse femoral arteries were injured by insertion of coiled wire. Four weeks after vascular injury, fixed arteries were decolorized. Vascular remodeling, including intimal hyperplasia and adventitial microvessel formation were estimated in a three-dimensional view. Vascular fragility, including blood leakiness was estimated by extravasation of fluorescein isothiocyanate (FITC)-lectin or FITC-dextran from microvessels. Ninj1 expression was increased in pericytes in response to vascular injury. NG2-CreER/Ninj1loxp mice were treated with tamoxifen (Tam) to induce deletion of Ninj1 in pericyte (Ninj1 KO). Tam-treated NG2-CreER or Tam-nontreated NG2-CreER/Ninj1loxp mice were used as controls. Intimal hyperplasia was significantly enhanced in Ninj1 KO compared with controls. Vascular leakiness was significantly enhanced in Ninj1 KO. In Ninj1 KO, the number of infiltrated macrophages in adventitia was increased, along with the expression of inflammatory cytokines. In conclusion, deletion of Ninj1 in pericytes induces the immature vasa vasorum formation of injured vasculature and exacerbates adventitial inflammation and intimal hyperplasia. Thus, Ninj1 contributes to the vasa vasorum maturation in response to vascular injury and to reduction of vascular remodeling.NEW & NOTEWORTHY Although abnormalities of adventitial vasa vasorum are associated with vascular remodeling such as atherosclerosis, the mechanisms of vasa vasorum malformation and its role in vascular remodeling have not been fully clarified. The present study provides a line of novel evidence that ninjurin-1 contributes to adventitial microvascular maturation during vascular injury and regulates vascular remodeling.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Femoral Artery/metabolism , Neointima/genetics , Nerve Growth Factors/genetics , Pericytes/metabolism , Vasa Vasorum/metabolism , Vascular Remodeling/genetics , Adventitia/metabolism , Adventitia/pathology , Animals , Femoral Artery/injuries , Femoral Artery/pathology , Gene Knockout Techniques , Hyperplasia/genetics , Inflammation/genetics , Inflammation/metabolism , Macrophages/pathology , Mice , Neointima/pathology , Neovascularization, Physiologic/genetics , Transcriptome , Tunica Intima/metabolism , Tunica Intima/pathology , Vasa Vasorum/pathology , Vascular System Injuries/genetics , Vascular System Injuries/metabolism , Vascular System Injuries/pathology
6.
Am J Physiol Cell Physiol ; 319(1): C183-C193, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32432925

ABSTRACT

The vasa vasorum (VV), the microvascular network around large vessels, has been recognized as an important contributor to the pathological vascular remodeling in cardiovascular diseases. In bovine and rat models of hypoxic pulmonary hypertension (PH), we have previously shown that chronic hypoxia profoundly increased pulmonary artery (PA) VV permeability, associated with infiltration of inflammatory and progenitor cells in the arterial wall, perivascular inflammation, and structural vascular remodeling. Extracellular adenosine was shown to exhibit a barrier-protective effect on VV endothelial cells (VVEC) via cAMP-independent mechanisms, which involved adenosine A1 receptor-mediated activation of Gi-phosphoinositide 3-kinase-Akt pathway and actin cytoskeleton remodeling. Using VVEC isolated from the adventitia of calf PA, in this study we investigated in more detail the mechanisms linking Gi activation to downstream barrier protection pathways. Using a small-interference RNA (siRNA) technique and transendothelial electrical resistance assay, we found that the adaptor protein, engulfment and cell motility 1 (ELMO1), the tyrosine phosphatase Src homology region 2 domain-containing phosphatase-2, and atypical Gi- and Rac1-mediated protein kinase A activation are implicated in VVEC barrier enhancement. In contrast, the actin-interacting GTP-binding protein, girdin, and the p21-activated kinase 1 downstream target, LIM kinase, are not involved in this response. In addition, adenosine-dependent cytoskeletal rearrangement involves activation of cofilin and inactivation of ezrin-radixin-moesin regulatory cytoskeletal proteins, consistent with a barrier-protective mechanism. Collectively, our data indicate that targeting adenosine receptors and downstream barrier-protective pathways in VVEC may have a potential translational significance in developing pharmacological approach for the VV barrier protection in PH.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenosine/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelial Cells/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Vasa Vasorum/metabolism , rac1 GTP-Binding Protein/metabolism , Adenosine/pharmacology , Animals , Cattle , Endothelial Cells/drug effects , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Male , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Vasa Vasorum/drug effects
7.
Cardiovasc Res ; 116(3): 708-720, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31241138

ABSTRACT

AIMS: Adventitial vasa vasorum provides oxygen and nourishment to the vascular wall, but whether it regulates vascular disease remains unclear. We have previously shown that an increased expression of VEGF (vascular endothelial growth factor) is associated with macrophage infiltration. This study aims to determine whether adventitial fibroblast (AF)-derived VEGF increases the number of vasa vasorum contributing to neointima formation through macrophage recruitment. METHODS AND RESULTS: In rat balloon injury model, vasa vasorum count was increased particularly in the adventitia accompanied by cell proliferation and VEGF expression. Both endogenous and PKH26-labelled exogenous macrophages were mainly distributed in adventitia around vasa vasorum. Interestingly, perivascular delivery of Ranibizumab preferentially concentrated in adventitia resulted in a decrease of neointima formation with concurrent reduction of vasa vasorum count and macrophage infiltration. AFs with adenovirus-mediated VEGF over-expression delivered to the adventitia significantly enhanced these pathological changes after injury. In Tie2-cre/Rosa-LoxP-RFP mice, endothelial cells were increased in the adventitia after wire injury. By using multiphoton laser scanning microscopy, macrophage rolling, adhesion and transmigration were observed in vasa vasorum. Moreover, adoptive transfer of macrophages accelerated injury-induced neointima formation. VEGF-neutralizing antibody administration also attenuated wire injury-induced neointima formation and macrophage infiltration. In primary cultured AFs, exogenous VEGF increased VEGF expression and secretion in a time- and dose-dependent manner. AF-conditioned medium promoted endothelial cell angiogenesis, vascular cell adhesion molecule-1 expression and macrophage adhesion was blocked by VEGF-neutralizing antibody and VEGFR2 inhibitor ZM323881, which also inhibited activation of VEGFR2/ERK1/2 pathway. CONCLUSION: These results demonstrate that AF-derived VEGF plays a significant role in the increase of vasa vasorum count which is involved in macrophage recruitment and neointima formation.


Subject(s)
Adventitia/metabolism , Carotid Arteries/metabolism , Carotid Artery Injuries/metabolism , Femoral Artery/metabolism , Fibroblasts/metabolism , Macrophages/metabolism , Neointima , Vasa Vasorum/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular System Injuries/metabolism , Adoptive Transfer , Adventitia/drug effects , Adventitia/pathology , Angiogenesis Inhibitors/pharmacology , Animals , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Carotid Artery Injuries/prevention & control , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Femoral Artery/drug effects , Femoral Artery/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Macrophages/drug effects , Macrophages/pathology , Macrophages/transplantation , Male , Mice, Inbred C57BL , Paracrine Communication , Rats, Sprague-Dawley , Signal Transduction , Tissue Culture Techniques , Vasa Vasorum/drug effects , Vasa Vasorum/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular System Injuries/genetics , Vascular System Injuries/pathology , Vascular System Injuries/prevention & control
8.
Biomed Pharmacother ; 114: 108785, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30909142

ABSTRACT

OBJECTIVE: To observe the intervention effect of Si-miao-Yong-An (SMYA) on atheroosclerosis (AS) vulnerable plaque, and to explore the mechanism by Vasa Vasorum (VV) maturation as a starting point. MATERIALS AND METHODS: SPF-class healthy male ApoE-/- mice were randomly divided into model group, SMYA group and simvastatin group, and C57BL/6 mice were used as a control group. After 8 weeks of drug intervention, the plaques of AS were observed by HE staining. The pericytes of aortic root plaques were observed by immunofiuorescence double staining (CD34, Desmin) and the density of VV. The expression of Dll4, Notch1, Hey1 and VEGF mRNA in aortic tissues was detected by real-time qPCR. RESULTS: SMYA significantly reduced the area of aortic plaque in ApoE-/- mice, significantly reduced plaque area and the ratio of plaque to lumen area, and reduced the intima medium thickness, it's effect was greater than that of simvastatin; it significantly increased the density of VV in plaque. SMYA increased the expression of Dll4 and Notch1 and Hey1mRNA, and decreased the expression of VEGF mRNA, and its effect was greater than that of simvastatin. CONCLUSION: SMYA can reduce the AS plaque area in ApoE-/- mice, promote the recruitment of VV pericytes, and stabilize AS vulnerable plaques. The mechanism may be regulate of Dll4/Notch1/ Hey1/VEGF signaling pathway. At the same time, it has a dual-direction regulation on the VV.


Subject(s)
Apolipoproteins E/metabolism , Atherosclerosis/drug therapy , Drugs, Chinese Herbal/pharmacology , Plaque, Atherosclerotic/drug therapy , Vasa Vasorum/drug effects , Animals , Atherosclerosis/metabolism , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Plaque, Atherosclerotic/metabolism , Simvastatin/pharmacology , Vasa Vasorum/metabolism
9.
Cardiovasc Hematol Agents Med Chem ; 16(2): 114-119, 2018.
Article in English | MEDLINE | ID: mdl-30394217

ABSTRACT

BACKGROUND: Atherosclerosis is a chronic inflammatory disease which may lead to major cardiovascular events. The primary cause of atherosclerosis is Dyslipidemia. The increased level of lipid profile triggers endothelial dysfunction. This results in inflammation with the recruitment of monocyte, macrophage, T lymphocyte, and Mast cells secreted by an Lp-PLA2 enzyme which causes binding between macrophage and oxidized LDL. This binding results in the formation of foam cells and also the migration of smooth muscle cells. Following that, an Lp-PLA2 receptor hydrolizes OxPC which results in LysoPC and OxNEFA, bioactive compounds which stimulate the progression of atherosclerosis plaques. This process leads to cell hypoxia, which may result in the increase of HIF-1α and VEGF expressions and induction of vasa vasorum angiogenesis. Employing darapladib as an agent of Lp-PLA2 selective inhibitors, this study aimed to find out the effect of darapladib as an Lp- PLA2 selective inhibitor agent on the formation of vasa vasorum angiogenesis and the decrease of HIF-1α and VEGF expression in aortic tissue of rats with dyslipidemia. METHOD: A true laboratory experiment with a randomized post-test control group design used 30 male spraque dowley rats as animal models which were divided into 6 groups: Normal 8 weeks, Normal 16 weeks, Dyslipidemia (DL) 8 weeks, Dyslipidemia (DL) 16 weeks, Dyslipidemia with darapladib treatment (DLDP) 8 weeks and Dyslipidemia with darapladib treatment (DLDP) 16 weeks. The data measured in this study were the lipid profile (total cholesterol, HDL, and LDL). Using EnzyChrom TM kit, hematoxylin eosin, and double-labelling immunofluorescene, the levels of lipid profile, vasa vasorum, HIF-1α and VEGF were measured. RESULTS: The study results which were analyzed using NOVA test showed that with darapladib administration, there was a significant decrease in vasa vasorum angiogenesis (p=0.000), HIF-1α (p=0.005) and VEGF (p=0.009) expression in each time series. This result proves that Lp-PLA2 inhibitor reduces inflammatory process. CONCLUSION: Darapladib injection as an Lp-PLA2 selective inhibitor correlates with the decreasing vasa vasorum angiogenesis through alteration in HIF-1α and VEGF expressions in the aorta of high fat diet rats. We recommend further experiments to determine the effectiveness of darapladib with earlier time series in the atherosclerosis process.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/antagonists & inhibitors , Atherosclerosis/complications , Atherosclerosis/drug therapy , Benzaldehydes/therapeutic use , Dyslipidemias/complications , Neovascularization, Pathologic/complications , Neovascularization, Pathologic/drug therapy , Oximes/therapeutic use , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Dyslipidemias/metabolism , Dyslipidemias/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Rats, Sprague-Dawley , Vasa Vasorum/drug effects , Vasa Vasorum/metabolism , Vasa Vasorum/pathology , Vascular Endothelial Growth Factor A/metabolism
10.
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
11.
Oxid Med Cell Longev ; 2018: 2028936, 2018.
Article in English | MEDLINE | ID: mdl-29743974

ABSTRACT

Heme oxygenase-1 (HO-1), encoded by HMOX1 gene and regulated by Nrf2 transcription factor, is a cytoprotective enzyme. Its deficiency may exacerbate abdominal aortic aneurysm (AAA) development, which is also often associated with hyperlipidemia. Beneficial effects of statins, the broadly used antilipidemic drugs, were attributed to modulation of Nrf2/HO-1 axis. However, the effect of statins on Nrf2/HO-1 pathway in patients with AAA has not been studied yet. We analyzed AAA tissue from patients treated with simvastatin (N = 28) or without statins (N = 14). Simvastatin treatment increased HO-1 protein level in AAA, both in endothelial cells (ECs) and in smooth muscle cells (SMCs), but increased Nrf2 localization was restricted only to vasa vasorum. Nrf2 target genes HMOX1, NQO1, and GCLM expression remained unchanged in AAA. In vitro studies showed that simvastatin raises HO-1 protein level slightly in ECs and to much higher extent in SMCs, which is not related to Nrf2/ARE activation, although HMOX1 expression is upregulated by simvastatin in both cell types. In conclusion, simvastatin-induced modulation of HO-1 level in ECs and SMCs in vitro is not related to Nrf2/ARE activity. Likewise, divergent HO-1 and Nrf2 localization together with stable expression of Nrf2 target genes, including HMOX1, in AAA tissue denotes Nrf2 independency.


Subject(s)
Aortic Aneurysm, Abdominal/drug therapy , Endothelial Cells/physiology , Heme Oxygenase-1/metabolism , Hyperlipidemias/drug therapy , Myocytes, Smooth Muscle/physiology , Simvastatin/therapeutic use , Vasa Vasorum/metabolism , Aged , Aged, 80 and over , Animals , Aortic Aneurysm, Abdominal/pathology , Cells, Cultured , Female , Heme Oxygenase-1/genetics , Humans , Male , Middle Aged , NF-E2-Related Factor 2/metabolism , Up-Regulation
12.
Front Immunol ; 9: 706, 2018.
Article in English | MEDLINE | ID: mdl-29719532

ABSTRACT

Plaque microvascularization and increased endothelial permeability are key players in the development of atherosclerosis, from the initial stages of plaque formation to the occurrence of acute cardiovascular events. First, endothelial dysfunction and increased permeability facilitate the entry of diverse inflammation-triggering molecules and particles such as low-density lipoproteins into the artery wall from the arterial lumen and vasa vasorum (VV). Recognition of entering particles by resident phagocytes in the vessel wall triggers a maladaptive inflammatory response that initiates the process of local plaque formation. The recruitment and accumulation of inflammatory cells and the subsequent release of several cytokines, especially from resident macrophages, stimulate the expansion of existing VV and the formation of new highly permeable microvessels. This, in turn, exacerbates the deposition of pro-inflammatory particles and results in the recruitment of even more inflammatory cells. The progressive accumulation of leukocytes in the intima, which trigger proliferation of smooth muscle cells in the media, results in vessel wall thickening and hypoxia, which further stimulates neoangiogenesis of VV. Ultimately, this highly inflammatory environment damages the fragile plaque microvasculature leading to intraplaque hemorrhage, plaque instability, and eventually, acute cardiovascular events. This review will focus on the pivotal roles of endothelial permeability, neoangiogenesis, and plaque microvascularization by VV during plaque initiation, progression, and rupture. Special emphasis will be given to the underlying molecular mechanisms and potential therapeutic strategies to selectively target these processes.


Subject(s)
Neovascularization, Pathologic , Vasa Vasorum/metabolism , Vasa Vasorum/pathology , Adaptation, Biological , Animals , Atherosclerosis/drug therapy , Atherosclerosis/etiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biomarkers , Capillary Permeability , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Disease Models, Animal , Disease Progression , Disease Susceptibility , Endothelial Cells/metabolism , Energy Metabolism , Epigenesis, Genetic , Humans , MicroRNAs/genetics , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/metabolism , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Risk Factors , Tunica Intima/growth & development , Tunica Intima/metabolism , Tunica Intima/pathology , Vasa Vasorum/drug effects , Vasculitis/complications , Vasculitis/pathology
13.
Int J Cardiol ; 265: 181-187, 2018 Aug 15.
Article in English | MEDLINE | ID: mdl-29685689

ABSTRACT

BACKGROUND: Vasa vasorum (VV) angiogenesis is increased in type 2 diabetes mellitus (T2DM) and may promote atherosclerotic plaque rupture. We sought to determine whether insulin resistance adipocyte-derived exosomes (IRADEs) played a major role in modulating VV angiogenesis and the mechanisms involved. METHODS: The characterization of IRADEs was performed by electron microscopy, NTA (Nanoparticle Tracking Analysis) and western blot. The cellular effects of IRADEs on angiogenesis were explored in human umbilical vein endothelial cells (HUVECs) and murine aortic endothelial cells (MAECs) in vitro. The roles of IRADEs in angiogenesis were demonstrated with aortic ring and matrigel plug assays ex vivo and the plaque burden, plaque stability and angiogenesis-related protein expression in vivo were evaluated by ultrasonography, immunohistochemistry and western blot. RESULTS: The IRADEs had a cup-shaped morphology, could be taken up by HUVECs and atherosclerotic plaques, and promoted tube formation by shh in vitro. In the aortic ring and matrigel plug assays, angiogenesis was significantly increased in the IRADEs group. Exogenously administered shh-containing IRADEs increased VV angiogenesis, the plaque burden, the vulnerability index and the expression of angiogenesis-related factors, whereas these effects were attenuated by silencing shh in IRADEs. CONCLUSIONS: In conclusion, IRADEs promote plaque burden and plaque vulnerability partly by inducing VV angiogenesis, which occurs partly through shh. Accordingly, the application of IRADEs may serve as a novel therapeutic approach to treat diabetic atherosclerosis.


Subject(s)
Adipocytes/metabolism , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin Resistance/physiology , Vasa Vasorum/metabolism , 3T3 Cells , Adipocytes/pathology , Animals , Atherosclerosis/pathology , Cells, Cultured , Diabetes Mellitus, Type 2/pathology , Exosomes/metabolism , Exosomes/pathology , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Knockout , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Vasa Vasorum/pathology
14.
Curr Drug Targets ; 19(11): 1327-1332, 2018.
Article in English | MEDLINE | ID: mdl-29359664

ABSTRACT

BACKGROUND: An abdominal aortic aneurysm (AAA), which affects approximately 10% of Japanese men aged ≥ 65 years, is frequently associated with hypertension, dyslipidemia, and other lifestyle- related diseases. The development of an AAA is attributed to chronic inflammation concomitant with arteriosclerotic changes. However, an accurate pathomechanism associated with AAA remains uncertain, and questions such as why only a particular group/percentage of patients with arteriosclerosis develop aneurysms and how diabetes suppresses aneurysm development remain unanswered. OBJECTIVE: We examined a novel mechanism to develop AAA based on histopathological findings following analysis of the human AAA tissues. Additionally, based on these findings, we developed a new animal model of AAA, in which the histopathological characteristics are similar to human AAA tissue. RESULTS: Recently, we identified stenosis of the vasa vasorum (VV) in aortic segments showing dilatation. The aorta is the largest artery in our circulatory system. Under physiological conditions, the inner layer of the aorta is nourished via direct diffusion of nutrients from the luminal blood flow, whereas the outer adventitia is primarily perfused by the VV. Therefore, hypoperfusion of the VV induces hypoxia and subsequent inflammation and tissue degeneration of the aortic wall, resulting in aneurysm formation. Based on these findings, we established an AAA animal model by reducing the blood flow through the VV to the aortic wall. AAA was successfully reproduced in our animal model. Histopathological findings in this model were indistinguishable from those observed in humans, and pronounced abnormality in lipid composition in blood vessel adventitia was also observed. CONCLUSION: Thus, hypoperfusion of the aortic wall appeared to be sufficient to cause inflammationinduced AAA. These findings may provide potential targets for novel therapeutics for the management of an AAA.


Subject(s)
Adventitia/pathology , Aortic Aneurysm, Abdominal/pathology , Vasa Vasorum/pathology , Aged , Aged, 80 and over , Animals , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/metabolism , Disease Models, Animal , Humans , Japan , Lipid Metabolism , Male , Rats , Research Design , Vasa Vasorum/immunology , Vasa Vasorum/metabolism
15.
Vascul Pharmacol ; 96-98: 5-10, 2017 09.
Article in English | MEDLINE | ID: mdl-28830735

ABSTRACT

Vasa vasorum are blood microvessels which penetrate the adventitia and outer layers of the media of large blood vessels, supplying them with nutrients and oxygen. A growing body of evidence suggests that vasa vasorum play a central role in the pathogenesis of atherosclerosis. In this review, we will make a case for the role of microvascular dysfunction in the initiation of disease. When seen through this lens, new therapeutic opportunities for prevention can be envisioned. In particular, we discuss how targeting the cellular metabolism and epigenetic machinery of vasa vasorum neovessels could be harnessed to render vasa vasorum endothelial cells less sensitive to atherogenic stimuli.


Subject(s)
Atherosclerosis/prevention & control , Cardiovascular Agents/therapeutic use , Vasa Vasorum/drug effects , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Capillary Permeability , Epigenesis, Genetic/drug effects , Humans , Inflammation Mediators/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Microcirculation/drug effects , Neovascularization, Pathologic , Signal Transduction/drug effects , Vasa Vasorum/metabolism , Vasa Vasorum/pathology , Vasa Vasorum/physiopathology
16.
Mediators Inflamm ; 2017: 3795142, 2017.
Article in English | MEDLINE | ID: mdl-28133420

ABSTRACT

Chronic kidney disease (CKD) patients, characterized by traditional and nontraditional risk factors, are prone to develop atheromatosis and thus cardiovascular events and mortality. The angiogenesis of the adventitial vasa vasorum (aVV) surrounding the carotid has been described as the atheromatosis initiator. Therefore, the aim of the study was to (1) evaluate if the carotid aVV in CKD patients increases in comparison to its physiological value of healthy patients; (2) explore which traditional or nontraditional risk factor including inflammation, bone and mineral metabolism, and anemia could be related to the aVV angiogenesis. CKD patients without previous cardiovascular events (44, stages 3-4; 37, stage 5D) and 65 healthy subjects were compared. The carotid aVV and the intima-media thickness (cIMT) were evaluated by ultrasound. CKD patients at stages 3-4 showed higher aVV of the right carotid artery even after adjusting for age. Importantly, a multiple linear regression model showed hemoglobin levels > 12.5 g/dL as the factor for an estimated higher aVV of the right carotid artery. In conclusion, the association of hemoglobin with higher aVV could suggest the role of high hemoglobin in the higher incidence of adverse cardiovascular outcomes in CKD patients.


Subject(s)
Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Hemoglobins/metabolism , Renal Insufficiency, Chronic/metabolism , Vasa Vasorum/metabolism , Vasa Vasorum/pathology , Adult , Aged , C-Reactive Protein/metabolism , Cholesterol/blood , Cross-Sectional Studies , Female , Ferritins/blood , Humans , Male , Middle Aged , Regression Analysis , Renal Insufficiency, Chronic/pathology , Triglycerides/blood , Vitamin D/analogs & derivatives , Vitamin D/blood
17.
J Biomed Mater Res A ; 105(3): 814-823, 2017 03.
Article in English | MEDLINE | ID: mdl-27779830

ABSTRACT

There is considerable global demand for three-dimensional (3D) functional tissues which mimic our native organs and tissues for use as in vitro drug screening systems and in regenerative medicine. In particular, there has been an increasing number of patients who suffer from arterial diseases such as arteriosclerosis. As such, in vitro 3D arterial wall models that can evaluate the effects of novel medicines and a novel artificial graft for the treatment are required. In our previous study, we reported the rapid construction of 3D tissues by employing a layer-by-layer (LbL) technique and revealed their potential applications in the pharmaceutical fields and tissue engineering. In this study, we successfully constructed a 3D arterial wall model containing vasa vasorum by employing a LbL technique for the first time. The cells were coated with extracellular matrix nanofilms and seeded into a culture insert using a cell accumulation method. This model had a three-layered hierarchical structure: a fibroblast layer, a smooth muscle layer, and an endothelial layer, which resembled the native arterial wall. Our method could introduce vasa vasorum into a fibroblast layer in vitro and the 3D arterial wall model showed barrier function which was evaluated by immunostaining and transendothelial electrical resistance measurement. Furthermore, electron microscopy observations revealed that the vasa vasorum was composed of single-layered endothelial cells, and the endothelial tubes were surrounded by the basal lamina, which are known to promote maturation and stabilization in native blood capillaries. These models should be useful for tissue engineering, regenerative medicine, and pharmaceutical applications. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 814-823, 2017.


Subject(s)
Arteries , Arteriosclerosis , Models, Cardiovascular , Tissue Engineering , Vasa Vasorum , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Arteriosclerosis/physiopathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Membranes, Artificial , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Nanostructures , Vasa Vasorum/metabolism , Vasa Vasorum/pathology , Vasa Vasorum/physiopathology
19.
Stem Cell Res Ther ; 7(1): 114, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27526687

ABSTRACT

BACKGROUND: Proliferation of the vasa vasorum has been implicated in the pathogenesis of atherosclerosis, and the vasa vasorum is closely associated with resident stem cells within the vasculature. C-reactive protein (CRP) is positively correlated with cardiovascular disease risk, and our previous study demonstrated that it induces inflammatory reactions of perivascular adipose tissue by targeting adipocytes. METHODS: Here we investigated whether CRP affected the proliferation and proangiogenic paracrine activity of adipose-derived stem cells (ADSCs), which may contribute to vasa vasorum angiogenesis. RESULTS: We found that CRP did not affect ADSC apoptosis, cell cycle, or proliferation but did increase their migration by activating the PI3K/Akt pathway. Our results demonstrated that CRP can upregulate vascular endothelial growth factor-A (VEGF-A) expression by activating hypoxia inducible factor-1α (HIF-1α) in ADSCs, which significantly increased tube formation on Matrigel and functional vessels in the Matrigel plug angiogenesis assay. The inhibition of CRP-activated phosphorylation of ERK and Akt can suppress CRP-stimulated HIF-1α activation and VEGF-A expression. CRP can also stimulate proteolytic activity of matrix metalloproteinase-2 in ADSCs. Furthermore, CRP binds activating CD64 on ADSCs, rather than CD16/32. CONCLUSION: Our findings implicate that CRP might play a role in vasa vasorum growth by activating the proangiogenic activity of ADSCs.


Subject(s)
Adipose Tissue/metabolism , C-Reactive Protein/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Pathologic/metabolism , Signal Transduction/physiology , Stem Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adipose Tissue/pathology , Animals , Cell Proliferation/physiology , MAP Kinase Signaling System/physiology , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/pathology , Paracrine Communication/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, IgG/metabolism , Stem Cells/pathology , Up-Regulation/physiology , Vasa Vasorum/metabolism , Vasa Vasorum/pathology
20.
PLoS One ; 11(3): e0150862, 2016.
Article in English | MEDLINE | ID: mdl-27010927

ABSTRACT

OBJECTIVES: It is generally believed that low-density lipoprotein enters the vascular wall from its lumen and oxidized (oxLDL), after which it plays an important role in atherosclerosis. Because voluminous epicardial adipose tissue is a risk factor for coronary events, there is a possibility that the pericoronary adipose tissue (PCAT), which is a part of epicardial adipose tissue, acts as a risk factor by supplying oxLDL to the coronary arterial wall. The present study was performed whether PCAT stores and supplies oxLDL to the coronary wall. METHODS: Localization of oxLDL in PCAT and its relation to plaque morphology were examined by immunohistochemical techniques in 27 epicardial coronary arteries excised from 9 human autopsy cases. RESULTS: OxLDL deposited in all PCAT of the studied cases. The percent (%) incidence of oxLDL in the intima of 25 normal segment, 19 white plaques, 15 yellow plaques without necrotic core (NC) and 10 yellow plaques with NC, was 32, 84, 93 (p<0.05 vs normal segments and yellow plaques with NC), and 30, respectively. OxLDL deposited either in dotted or diffuse pattern. Double immunohistochemical staining revealed that the dotted oxLDL was that contained in CD68(+)-macrophages. The oxLDL-containing macrophages were observed in the interstitial space but not inside of the vasa vasorum, and they traversed PCAT, adventitia, external and internal elastic laminae, suggesting their migration towards the intima. Diffuse oxLDL deposits were observed in 17 preparations, the majority of which were co-localized with the vasa vasorum in outer or in both inner and outer halves of intima, and rarely in the inner half alone. CONCLUSIONS: The results suggested that PCAT is a supply source of oxLDL to coronary intima and acts as a risk factor for coronary events, that oxLDL increasingly deposits in the intima with plaque growth and decreases after plaque maturation, and therefore molecular therapies targeting the PCAT before plaque growth could be effective in preventing human coronary atherosclerosis.


Subject(s)
Adipose Tissue/pathology , Coronary Vessels/pathology , Lipoproteins, LDL/analysis , Lipoproteins, LDL/metabolism , Plaque, Atherosclerotic/pathology , Adipose Tissue/metabolism , Antigens, CD/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , Autopsy , Coronary Vessels/metabolism , Female , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Middle Aged , Plaque, Atherosclerotic/metabolism , Tunica Intima/metabolism , Tunica Intima/pathology , Vasa Vasorum/metabolism , Vasa Vasorum/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...