Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 239
Filter
1.
Am J Physiol Heart Circ Physiol ; 320(4): H1724-H1737, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33710913

ABSTRACT

The surface of vascular endothelial cells (ECs) is covered by a protective negatively charged layer known as the endothelial glycocalyx. Herein, we hypothesized its transport barrier and mechanosensory role in transmural water flux and low-density lipoprotein (LDL) transport in an isolated rat abdominal aorta perfused under 85 mmHg and 20 dyn/cm2 ex vivo. The endothelial glycocalyx was digested by hyaluronidase (HAase) from bovine tests. Water infiltration velocity (Vw) was measured by a graduated pipette. LDL coverage and mean maximum infiltration distance (MMID) in the vessel wall were quantified by confocal laser scanning microscopy. EC apoptosis was determined by the terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) technique, and leaky junction rates were evaluated by electron microscopy. The results showed that a 42% degradation of the endothelial glycocalyx by HAase treatment increased Vw, LDL coverage, and MMID. Shear stress increased Vw, which cannot be inhibited by HAase treatment. Four hour-shear application increased about fourfolds of LDL coverage, whereas exerted no significant effects on its MMID, EC apoptosis, and the leaky junctions. On the contrary, 24-h shear exposure has no significant effects on LDL coverage, whereas increased 2.74-folds of MMID and about 53% of EC apoptotic rates that could be inhibited by HAase treatment. These results suggest endothelial glycocalyx acts as a transport barrier by decreasing water and LDL transport, as well as a mechanosensor of shear to regulate EC apoptosis, thus affecting leaky junctions and regulating LDL transport into the vessel wall.NEW & NOTEWORTHY A 42% degradation of the endothelial glycocalyx by hyaluronidase of the isolated rat abdominal aorta facilitated water and LDL transport across the vessel wall, suggesting endothelial glycocalyx as a transport barrier. A 24-h shear exposure increased LDL mean maximum infiltration distance, and enhanced EC apoptosis, which could be both inhibited by hyaluronidase treatment, suggesting endothelial glycocalyx may also act as a mechanosensor of shear to regulate EC apoptosis, thus affecting leaky junctions and regulating LDL transport.


Subject(s)
Aorta, Abdominal/metabolism , Endothelial Cells/metabolism , Glycocalyx/metabolism , Lipoproteins, LDL/metabolism , Water/metabolism , Animals , Aorta, Abdominal/ultrastructure , Apoptosis , Biological Transport , Endothelial Cells/ultrastructure , Glycocalyx/ultrastructure , Hyaluronoglucosaminidase/metabolism , In Vitro Techniques , Male , Mechanotransduction, Cellular , Permeability , Rats, Sprague-Dawley , Regional Blood Flow , Stress, Mechanical
2.
J Vasc Res ; 58(3): 148-158, 2021.
Article in English | MEDLINE | ID: mdl-33601368

ABSTRACT

BACKGROUND AND OBJECTIVE: Epidemiological evidence suggests that the antidiabetic drug metformin (MET) can also inhibit abdominal aortic aneurysm (AAA) formation. However, the underlying protective mechanism remains unknown. It has been reported that phosphorylated AMP-activated protein kinase (AMPK) levels are significantly lower in AAA tissues than control aortic tissues. AMPK activation can inhibit the downstream signaling molecule called mechanistic target of rapamycin (mTOR), which has also been reported be upregulated in thoracic aneurysms. Thus, blocking mTOR signaling could attenuate AAA progression. MET is a known agonist of AMPK. Therefore, in this study, we investigated if MET could inhibit formation of AAA by activating the AMPK/mTOR signaling pathway. MATERIALS AND METHODS: The AAA animal model was induced by intraluminal porcine pancreatic elastase (PPE) perfusion in male Sprague Dawley rats. The rats were treated with MET or compound C (C.C), which is an AMPK inhibitor. AAA formation was monitored by serial ultrasound. Aortas were collected 4 weeks after surgery and subjected to immunohistochemistry, Western blot, and transmission electron microscopy analyses. RESULTS: MET treatment dramatically inhibited the formation of AAA 4 weeks after PPE perfusion. MET reduced the aortic diameter, downregulated both macrophage infiltration and matrix metalloproteinase expression, decreased neovascularization, and preserved the contractile phenotype of the aortic vascular smooth muscle cells. Furthermore, we detected an increase in autophagy after MET treatment. All of these effects were reversed by the AMPK inhibitor C.C. CONCLUSION: This study demonstrated that MET activates AMPK and suppresses AAA formation. Our study provides a novel mechanism for MET and suggests that MET could be potentially used as a therapeutic candidate for preventing AAA.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aorta, Abdominal/drug effects , Aortic Aneurysm, Abdominal/prevention & control , Metformin/pharmacology , TOR Serine-Threonine Kinases/metabolism , Vascular Remodeling/drug effects , Animals , Aorta, Abdominal/enzymology , Aorta, Abdominal/ultrastructure , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/enzymology , Aortic Aneurysm, Abdominal/pathology , Dilatation, Pathologic , Disease Models, Animal , Enzyme Activation , Macrophages/drug effects , Macrophages/metabolism , Male , Neovascularization, Pathologic , Pancreatic Elastase , Phosphorylation , Rats, Sprague-Dawley , Signal Transduction
3.
Bull Exp Biol Med ; 169(4): 525-530, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32910389

ABSTRACT

We evaluated the efficiency of an original method for studying of the microvascular bed under conditions of normal microanatomy and pathological neovascularization. The blood vessels, tissues surrounding the stent in the pulmonary artery and subcutaneously implanted titanium nickelide plate, atherosclerotic plaque, and vascular stent with restenosis were examined. The specimens were fixed in formalin and stained in OsO4, embedded into fresh epoxy resin, grinded, polished, and counterstained with uranyl acetate and lead citrate. Numerous vasa vasorum were found in all native vessels. Around the pulmonary artery stent and metal plates, numerous newly formed vessels of small diameter were seen. The intensity of neovascularization in atherosclerosis and carotid stent restenosis differed significantly. Our technique can be successfully used for evaluation of the microvascular bed.


Subject(s)
Aorta, Abdominal/ultrastructure , Microscopy, Electron, Scanning/methods , Neovascularization, Pathologic/diagnostic imaging , Plaque, Atherosclerotic/ultrastructure , Saphenous Vein/ultrastructure , Thoracic Arteries/ultrastructure , Animals , Aorta, Abdominal/anatomy & histology , Cattle , Coated Materials, Biocompatible/chemistry , Coronary Restenosis/pathology , Formaldehyde , Humans , Male , Neovascularization, Physiologic , Plaque, Atherosclerotic/pathology , Rats , Rats, Wistar , Saphenous Vein/anatomy & histology , Staining and Labeling/methods , Stents , Subcutaneous Tissue/pathology , Subcutaneous Tissue/ultrastructure , Thoracic Arteries/anatomy & histology , Tissue Fixation/methods
4.
J Vasc Res ; 56(5): 230-240, 2019.
Article in English | MEDLINE | ID: mdl-31307051

ABSTRACT

OBJECTIVE: The relationship between methionine (Met) and abdominal aortic aneurysm (AAA) has been previously demonstrated, but the mechanisms controlling this association remain unclear. This study investigated the potential contribution of hypermethioninemia (HMet) to the development of AAA. METHODS: A model of AAA was induced by intraluminal porcine pancreatic elastase (PPE) infusion in 60 male Sprague-Dawley rats divided into 4 groups (n = 15 per group). Met was supplied by intragastric administration (1 g/kg body weight/day) from 1 week before surgery until 4 weeks after surgery. The aortic diameter was measured by ultrasound. Aortas were collected 4 weeks after surgery and subjected to biochemical analysis, histological assays, and transmission electron microscopy. RESULTS: After 5 weeks of Met supplementation, HMet increased the dilation ratio of the HMet + PPE group, and hyperhomocysteinemia was also induced in HMet and HMet + PPE rats. Increased matrix metalloproteinase-2 (MMP-2), osteopontin, and interleukin-6 expression was detected in HMet + PPE rats. Furthermore, increased autophagy was detected in the HMet + PPE group. CONCLUSION: This study demonstrates that HMet may exacerbate the formation of AAA due to the increased dilation ratio partially via enhancing MMP-2 and inflammatory responses.


Subject(s)
Amino Acid Metabolism, Inborn Errors/chemically induced , Aortic Aneurysm, Abdominal/chemically induced , Glycine N-Methyltransferase/deficiency , Methionine , Amino Acid Metabolism, Inborn Errors/blood , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/ultrastructure , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Dilatation, Pathologic , Disease Models, Animal , Disease Progression , Glycine N-Methyltransferase/blood , Interleukin-6/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Osteopontin/metabolism , Pancreatic Elastase , Rats, Sprague-Dawley , Risk Factors , Time Factors
5.
Arterioscler Thromb Vasc Biol ; 38(2): 363-372, 2018 02.
Article in English | MEDLINE | ID: mdl-29217507

ABSTRACT

OBJECTIVE: Vascular calcification significantly increases morbidity in life-threatening diseases, and no treatments are available because of lack of understanding of the underlying molecular mechanism. Here, we study the physicochemical details of mineral nucleation and growth in an animal model that faithfully recapitulates medial arterial calcification in humans, to understand how pathological calcification is initiated on the vascular extracellular matrix. APPROACH AND RESULTS: MGP (matrix Gla protein) is a potent mineralization inhibitor. We study the evolution of medial calcification in MGP-deficient mice over the course of 5 weeks using a combination of material science techniques and find that mineral composition and crystallinity evolve over time and space. We show that calcium is adsorbed first and then amorphous calcium phosphate and octacalcium phosphate forms, which then transform into hydroxyapatite and carbonated apatite. These events are repeated after each nucleation event, providing a snapshot of the overall mineral evolution at each time point analyzed. CONCLUSIONS: Our results show that an interdisciplinary approach combining animal models and materials science can provide insights into the mechanism of vascular calcification and suggest the importance of analyzing mineral phases, rather than just overall mineralization extent, to diagnose and possibly prevent disease development.


Subject(s)
Aorta, Abdominal/metabolism , Aorta, Thoracic/metabolism , Aortic Diseases/metabolism , Calcium-Binding Proteins/metabolism , Calcium/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Vascular Calcification/metabolism , Animals , Aorta, Abdominal/ultrastructure , Aorta, Thoracic/ultrastructure , Aortic Diseases/genetics , Aortic Diseases/pathology , Apatites/metabolism , Calcium Phosphates/metabolism , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Crystallization , Disease Models, Animal , Disease Progression , Durapatite/metabolism , Extracellular Matrix/pathology , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Time Factors , Vascular Calcification/genetics , Vascular Calcification/pathology , Matrix Gla Protein
6.
J Vasc Res ; 55(6): 338-349, 2018.
Article in English | MEDLINE | ID: mdl-30485863

ABSTRACT

In the field of vascular graft research, poly-ε-caprolactone (PCL) is used owing to its good mechanical strength and biocompatibility. In this study, PCL scaffold was prepared by electrospinning and surface modification with heparin via hexamethylenediamine. Then the scaffolds were implanted into the infrarenal abdominal aorta of Wistar rats and contrast-enhanced micro-ultrasound was used to monitor the patency of grafts after implantation. These grafts were extracted from the rats at 1, 3, and 6 months for histological analysis, immunofluorescence staining, and scanning electron microscopy observation. Although some grafts experienced aneurysmal change, results showed that all implanted grafts were patent during the course of 6 months and these grafts demonstrated well-organized neotissue with endothelium formation, smooth muscle regeneration, and extracellular matrix formation. Such findings confirm feasibility to create heparin-conjugated scaffolds of next-generation vascular grafts.


Subject(s)
Aorta, Abdominal/surgery , Heparin/chemistry , Polyesters/chemistry , Tissue Scaffolds/chemistry , Vascular Remodeling , Animals , Anticoagulants , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/ultrastructure , Biocompatible Materials , Blood Vessel Prosthesis , Endothelium, Vascular/physiology , Extracellular Matrix/physiology , Microscopy, Electron, Scanning , Models, Animal , Muscle, Smooth, Vascular/physiology , Rats , Rats, Wistar , Regeneration , Ultrasonography , Vascular Grafting/methods
7.
J Vasc Surg ; 67(4): 1234-1246.e2, 2018 04.
Article in English | MEDLINE | ID: mdl-28899569

ABSTRACT

OBJECTIVE: Parameters other than maximum diameter that predict rupture of abdominal aortic aneurysms (AAAs) may be helpful for risk-benefit analysis in individual patients. The aim of this study was to characterize the biomechanical-structural characteristics associated with AAA walls to better identify the related mechanistic variables required for an accurate prediction of rupture risk. METHODS: Anterior AAA wall (n = 40) and intraluminal thrombus (ILT; n = 114) samples were acquired from 18 patients undergoing open surgical repair. Biomechanical characterization was performed using controlled circumferential stretching tests combined with a speckle-strain tracking technique to quantify the spatial heterogeneity in deformation and localized strains in the AAA walls containing calcification. After mechanical testing, the accompanying microstructural characteristics of the AAA wall and ILT types were examined using electron microscopy. RESULTS: No significant correlation was found between the AAA diameter and the wall mechanical properties in terms of Cauchy stress (rs = -0.139; P = .596) or stiffness (rs = -0.451; P = .069). Quantification of significant localized peak strains, which were concentrated in the tissue regions surrounding calcification, reveals that peak strains increased by a mean of 174% as a result of calcification and corresponding peak stresses by 18.2%. Four ILT types characteristic of diverse stages in the evolving tissue microstructure were directly associated with distinct mechanical stiffness properties of the ILT and underlying AAA wall. ILT types were independent of geometric factors, including ILT volume and AAA diameter measures (ILT stiffness and AAA diameter [rs = -0.511; P = .074]; ILT stiffness and ILT volume [rs = -0.245; P = .467]). CONCLUSIONS: AAA wall stiffness properties are controlled by the load-bearing capacity of the noncalcified tissue portion, and low stiffness properties represent a highly degraded vulnerable wall. The presence of calcification that is contiguous with the inner wall causes severe tissue overstretching in surrounding tissue areas. The results highlight the use of additional biomechanical measures, detailing the biomechanical-structural characteristics of AAA tissue, that may be a helpful adjunct to improve the accuracy of rupture prediction.


Subject(s)
Aorta, Abdominal/physiopathology , Aortic Aneurysm, Abdominal/complications , Aortic Rupture/etiology , Thrombosis/complications , Vascular Calcification/complications , Aged , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/surgery , Aorta, Abdominal/ultrastructure , Aortic Aneurysm, Abdominal/diagnostic imaging , Aortic Aneurysm, Abdominal/physiopathology , Aortic Aneurysm, Abdominal/surgery , Aortic Rupture/diagnostic imaging , Aortic Rupture/physiopathology , Aortography/methods , Biomechanical Phenomena , Computed Tomography Angiography , Female , Humans , Male , Microscopy, Electron, Scanning , Middle Aged , Multidetector Computed Tomography , Regional Blood Flow , Risk Assessment , Risk Factors , Spectroscopy, Fourier Transform Infrared , Thrombosis/diagnostic imaging , Thrombosis/physiopathology , Thrombosis/surgery , Vascular Calcification/diagnostic imaging , Vascular Calcification/physiopathology , Vascular Calcification/surgery
8.
J Cell Physiol ; 232(1): 7-13, 2017 01.
Article in English | MEDLINE | ID: mdl-26991605

ABSTRACT

Stiffness of intact endothelial cells (ECs) in the abdominal aorta (AA) and in the medial and lateral wall of the common iliac artery (CIA(Medial) and CIA(Lateral), respectively), which were freshly obtained from cholesterol-fed rabbits, were measured with an atomic force microscopic indentation method. In the areas away from atherosclerotic plaques (Off-plaque), ECs were significantly stiffer in CIA(Medial) than in the other two locations; this result was similar to that from normal diet-fed animals. On the other hand, there were no significant differences in the stiffness of ECs located on atherosclerotic plaques (On-plaque) among the three sites; the stiffness was equal to those in "Off-plaque" wall of CIA(Lateral) and AA. Moreover, the stiffness of ECs covering plaques decreased with the progression of atherosclerosis. The precise quantification of the stiffness of vascular ECs would provide a better understanding of cellular remodeling and adaptation in atherosclerosis. J. Cell. Physiol. 232: 7-13, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Aorta, Abdominal/ultrastructure , Atherosclerosis , Endothelial Cells/ultrastructure , Microscopy, Atomic Force , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Atherosclerosis/pathology , Cholesterol/metabolism , Disease Models, Animal , Endothelial Cells/pathology , Male , Rabbits
9.
Artif Organs ; 41(1): 98-106, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27087603

ABSTRACT

Decellularization is a proposed method of preparing nonautologous biological arterial vascular scaffolding; however, the fate of the supporting medial elastic fiber, which is important in preserving the vascular structural integrity, is uncertain. The influence of losartan on preserving the medial elastic fiber integrity in decellularized small diameter vascular conduits (SDVC) was investigated. Decellularized infrarenal abdominal aortic allografts were implanted in Sprague-Dawley rats treated either with (study rats, n = 6) or without oral losartan (control rats, n = 6) and graded 8 weeks later according to a remodeling scoring system (1-mild, 2-moderate, 3-severe) which we devised based on the intimal hyperplasia degree, morphologic changes, and elastic fiber fragmentation of the conduits. DAPI immunohistochemistry analysis was performed in 47 (25 decellularization only and 22 losartan treatment) cross-sectional slide specimens. The losartan versus decellularization only SDVC showed a significantly lower medial elastic fragmentation score (1.32 vs. 2.24, P < 0.001), superior medial layer preservation, and relatively more normal appearing intimal cellular morphology. The results suggested rats receiving decellularized SDVCs treated with losartan may yield superior medial layer elastic fiber preservation.


Subject(s)
Antihypertensive Agents/pharmacology , Aorta, Abdominal/drug effects , Aorta, Abdominal/transplantation , Bioprosthesis , Blood Vessel Prosthesis , Losartan/pharmacology , Allografts , Animals , Aorta, Abdominal/ultrastructure , Biomechanical Phenomena/drug effects , Elasticity/drug effects , Female , Rats, Sprague-Dawley , Tissue Scaffolds/chemistry , Transplantation, Homologous
10.
J Artif Organs ; 20(3): 221-229, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28500497

ABSTRACT

In order to develop small-diameter vascular grafts, it is necessary to evaluate endothelialization, especially, in the center part at early stage. For implantation of vascular grafts of 1 cm in length to abdominal aortae of rat, endothelial cells can be formed easily by stretching anastomosis. We evaluated the endothelialization in the center part of vascular grafts by implanting vascular grafts using transgenic (TG) silk fibroin (SF) of 3 cm in length. Vascular grafts were prepared 1.5 mm in diameter and 1 and 3 cm in length using wild type (WT) SF and TG SF by braiding structure, respectively. The grafts were removed after 2 weeks or 3 months and evaluated pathologically. Endothelialization was not confirmed totally after 3 months of implantation. However, endothelialization in the center part of grafts was significantly higher in TG SF than in WT SF. No significant difference was found regarding tissue infiltration and internal diameter. The TG SF revealed migration of the endothelial cells into the center part of the vessels at the early stage. Also, tissue infiltration and remodeling is expected using SF. The 3 cm length vascular grafts can be evaluated as a new experimental system.


Subject(s)
Aorta, Abdominal/surgery , Bioprosthesis , Blood Vessel Prosthesis , Endothelium, Vascular/ultrastructure , Vascular Diseases/surgery , Vascular Grafting/methods , Animals , Aorta, Abdominal/ultrastructure , Disease Models, Animal , Female , Microscopy, Electron, Scanning , Rats , Rats, Sprague-Dawley , Vascular Diseases/pathology
11.
J Vasc Res ; 53(1-2): 49-57, 2016.
Article in English | MEDLINE | ID: mdl-27532120

ABSTRACT

Topical application of elastase to induce arterial aneurysm formation is an emerging murine model of vascular disease. In the context of aortic abdominal aneurysm (AAA), angiotensin II infusion and porcine pancreatic elastase perfusion models are commonly used today. This study, therefore, compares matrix remodeling, inflammation, and angiogenesis as distinct features of aneurysms in two models treated with intra-/extraluminal elastase. C57BL/6 mice underwent intra-/extraluminal elastase application via laparotomy and were followed up for 4 weeks. Basic histology and immunohistochemistry were performed at different time points along with transmission electron microscopy, PCR analysis, TUNEL assays, and blood analysis. Both models did not differ in aneurysm growth rate, but they showed distinct features and results depending on the way of elastase application. Extraluminal aneurysm induction preserved endothelial cell function and elastic fibers but showed ongoing acute inflammation, mainly in the adventitia. The destruction of elastic layers followed by chronic inflammation was a characteristic of intraluminal elastase perfusion, as well as medial angiogenesis, a key feature in human AAA. Different animal models harbor different features of human AAA and must, therefore, be chosen wisely. External elastase application mimics an acute inflammatory aneurysm, whereas intraluminal elastase perfusion shows chronic inflammation with angiogenesis and endothelial destruction, thus better mimicking human disease.


Subject(s)
Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Elastic Tissue/pathology , Pancreatic Elastase , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/ultrastructure , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Disease Models, Animal , Disease Progression , Elastic Tissue/metabolism , Elastic Tissue/ultrastructure , Endothelial Cells/pathology , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Male , Mice, Inbred C57BL , Neovascularization, Pathologic , Phenotype , Proteolysis , Time Factors
12.
J Vasc Surg ; 63(5): 1341-50, 2016 05.
Article in English | MEDLINE | ID: mdl-25701495

ABSTRACT

OBJECTIVE: Adventitial collagen structure provides the aorta with tensile strength. Like other collagen-rich tissues, it can be affected by internal factors including aging and location. We determined whether the structural characteristics of human aortic adventitial collagen change with aging, location, and aneurysm formation. METHODS: Nonatherosclerotic nonaneurysmal (NANA) human abdominal aortas were collected from 15 individuals who had died of noncardiovascular diseases (<40 years old, NANA young, n = 5; >60 years old, NANA old, n = 5). The architecture of adventitial collagen in the aortas was assessed by scanning electron microscopy, and fiber orientation was assessed by polarized microscopy with two-dimensional fast Fourier transform. We then analyzed retardation as an anisotropic property of adventitial collagen by polarized light microscopy. The orientation and retardation of NANA aortas were compared with those of abdominal aortic specimens from patients who were surgically treated for abdominal aortic aneurysm (AAA) (>60 years old, n = 11). RESULTS: Adventitial collagen of the abdominal aortas on scanning electron microscopy images appeared as wavy, ropy fibers in aortas from young individuals (NANA young, n = 5) and were essentially flattened in those from older patents (NANA old, n = 5) and from those with AAA. Collagen fibers were thicker but sparser in the adventitia of aortas with AAA. Orientation maintained in the collagen fibers of NANA aortas (n = 15) on two-dimensional fast Fourier transform analysis was unrelated to either location or age and did not differ between NANA aortas and those with AAA. However, collagen fibrils in NANA aortas (n = 15) were significantly less retarded only at the level of the inferior mesenteric artery compared with other aortic locations. In addition, retardation was significantly reduced in abdominal aortas with AAA at the level of the inferior mesenteric artery. CONCLUSIONS: The basic structure of adventitial collagen fiber was maintained in abdominal aortas regardless of location or age. Because the molecular structure at the subfibril level changed at abdominal aorta and enhanced in aortas with AAA, alterations in the molecular structure of adventitial collagen might be associated with aneurysmal formation.


Subject(s)
Adventitia/ultrastructure , Aging/pathology , Aorta, Abdominal/ultrastructure , Aortic Aneurysm, Abdominal/pathology , Fibrillar Collagens/ultrastructure , Adult , Age Factors , Aged , Aged, 80 and over , Aortic Aneurysm, Abdominal/etiology , Case-Control Studies , Humans , Male , Microscopy, Electron, Scanning , Microscopy, Polarization , Middle Aged , Protein Conformation , Risk Factors , Tensile Strength
13.
J Nanosci Nanotechnol ; 16(1): 885-91, 2016 Jan.
Article in English | MEDLINE | ID: mdl-27398541

ABSTRACT

Learning from nature concerning how nanostructured surfaces interact with liquids may provide insight into better understanding of inside living biological interfaces bearing these nanostructures and further development of innovative materials contacting water. Here we investigate the dynamic behaviour of water droplet interacting with one-dimensional nano-wrinkles of different size on polydimethylsiloxane (PDMS) surface. The structure design of the variationally one-dimensional nano-wrinkles is inspired by in vivo responding topographic changes in aortic intima, which was characterized with liquid-phase atomic force microscopy. We show here that increasing the amplitude of the wrinkles promotes the spreading and energy dissipation of liquid droplets on the wrinkled interfaces. This result suggests a possible bio-protection mechanism of blood vessels via its structural changes on the aortic intima against elevated flowing blood, and provides a basis for tuning interfacial nanostructure of optimal durability against wearing by the liquid behaviors.


Subject(s)
Aorta, Abdominal/chemistry , Dimethylpolysiloxanes/chemistry , Nanostructures/chemistry , Nylons/chemistry , Tunica Intima/chemistry , Water/chemistry , Animals , Aorta, Abdominal/ultrastructure , Microscopy, Atomic Force/methods , Nanostructures/ultrastructure , Phase Transition , Rats , Rats, Wistar , Tunica Intima/ultrastructure
14.
Morfologiia ; 147(1): 21-6, 2015.
Article in Russian | MEDLINE | ID: mdl-25958724

ABSTRACT

The peculiarities of the structure, skeletotopy, and syntopy of the lumbar lymphatic collector were studied on 20 5-8 week-old embryos and on 80 9-36 week-old fetuses using a complex macro-microscopic method. It is found that the lumbar lymphatic collector in fetuses at 9-10 weeks was represented by retroperitoneal and retroaortic lymphatic sacs that had a fusion mode of formation and were interconnected. Retroperitoneal sac was located in the projection of L(I)-L(IV) and was in contact with the anterior surface of the abdominal aorta and inferior vena cava, aortic lumbar paraganglia, abdominal aortic plexus and ganglia of sympathetic trunk. Retroaortic sack at L(I)-L(II) was adjacent to posterior surface of the aorta, the lumbar vertebrae and the medial crus of the diaphragm. These topical relations were preserved throughout the whole fetal period. However, in fetuses of 11-13 weeks lymphatic sacs formed the lymphatic plexuses, while in fetuses of 14-36 weeks they formed lumbar lymph nodes and their interconnecting vessels.


Subject(s)
Lumbar Vertebrae/ultrastructure , Lumbosacral Region , Lymph Nodes/ultrastructure , Retroperitoneal Space/growth & development , Aorta, Abdominal/growth & development , Aorta, Abdominal/ultrastructure , Embryonic Development , Fetus/ultrastructure , Humans , Lumbar Vertebrae/growth & development , Lymph Nodes/growth & development
15.
Arterioscler Thromb Vasc Biol ; 32(12): 2901-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23042818

ABSTRACT

OBJECTIVE: The purpose of this study was to evaluate potential mechanisms promoting abdominal aortic aneurysm development with tobacco smoke (TS) exposure. METHODS AND RESULTS: Experiments used the elastase perfusion model of abdominal aortic aneurysms with smoke-free controls. The effect of TS exposure was evaluated in C57/Bl6 mice, after broad-spectrum matrix metalloproteinase inhibition with doxycycline and in mice deficient in matrix metalloproteinase-9, matrix metalloproteinase-12, Cathepsin-S, and Neutrophil Elastase. Preparations of washed marrow, spleen, and peripheral blood leukocytes were transferred to smoke-free mice from 6-week TS-exposed mice or smoke-free mice. All mice were euthanized 14 days after elastase perfusion, and the percentage of change in aortic diameter (%Δ aortic diameter) was calculated. Electron microscopy of aortic tissue from animals exposed to TS without elastase exposure did not demonstrate any ultrastructural changes. Neither doxycycline nor any specific elastase deficiency was effective at preventing an increase in %Δ aortic diameter in TS-exposed animals. Smoke exposure for 6 weeks increased the %Δ aortic diameter after a smoke-free interval of up to 6 weeks before elastase perfusion. Leukocyte preparations from TS-exposed mice localized to abdominal aortic aneurysms and increased the %Δ aortic diameter in smoke-free mice. CONCLUSIONS: The effect of TS on the development of abdominal aortic aneurysms is not dependent on the activity of elastolytic enzymes and persists for long periods despite cessation of TS. Alterations in leukocyte response to aortic injury appear to mediate this effect.


Subject(s)
Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/physiopathology , Leukocytes, Mononuclear/pathology , Leukocytes, Mononuclear/physiology , Smoking/adverse effects , Animals , Aorta, Abdominal/pathology , Aorta, Abdominal/physiopathology , Aorta, Abdominal/ultrastructure , Aortic Aneurysm, Abdominal/pathology , Cathepsins/deficiency , Cathepsins/genetics , Cathepsins/physiology , Cell Count , Disease Models, Animal , Doxycycline/pharmacology , Leukocyte Elastase/deficiency , Leukocyte Elastase/genetics , Leukocyte Elastase/physiology , Male , Matrix Metalloproteinase 12/deficiency , Matrix Metalloproteinase 12/genetics , Matrix Metalloproteinase 12/physiology , Matrix Metalloproteinase 9/deficiency , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/physiology , Matrix Metalloproteinases/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Artif Organs ; 36(1): 54-61, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21848935

ABSTRACT

No small-diameter synthetic graft has yet shown comparable performance to autologous vessels. Synthetic conduits fail due to their inherent surface thrombogenicity and the development of intimal hyperplasia. In addressing these shortcomings, electrospinning offers an interesting alternative to other nanostructured, cardiovascular substitutes because of the close match of electrospun materials to the biomechanical and structural properties of native vessels. In this study, we investigated the in vivo behavior of electrospun, small-diameter conduits in a rat model. Vascular grafts composed of polyurethane were fabricated by electrospinning. Prostheses were implanted into the abdominal aorta in 40 rats for either 7 days, 4 weeks, 3 months, or 6 months. Retrieved specimens were evaluated by histology, immunohistochemical staining, confocal laser scanning microscopy, and scanning electron microscopy. At all time points, we found no evidence of foreign body reaction or graft degradation. The overall patency rate of the intravascular implants was 95%. Within 7 days, grafts revealed ingrowth of host cells. CD34+ cells increased significantly from 7 days up to 6 months of implantation (P < 0.05). Myofibroblasts and myocytes showed increasing cell numbers up to 3 months (P < 0.05). Ki67 staining indicated unaltered cell proliferation during the whole follow-up period. Besides biomechanical benefits, electrospun polyurethane grafts exhibit excellent biocompatibility in vivo. Cell immigration and differentiation seems to be promoted by the nanostructured artificial matrix.


Subject(s)
Biocompatible Materials , Blood Vessel Prosthesis , Polyurethanes , Vascular Grafting/instrumentation , Animals , Aorta, Abdominal/pathology , Aorta, Abdominal/surgery , Aorta, Abdominal/ultrastructure , Cell Culture Techniques , Cell Differentiation , Cell Movement , Cell Proliferation , Male , Models, Animal , Myocytes, Smooth Muscle/pathology , Myofibroblasts/pathology , Nanotechnology , Rats , Rats, Sprague-Dawley , Tissue Scaffolds , Vascular Grafting/methods , Vascular Patency
17.
Postepy Hig Med Dosw (Online) ; 66: 469-74, 2012 Jul 02.
Article in English | MEDLINE | ID: mdl-22922147

ABSTRACT

INTRODUCTION: The experiment was designed in order to determine the immunological processes that occur during the healing in synthetic vascular grafts, especially to establish the differences in the location of the complement system proteins between the proximal and distal anastomosis and the differences in the arrangement of inflammatory cells in those anastomoses. The understanding of those processes will provide a true basis for determining risk factors for complications after arterial repair procedures. MATERIAL/METHODS: The experiment was carried out on 16 dogs that underwent implantation of unilateral aorto-femoral bypass with expanded polytetrafluoroethylene (ePTFE). After 6 months all animals were euthanized to dissect the vascular grafts. Immunohistochemical assays and electron microscopic examinations were performed. RESULTS: Immunohistochemical findings in the structure of neointima between anastomoses of vascular prostheses demonstrated significant differences between humoral and cellular responses. The area of proximal anastomosis revealed the presence of fibroblasts, but no macrophages were detected. The histological structure of the proximal anastomosis indicates that inflammatory processes were ended during the prosthesis healing. The immunological response obtained in the distal anastomosis corresponded to the chronic inflammatory reaction with the presence of macrophages, myofibroblasts and deposits of complement C3. DISCUSSION: The identification of differences in the presence of macrophages and myofibroblasts and the presence of the C3 component between the anastomoses is the original achievement of the present study. In the available literature, no such significant differences have been shown so far in the humoral and cellular immune response caused by the presence of an artificial vessel in the arterial system.


Subject(s)
Blood Vessel Prosthesis , Complement C3/immunology , Complement System Proteins/immunology , Macrophages/immunology , Neointima/immunology , Neointima/pathology , Wound Healing/immunology , Anastomosis, Surgical , Animals , Aorta, Abdominal/immunology , Aorta, Abdominal/surgery , Aorta, Abdominal/ultrastructure , Biocompatible Materials , Blood Vessel Prosthesis/adverse effects , Collagen/immunology , Complement Activation/immunology , Dogs , Femoral Artery/immunology , Femoral Artery/surgery , Immunity, Cellular , Immunity, Humoral , Inflammation/immunology , Myofibroblasts/immunology , Polytetrafluoroethylene
18.
J Vasc Res ; 48(2): 119-29, 2011.
Article in English | MEDLINE | ID: mdl-20926892

ABSTRACT

AIMS: The Williams-Beuren syndrome (WBS) is a genetic disorder caused by a heterozygous ~1.5-Mb deletion. The aim of this study was to determine how the genetic changes in a Wbs mouse model alter Eln expression, blood pressure, vessel structure, and abdominal aortic wall dynamics in vivo. METHODS: Elastin (ELN) transcript levels were quantified by qRT-PCR and blood pressure was measured with a tail cuff system. M-mode ultrasound was used to track pulsatile abdominal aortic wall motion. Aortas were sectioned and stained to determine medial lamellar structure. RESULTS: ELN transcript levels were reduced by 38-41% in Wbs mice lacking one copy of the ELN gene. These mice also had a 10-20% increase in mean blood pressure and significantly reduced circumferential cyclic strain (p < 0.001). Finally, histological sections showed disorganized and fragmented elastin sheets in Wbs mice, but not the characteristic increase in lamellar units seen in Eln(+/-) mice. CONCLUSIONS: The deletion of Eln in this Wbs mouse model results in lower gene expression, hypertension, reduced cyclic strain, and fragmented elastin sheets. The observation that the number of medial lamellar units is normal in Wbs deletion mice, which is in contrast to Eln(+/-) mice, suggests other genes may be involved in vascular development.


Subject(s)
Blood Pressure/genetics , Chromosome Deletion , Elastin/genetics , Elastin/metabolism , Williams Syndrome/genetics , Animals , Aorta, Abdominal/diagnostic imaging , Aorta, Abdominal/ultrastructure , Cardiovascular Abnormalities/genetics , Cardiovascular Abnormalities/metabolism , Disease Models, Animal , Elasticity , Female , Gene Expression , Male , Mice , Mice, Inbred C57BL , Phenotype , Ultrasonography , Williams Syndrome/metabolism , Williams Syndrome/physiopathology
19.
Eur J Vasc Endovasc Surg ; 40(2): 202-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20547463

ABSTRACT

OBJECTIVE: The aim of study was to assess how the ultrastructure of the wall of aortic aneurysms, sac and neck influences aortic wall distensibility and proximal dilatation 2 years after open repair. METHODS: Biopsies for electron microscopy were taken from aneurysmal sac and neck of 30 patients. Patients were assessed by computed tomography (CT) and ultrasound for aneurysm diameter and distensibility (M-mode ultrasonography). RESULTS: Postoperative CT of the aortic stump distinguished two groups. Group I (n = 11) with little enlargement, median 1 mm (1-3 mm) and group II (n = 19) with significant aortic enlargement, median 5.2 mm (4-12 mm). In group II, changes in elastic fibres in the aneurysm neck were comparable to, but as extreme as in the aneurysm sac. For group I, the distensibility of the aneurysmal sac was significantly lower than in the neck or at the renal arteries. For group II, the distensibility in both the neck and sac was significantly lower than at the juxtarenal segment (p = 0.01). The biopsies of group II patients showed the extensive degeneration of normal architecture, which was associated with altered wall distensibility in both the aneurysmal neck and sac. CONCLUSIONS: Disorganisation and destruction of normal aortic architecture at the ultrastructural level are associated with decreasing aortic distensibility. Low aortic neck distensibility is associated with proximal aortic dilatation at 2 years postoperatively.


Subject(s)
Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/surgery , Aged , Aged, 80 and over , Aorta, Abdominal/ultrastructure , Aortic Aneurysm, Abdominal/physiopathology , Elasticity , Endothelium, Vascular/pathology , Endothelium, Vascular/ultrastructure , Humans , Male , Middle Aged , Vascular Patency/physiology
20.
J Am Heart Assoc ; 9(8): e014757, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32308093

ABSTRACT

Background The protective effects of polyamines on cardiovascular disease have been demonstrated in many studies. However, the roles of spermidine, a natural polyamine, in abdominal aortic aneurysm (AAA) disease have not been studied. In this study, we investigated the influence and potential mechanisms of spermidine treatment on experimental AAA disease. Methods and Results Experimental AAAs were induced in 8- to 10-week-old male C57BL/6J mice by transient intra-aortic infusion of porcine pancreatic elastase. Spermidine was administered via drinking water at a concentration of 3 mmol/L. Spermidine treatment prevented experimental AAA formation with preservation of medial elastin and smooth muscle cells. In immunostaining, macrophages, T cells, neutrophils, and neovessels were significantly reduced in aorta of spermidine-treated, as compared with vehicle-treated elastase-infused mice. Additionally, flow cytometric analysis showed that spermidine treatment reduced aortic leukocyte infiltration and circulating inflammatory cells. Furthermore, we demonstrated that spermidine treatment promoted autophagy-related proteins in experimental AAAs using Western blot analysis, immunostaining, and transmission electron microscopic examination. Autophagic function was evaluated for human abdominal aneurysmal and nonaneurysmal adjacent aortae from AAA patients using Western blot analysis and immunohistochemistry. Dysregulated autophagic function, as evidenced by increased SQSTM1/p62 protein and phosphorylated mTOR, was found in aneurysmal, as compared with nonaneurysmal, aortic segments. Conclusions Our results suggest that spermidine supplementation limits experimental AAA formation associated with preserved aortic structural integrity, attenuated aortic inflammatory infiltration, reduced circulating inflammatory monocytes, and increased autophagy-related proteins. These findings suggest that spermidine may be a promising treatment for AAA disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Aorta, Abdominal/drug effects , Aortic Aneurysm, Abdominal/prevention & control , Spermidine/pharmacology , Vascular Remodeling/drug effects , Aged , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/ultrastructure , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Autophagy/drug effects , Autophagy-Related Proteins/metabolism , Chemotaxis, Leukocyte/drug effects , Dilatation, Pathologic , Disease Models, Animal , Female , Humans , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Middle Aged , Pancreatic Elastase , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL