Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
J Cell Physiol ; 235(12): 9876-9883, 2020 12.
Article in English | MEDLINE | ID: mdl-32437059

ABSTRACT

Quercetin, an antioxidant derived from plants, can play a beneficial role in the protection of various tissues against ischemia-reperfusion injuries (IRI). The purpose of the present research was to investigate the protective effects of quercetin on gastrocnemius muscle ischemia-reperfusion. A total of 80 adult male Wistar rats (weights: 250-300 g) were divided into ten groups (n = 8 per group). We used silk 6.0 surgical thread to create a knit to occlude the femoral artery and vein for 3 hr. The treated groups, which comprised half of each experimental group, received intraperitoneal injections of 150 mg/kg quercetin after the ischemia. Blood flow was subsequently reestablished in the reperfusion phase. The rats were kept in reperfusion for 3, 7, 14, or 28 days after which they were killed with high doses of anesthetic drugs, and the gastrocnemius muscles were removed and fixed. Tissue processing, hematoxylin and eosin and toluidine blue staining, and immunohistochemistry were used to assess tumor necrosis factor-α (TNF-α) and nuclear factor κB (NF-κB) levels. A comparison between treated and untreated ischemic sites showed that on the third day of reperfusion, the severity of edema and NF-κB level decreased significantly; on the 7th day of reperfusion, the severity of edema and the levels of TNF-α and NF-κB decreased significantly; and on the 14th day of reperfusion, all of the parameters showed significant decreases. On the 28th day of reperfusion, there were significantly decreased levels of TNF-α and NF-κB, and decreased mast cell infiltration when compared with the untreated groups. According to the results, administration of quercetin after ischemia could significantly prevent gastrocnemius muscle IRI.


Subject(s)
Femoral Artery/drug effects , Muscle, Skeletal/drug effects , Quercetin/pharmacology , Reperfusion Injury/drug therapy , Animals , Antioxidants/pharmacology , Disease Models, Animal , Femoral Artery/growth & development , Femoral Artery/pathology , Humans , Muscle, Skeletal/pathology , NF-kappa B/genetics , Rats , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Tumor Necrosis Factor-alpha/genetics
2.
Int J Mol Sci ; 20(14)2019 Jul 15.
Article in English | MEDLINE | ID: mdl-31311132

ABSTRACT

Cardiovascular risk associated with fetal growth restriction (FGR) could result from an early impaired vascular function. However, whether this effect results in premature vascular aging has not been addressed. We studied the ex vivo reactivity of carotid and femoral arteries in fetal (near term), adults (eight months-old) and aged (16 months-old) guinea pigs in normal (control) and FGR offspring. Additionally, an epigenetic marker of vascular aging (i.e., LINE-1 DNA methylation) was evaluated in human umbilical artery endothelial cells (HUAEC) from control and FGR subjects. Control guinea pig arteries showed an increased contractile response (KCl-induced) and a progressive impairment of NO-mediated relaxing responses as animals get older. FGR was associated with an initial preserved carotid artery reactivity as well as a later significant impairment in NO-mediated responses. Femoral arteries from FGR fetuses showed an increased contractility but a decreased relaxing response compared with control fetuses, and both responses were impaired in FGR-adults. Finally, FGR-HUAEC showed decreased LINE-1 DNA methylation compared with control-HUAEC. These data suggest that the aging of vascular function occurs by changes in NO-mediated responses, with limited alterations in contractile capacity. Further, these effects are accelerated and imposed at early stages of development in subjects exposed to a suboptimal intrauterine environment.


Subject(s)
Aging/pathology , Endothelium, Vascular/growth & development , Fetal Growth Retardation/pathology , Animals , Carotid Arteries/growth & development , Carotid Arteries/pathology , Carotid Arteries/physiopathology , Cells, Cultured , DNA Methylation , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Femoral Artery/growth & development , Femoral Artery/pathology , Femoral Artery/physiopathology , Fetal Growth Retardation/genetics , Guinea Pigs , Humans , Long Interspersed Nucleotide Elements/genetics , Nitric Oxide/metabolism , Vasoconstriction , Vasodilation
3.
Int J Mol Sci ; 20(15)2019 Jul 24.
Article in English | MEDLINE | ID: mdl-31344780

ABSTRACT

Collaterals are unique blood vessels present in the microcirculation of most tissues that, by cross-connecting a small fraction of the outer branches of adjacent arterial trees, provide alternate routes of perfusion. However, collaterals are especially susceptible to rarefaction caused by aging, other vascular risk factors, and mouse models of Alzheimer's disease-a vulnerability attributed to the disturbed hemodynamic environment in the watershed regions where they reside. We examined the hypothesis that endothelial and smooth muscle cells (ECs and SMCs, respectively) of collaterals have specializations, distinct from those of similarly-sized nearby distal-most arterioles (DMAs) that maintain collateral integrity despite their continuous exposure to low and oscillatory/disturbed shear stress, high wall stress, and low blood oxygen. Examination of mouse brain revealed the following: Unlike the pro-inflammatory cobble-stoned morphology of ECs exposed to low/oscillatory shear stress elsewhere in the vasculature, collateral ECs are aligned with the vessel axis. Primary cilia, which sense shear stress, are present, unexpectedly, on ECs of collaterals and DMAs but are less abundant on collaterals. Unlike DMAs, collaterals are continuously invested with SMCs, have increased expression of Pycard, Ki67, Pdgfb, Angpt2, Dll4, Ephrinb2, and eNOS, and maintain expression of Klf2/4. Collaterals lack tortuosity when first formed during development, but tortuosity becomes evident within days after birth, progresses through middle age, and then declines-results consistent with the concept that collateral wall cells have a higher turnover rate than DMAs that favors proliferative senescence and collateral rarefaction. In conclusion, endothelial and SMCs of collaterals have morphologic and functional differences from those of nearby similarly sized arterioles. Future studies are required to determine if they represent specializations that counterbalance the disturbed hemodynamic, pro-inflammatory, and pro-proliferative environment in which collaterals reside and thus mitigate their risk factor-induced rarefaction.


Subject(s)
Blood Vessels/metabolism , Collateral Circulation/genetics , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic/genetics , Aging/genetics , Aging/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Blood Vessels/pathology , Collateral Circulation/physiology , Endothelial Cells/metabolism , Femoral Artery/growth & development , Femoral Artery/metabolism , Hindlimb/blood supply , Humans , Mice , Risk Factors , Signal Transduction
4.
Int J Mol Sci ; 20(13)2019 Jul 05.
Article in English | MEDLINE | ID: mdl-31284541

ABSTRACT

The process of arteriogenesis is severely compromised in patients with diabetes mellitus (DM). Earlier studies have reported the importance of Egr-1 in promoting collateral outward remodeling. However, the role of Egr-1 in the presence of DM in outward vessel remodeling was not studied. We hypothesized that Egr-1 expression may be compromised in DM which may lead to impaired collateral vessel growth. Here, we investigated the relevance of the transcription factor Egr-1 for the process of collateral artery growth in diabetic mice. Induction of arteriogenesis by femoral artery ligation resulted in an increased expression of Egr-1 on mRNA and protein level but was severely compromised in streptozotocin-induced diabetic mice. Diabetes mellitus mice showed a significantly reduced expression of Egr-1 endothelial downstream genes Intercellular Adhesion Molecule-1 (ICAM-1) and urokinase Plasminogen Activator (uPA), relevant for extravasation of leukocytes which promote arteriogenesis. Fluorescent-activated cell sorting analyses confirmed reduced leukocyte recruitment. Diabetes mellitus mice showed a reduced expression of the proliferation marker Ki-67 in growing collaterals whose luminal diameters were also reduced. The Splicing Factor-1 (SF-1), which is critical for smooth muscle cell proliferation and phenotype switch, was found to be elevated in collaterals of DM mice. Treatment of DM mice with insulin normalized the expression of Egr-1 and its downstream targets and restored leukocyte recruitment. SF-1 expression and the diameter of growing collaterals were normalized by insulin treatment as well. In summary, our results showed that Egr-1 signaling was impaired in DM mice; however, it can be rescued by insulin treatment.


Subject(s)
Diabetes Mellitus, Experimental/pathology , Early Growth Response Protein 1/metabolism , Femoral Artery/growth & development , Insulins/pharmacology , Morphogenesis/drug effects , Signal Transduction , Up-Regulation/drug effects , Animals , Antigens, CD/metabolism , Collateral Circulation/drug effects , Diabetes Mellitus, Experimental/genetics , Femoral Artery/drug effects , Gene Expression Regulation/drug effects , Leukocytes/drug effects , Leukocytes/metabolism , Leukocytes/pathology , Male , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects
5.
Proc Natl Acad Sci U S A ; 111(16): 6010-5, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24711414

ABSTRACT

Large soft tissue defects involve significant tissue loss, requiring surgical reconstruction. Autologous flaps are occasionally scant, demand prolonged transfer surgery, and induce donor site morbidity. The present work set out to fabricate an engineered muscle flap bearing its own functional vascular pedicle for repair of a large soft tissue defect in mice. Full-thickness abdominal wall defect was reconstructed using this engineered vascular muscle flap. A 3D engineered tissue constructed of a porous, biodegradable polymer scaffold embedded with endothelial cells, fibroblasts, and/or myoblasts was cultured in vitro and then implanted around the femoral artery and veins before being transferred, as an axial flap, with its vascular pedicle to reconstruct a full-thickness abdominal wall defect in the same mouse. Within 1 wk of implantation, scaffolds showed extensive functional vascular density and perfusion and anastomosis with host vessels. At 1 wk posttransfer, the engineered muscle flaps were highly vascularized, were well-integrated within the surrounding tissue, and featured sufficient mechanical strength to support the abdominal viscera. Thus, the described engineered muscle flap, equipped with an autologous vascular pedicle, constitutes an effective tool for reconstruction of large defects, thereby circumventing the need for both harvesting autologous flaps and postoperative scarification.


Subject(s)
Abdominal Wall/pathology , Abdominal Wall/surgery , Muscles/surgery , Plastic Surgery Procedures , Surgical Flaps/surgery , Tissue Engineering/methods , Animals , Biomechanical Phenomena , Dextrans/metabolism , Erythrocytes/metabolism , Femoral Artery/growth & development , Fibroblasts/cytology , Fibroblasts/transplantation , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Implants, Experimental , Mice , Myoblasts/cytology , Myoblasts/transplantation , Neovascularization, Physiologic , Perfusion , Surgical Flaps/blood supply , Ultrasonics
6.
Cytotherapy ; 18(2): 219-28, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26740280

ABSTRACT

BACKGROUND AIMS: Existing treatments have limited success in modifying the course of peripheral artery disease, which may eventually lead to limb-threatening ulcers and amputation. Cellular therapies have the potential to provide a new treatment option for this condition, but isolation of cells by conventional means has limitations with respect to reproducibility and scalability. METHODS: Induced pluripotent stem cells (iPSCs) were differentiated into precursor cells known as mesenchymoangioblasts (MCAs) and subsequently into mesenchymal stromal cells (MSCs). Hindlimb ischemia in mice was created by ligating both the iliac and femoral arteries of one hindlimb. Immediately after surgery, each animal received intramuscular injections of 5 × 10(6) cells or media in the ischemic limb. Toe necrosis was assessed visually, and hindlimb blood flow was measured by laser Doppler using a set region of interest (ROI) and by tracing the entire foot. Myofiber heterogeneity, nuclear centralization, fatty degeneration, fibrosis and capillary angiogenesis in the gastrocnemius muscle were assessed histologically. RESULTS: Blood flow in the MCA-derived MSC-treated animals was higher at each day (P <0.006), and these mice recovered faster than control animals (3.6 vs. 2.5 for set ROI; 7.5 vs. 4.1 foot tracing; slope; P <0.001). There was significantly less myofiber heterogeneity, nuclear centralization, fatty degeneration and fibrosis in MCA-derived MSC-treated animals, indicating less tissue damage. DISCUSSION: MCA-derived MSCs improved limb blood flow, reduced necrosis and maintained muscle mass and gross muscle appearance. We conclude that MCA-derived MSCs have a significant and protective effect against ischemic insults.


Subject(s)
Ischemia/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/physiology , Peripheral Arterial Disease/therapy , Regional Blood Flow/physiology , Animals , Cell Differentiation , Femoral Artery/growth & development , Femoral Artery/pathology , Hindlimb/blood supply , Hindlimb/injuries , Iliac Artery/growth & development , Iliac Artery/pathology , Induced Pluripotent Stem Cells/cytology , Mice , Muscle, Skeletal/injuries , Necrosis/pathology , Reproducibility of Results
7.
Mol Med ; 21: 605-15, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26181630

ABSTRACT

Toll-like receptors (TLRs) play an important role in regulating muscle regeneration and angiogenesis in response to ischemia. TLR2 knockout mice exhibit pronounced skeletal muscle necrosis and abnormal vessel architecture after femoral artery ligation, suggesting that TLR2 signaling is protective during ischemia. TLR4, an important receptor in inflammatory signaling, has been shown to regulate TLR2 expression in other systems. We hypothesize that a similar relationship between TLR4 and TLR2 may exist in hindlimb ischemia in which TLR4 upregulates TLR2, a mediator of angiogenesis and perfusion recovery. We examined the expression of TLR2 in unstimulated and in TLR-agonist treated endothelial cells (ECs). TLR2 expression (low in control ECs) was upregulated by lipopolysaccharide, the danger signal high mobility group box-1, and hypoxia in a TLR4-dependent manner. Endothelial tube formation on Matrigel as well as EC permeability was assessed as in vitro measures of angiogenesis. Time-lapse imaging demonstrated that ECs lacking TLR4 formed more tubes, whereas TLR2 knockdown ECs exhibited attenuated tube formation. TLR2 also mediated EC permeability, an initial step during angiogenesis, in response to high-mobility group box-1 (HMGB1) that is released by cells during hypoxic injury. In vivo, ischemia-induced upregulation of TLR2 required intact TLR4 signaling that mediated systemic inflammation, as measured by local and systemic IL-6 levels. Similar to our in vitro findings, vascular density and limb perfusion were both enhanced in the absence of TLR4 signaling, but not if TLR2 was deleted. These findings indicate that TLR2, in the absence of TLR4, improves angiogenesis and perfusion recovery in response to ischemia.


Subject(s)
Inflammation/genetics , Ischemia/genetics , Neovascularization, Physiologic/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Animals , Endothelial Cells/metabolism , Endothelial Cells/pathology , Femoral Artery/growth & development , Femoral Artery/pathology , Femoral Artery/surgery , Gene Expression Regulation , HMGB1 Protein/biosynthesis , Hindlimb/growth & development , Hindlimb/metabolism , Hindlimb/pathology , Humans , Inflammation/pathology , Interleukin-6/biosynthesis , Ischemia/metabolism , Ischemia/pathology , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Signal Transduction , Toll-Like Receptor 2/biosynthesis , Toll-Like Receptor 4/biosynthesis
8.
Circ Res ; 109(5): 524-33, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21719759

ABSTRACT

RATIONALE: Positive outward remodeling of pre-existing collateral arteries into functional conductance arteries, arteriogenesis, is a major endogenous rescue mechanism to prevent cardiovascular ischemia. Collateral arterial growth is accompanied by expression of kinin precursor. However, the role of kinin signaling via the kinin receptors (B1R and B2R) in arteriogenesis is unclear. OBJECTIVE: The purpose of this study was to elucidate the functional role and mechanism of bradykinin receptor signaling in arteriogenesis. METHODS AND RESULTS: Bradykinin receptors positively affected arteriogenesis, with the contribution of B1R being more pronounced than B2R. In mice, arteriogenesis upon femoral artery occlusion was significantly reduced in B1R mutant mice as evidenced by reduced microspheres and laser Doppler flow perfusion measurements. Transplantation of wild-type bone marrow cells into irradiated B1R mutant mice restored arteriogenesis, whereas bone marrow chimeric mice generated by reconstituting wild-type mice with B1R mutant bone marrow showed reduced arteriogenesis after femoral artery occlusion. In the rat brain 3-vessel occlusion arteriogenesis model, pharmacological blockade of B1R inhibited arteriogenesis and stimulation of B1R enhanced arteriogenesis. In the rat, femoral artery ligation combined with arterial venous shunt model resulted in flow-driven arteriogenesis, and treatment with B1R antagonist R715 decreased vascular remodeling and leukocyte invasion (monocytes) into the perivascular tissue. In monocyte migration assays, in vitro B1R agonists enhanced migration of monocytes. CONCLUSIONS: Kinin receptors act as positive modulators of arteriogenesis in mice and rats. B1R can be blocked or therapeutically stimulated by B1R antagonists or agonists, respectively, involving a contribution of peripheral immune cells (monocytes) linking hemodynamic conditions with inflammatory pathways.


Subject(s)
Arteries/growth & development , Receptor, Bradykinin B1/physiology , Receptor, Bradykinin B2/physiology , Signal Transduction/physiology , Animals , Arterial Occlusive Diseases/metabolism , Arterial Occlusive Diseases/physiopathology , Arteries/physiopathology , Cerebral Arteries/growth & development , Femoral Artery/growth & development , Hindlimb/blood supply , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/physiopathology , Rats , Rats, Sprague-Dawley
9.
Arterioscler Thromb Vasc Biol ; 31(2): 306-12, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21071685

ABSTRACT

OBJECTIVE: To investigate the role of recombinant human interleukin-11 (rhIL-11) on in vivo mobilization of CD34(+)/vascular endothelial growth factor receptor (VEGFR) 2(+) mononuclear cells and collateral vessel remodeling in a mouse model of hindlimb ischemia. METHODS AND RESULTS: We observed that treatment of Sv129 mice with continuous infusion of 200-µg/kg rhIL-11 per day led to in vivo mobilization of CD34(+)/VEGFR2(+) cells that peaked at 72 hours. Sv129 mice pretreated with rhIL-11 for 72 hours before femoral artery ligation showed a 3-fold increase in plantar vessel perfusion, leading to faster blood flow recovery; and a 20-fold increase in circulating CD34(+)/VEGFR2(+) cells after 8 days of rhIL-11 treatment. Histologically, experimental mice had a 3-fold increase in collateral vessel luminal diameter after 21 days of rhIL-11 treatment and a 4.4-fold influx of perivascular CD34(+)/VEGFR2(+) cells after 8 days of therapy. Functionally, rhIL-11-treated mice showed better hindlimb appearance and use scores when compared with syngeneic mice treated with PBS under the same experimental conditions. CONCLUSIONS: These novel findings show that rhIL-11 promotes in vivo mobilization of CD34(+)/VEGFR2(+) mononuclear cells, enhances collateral vessel growth, and increases recovery of perfusion after femoral artery ligation. Thus, rhIL-11 has a promising role for development as an adjunctive treatment of patients with peripheral vascular disease.


Subject(s)
Femoral Artery/drug effects , Femoral Artery/growth & development , Hindlimb/blood supply , Interleukin-11/pharmacology , Ischemia/metabolism , Recombinant Proteins/pharmacology , Animals , Antigens, CD34/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Disease Models, Animal , Femoral Artery/cytology , Humans , Ischemia/pathology , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Ligation , Mice , Neovascularization, Physiologic/physiology , STAT3 Transcription Factor/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
Am J Physiol Heart Circ Physiol ; 298(6): H1959-65, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20228260

ABSTRACT

Technologies to increase tissue vascularity are critically important to the fields of tissue engineering and cardiovascular medicine. Currently, limited technologies exist to encourage angiogenesis and arteriogenesis in a controlled manner. In the present study, we describe an injectable controlled release system consisting of VEGF encapsulated in poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs). The majority of VEGF was released gradually over 2-4 days from the NPs as determined by an ELISA release kinetics experiment. An in vitro aortic ring bioassay was used to verify the bioactivity of VEGF-NPs compared with empty NPs and no treatment. A mouse femoral artery ischemia model was then used to measure revascularization in VEGF-NP-treated limbs compared with limbs treated with naked VEGF and saline. 129/Sv mice were anesthetized with isoflurane, and a region of the common femoral artery and vein was ligated and excised. Mice were then injected with VEGF-NPs, naked VEGF, or saline. After 4 days, three-dimensional microcomputed tomography angiography was used to quantify vessel growth and morphology. Mice that received VEGF-NP treatment showed a significant increase in total vessel volume and vessel connectivity compared with 5 microg VEGF, 2.5 microg VEGF, and saline treatment (all P < 0.001). When the yield of the fabrication process was taken into account, VEGF-NPs were over an order of magnitude more potent than naked VEGF in increasing blood vessel volume. Differences between the VEGF-NP group and all other groups were even greater when only small-sized vessels under 300 mum diameter were analyzed. In conclusion, sustained VEGF delivery via PLGA NPs shows promise for encouraging blood vessel growth in tissue engineering and cardiovascular medicine applications.


Subject(s)
Biocompatible Materials , Lactic Acid , Nanoparticles , Neovascularization, Physiologic/drug effects , Polyglycolic Acid , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/pharmacology , Animals , Aorta/growth & development , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Femoral Artery/diagnostic imaging , Femoral Artery/growth & development , Hindlimb/blood supply , Ischemia/drug therapy , Ischemia/physiopathology , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , Polylactic Acid-Polyglycolic Acid Copolymer , Tomography, X-Ray Computed , Vascular Endothelial Growth Factor A/therapeutic use
11.
Mol Cell Biochem ; 322(1-2): 15-23, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18979232

ABSTRACT

Arteriogenesis or collateral growth is able to compensate for the stenosis of major arteries. Using differential display RT-PCR on growing and quiescent collateral arteries in a rabbit femoral artery ligation model, we cloned the rabbit full-length cDNA of osteoglycin/mimecan. Osteoglycin was present in the adventitia of collateral arteries as a glycosylated protein without keratan sulfate side chains, mainly produced by smooth muscle cells (SMCs) and perivascular fibroblasts. Northern blot, Western blot, and immunohistochemistry confirmed a collateral artery-specific downregulation of osteoglycin from 6 h to 3 weeks after the onset of arteriogenesis. Treatment of primary SMCs with the arteriogenic protein fibroblast growth factor-2 (FGF-2) resulted in a similar reduction of osteoglycin expression as observed in vivo. Application of the FGF-2 inhibitor polyanethole sulfonic acid (PAS) blocked the downregulation of osteoglycin and interfered with arteriogenesis. From our study we conclude that downregulation of osteoglycin is a fundamental requirement for proper arteriogenesis.


Subject(s)
Femoral Artery/growth & development , Proteoglycans/metabolism , Amino Acid Sequence , Animals , Arteries/growth & development , Base Sequence , DNA, Complementary/chemistry , Down-Regulation , Fibroblast Growth Factor 2/metabolism , Models, Animal , Molecular Sequence Data , Muscle, Smooth, Vascular/metabolism , Proteoglycans/chemistry , Proteoglycans/genetics , RNA, Messenger/metabolism , Rabbits , Reverse Transcriptase Polymerase Chain Reaction
12.
AJR Am J Roentgenol ; 192(6): W306-10, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19457794

ABSTRACT

OBJECTIVE: Regenerative therapy is a new treatment of vascular occlusive diseases. The purpose of this study was to examine the advantages of repeated low-dose growth factor infusions compared with a single high-dose infusion in an ischemic hind-limb rabbit model. MATERIALS AND METHODS: Thirty-two rabbits were used to construct an ischemic hind-limb model by resection of the left femoral artery. For the vascular regenerative method, basic fibroblast growth factor (bFGF) was impregnated into 3 mg of gelatin microspheres 30 microm in diameter and a reservoir system was implanted in the left femoral artery for infusion. The gelatin microspheres were then infused into the left internal iliac artery via the reservoir system. The rabbits were divided into three groups according to different infusion methods: single high-dose infusion, repeated low-dose infusions, and saline (control). Therapeutic effects were evaluated by thigh temperature, blood pressure, blood flow, angiography, and pathology. RESULTS: There was no significant difference between the two infusion methods in thigh temperature, blood pressure, blood flow, angiography, and pathology. In pathologic analyses at 2 and 4 weeks, both the repeated low-dose infusion and the single high-dose infusion groups showed significant differences in the number of vessels when compared with the control group. CONCLUSION: The efficacy of repeated bFGF infusions for neovascularization via the reservoir method was investigated. Despite the pathologic confirmation of neovascularization, there was no significant difference in treatment effect by the two administration methods.


Subject(s)
Femoral Artery/growth & development , Fibroblast Growth Factor 2/administration & dosage , Hindlimb/blood supply , Ischemia/drug therapy , Neovascularization, Physiologic/drug effects , Regeneration/drug effects , Animals , Dose-Response Relationship, Drug , Female , Femoral Artery/drug effects , Hindlimb/drug effects , Infusions, Intra-Arterial , Ischemia/diagnostic imaging , Rabbits , Radiography , Treatment Outcome
13.
Clin Hemorheol Microcirc ; 73(1): 195-201, 2019.
Article in English | MEDLINE | ID: mdl-31561347

ABSTRACT

BACKGROUND: In cardiovascular research small pigs breeds like Göttingen® minipigs (GM) are established animal models, but systematic data about the micromorphology of the GM vasculature at different ages are scarce. OBJECTIVE: The study was aimed at gaining knowledge about the micromorphology of the femoral artery (FA) from German Landrace pigs (DL) and GM during the period of growth over a body weight range of 10-40 kg. METHODS: FA samples from DL aged two or three months were compared to GM ones, aged 18 or 40 months using transmitted light microscopy. RESULTS: All FA samples showed typical characteristics of muscular arteries. Growth was associated with increased vessel wall thickness. In the GM this resulted in a slight decrease of the luminal diameter (LD), while in the DL pigs, an increase of the LD and smooth muscle cell content (10%) with decreased elastic fiber content (10%) has been detected. In contrast, within the 22 months lasting growth period of the GM, the tunica media content of smooth muscle cells and elastic fibers remained stable. CONCLUSIONS: FA maturation strongly depends on the pig breed and age. It can be different from what is described in humans.


Subject(s)
Femoral Artery/growth & development , Tunica Media/growth & development , Animals , Swine
14.
Arterioscler Thromb Vasc Biol ; 27(11): 2310-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17717295

ABSTRACT

OBJECTIVE: The immune system is thought to play a crucial role in regulating collateral circulation (arteriogenesis), a vital compensatory mechanism in patients with arterial obstructive disease. Here, we studied the role of lymphocytes in a murine model of hindlimb ischemia. METHODS AND RESULTS: Lymphocytes, detected with markers for NK1.1, CD3, and CD4, invaded the collateral vessel wall. Arteriogenesis was impaired in C57BL/6 mice depleted for Natural Killer (NK)-cells by anti-NK1.1 antibodies and in NK-cell-deficient transgenic mice. Arteriogenesis was, however, unaffected in J alpha281-knockout mice that lack NK1.1+ Natural Killer T (NKT)-cells, indicating that NK-cells, rather than NKT-cells, are involved in arteriogenesis. Furthermore, arteriogenesis was impaired in C57BL/6 mice depleted for CD4+ T-lymphocytes by anti-CD4 antibodies, and in major histocompatibility complex (MHC)-class-II-deficient mice that more selectively lack mature peripheral CD4+ T-lymphocytes. This impairment was even more profound in anti-NK1.1-treated MHC-class-II-deficient mice that lack both NK- and CD4+ T-lymphocytes. Finally, collateral growth was severely reduced in BALB/c as compared with C57BL/6 mice, 2 strains with different bias in immune responsiveness. CONCLUSIONS: These data show that both NK-cells and CD4+ T-cells modulate arteriogenesis. Promoting lymphocyte activation may represent a promising method to treat ischemic disease.


Subject(s)
Arterial Occlusive Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Collateral Circulation/immunology , Killer Cells, Natural/immunology , Neovascularization, Physiologic/immunology , Animals , Arterial Occlusive Diseases/physiopathology , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Femoral Artery/growth & development , Hindlimb/blood supply , Ischemia , Killer Cells, Natural/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout
15.
EBioMedicine ; 27: 237-246, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29233575

ABSTRACT

Midkine is a pleiotropic factor, which is involved in angiogenesis. However, its mode of action in this process is still ill defined. The function of midkine in arteriogenesis, the growth of natural bypasses from pre-existing collateral arteries, compensating for the loss of an occluded artery has never been investigated. Arteriogenesis is an inflammatory process, which relies on the proliferation of endothelial cells and smooth muscle cells. We show that midkine deficiency strikingly interferes with the proliferation of endothelial cells in arteriogenesis, thereby interfering with the process of collateral artery growth. We identified midkine to be responsible for increased plasma levels of vascular endothelial growth factor A (VEGFA), necessary and sufficient to promote endothelial cell proliferation in growing collaterals. Mechanistically, we demonstrate that leukocyte domiciled midkine mediates increased plasma levels of VEGFA relevant for upregulation of endothelial nitric oxide synthase 1 and 3, necessary for proper endothelial cell proliferation, and that non-leukocyte domiciled midkine additionally improves vasodilation. The data provided on the role of midkine in endothelial proliferation are likely to be relevant for both, the process of arteriogenesis and angiogenesis. Moreover, our data might help to estimate the therapeutic effect of clinically applied VEGFA in patients with vascular occlusive diseases.


Subject(s)
Femoral Artery/growth & development , Femoral Artery/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Nitric Oxide Synthase/metabolism , Organogenesis/drug effects , Vascular Endothelial Growth Factor A/metabolism , Animals , Biological Availability , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Proliferation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Femoral Artery/drug effects , Leukocytes/drug effects , Leukocytes/metabolism , Mice, Inbred C57BL , Midkine , Models, Biological , Nitroso Compounds/pharmacology
16.
J Clin Invest ; 101(6): 1225-32, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9502763

ABSTRACT

To begin to dissect atherogenesis as a complex genetic disorder affected by genetic makeup and environment, we have (a) generated a reproducible mouse model of neointimal growth; (b) evaluated the effect of disruption of a single gene, endothelial nitric oxide synthase, believed to be central to intimal growth, and (c) examined the modifying effects of gender and pregnancy upon the vascular response. Cuff placement around the femoral artery causes reproducible intimal growth. We assessed the response to injury by quantitative morphometry, measuring the intimal to medial (I/M) volume ratio. In wild-type mice, cuff placement causes pronounced intimal proliferation without affecting the media, resulting in I/M ratios of 31% (SV129 males) and 27% (C57BL/6 males). eNOS mutant male mice have a much greater degree of intimal growth (I/M ratio of 70%). Female mice show less intimal response than do males, although eNOS mutant female mice still have more response than do wild-type females. Most dramatic, however, is the effect of pregnancy, which essentially abolishes the intimal response to injury, even overriding the effect of eNOS mutation. We conclude that eNOS deficiency is a genetic predisposition to intimal proliferation that is enhanced by male gender, and that may be overridden by pregnancy.


Subject(s)
Endothelium, Vascular/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Animals , Disease Models, Animal , Endothelium, Vascular/growth & development , Endothelium, Vascular/injuries , Female , Femoral Artery/growth & development , Femoral Artery/injuries , Femoral Artery/metabolism , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Pregnancy , Sex Factors , Tunica Intima/growth & development , Tunica Intima/injuries , Tunica Intima/metabolism
17.
J Am Acad Orthop Surg ; 24(8): 515-26, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27428883

ABSTRACT

Throughout development, the vascular supply to the proximal femur and acetabulum undergoes a series of changes during which it is susceptible to injury. Before age 3 months, the ligamentum teres and lateral epiphyseal arteries are the dominant supply to the developing head. The dominant supply shifts to the lateral epiphyseal vessels by age 18 months. The distinct metaphyseal and epiphyseal circulations of the adult proximal femur form in adolescence when an increasingly rich metaphyseal circulation supplies the subphyseal region, terminating at the physeal plate. The acetabular blood supply derives from two independent systems, with the dominance of each changing throughout maturity. Most descriptions of the vascular contributions to the proximal femur and acetabulum have been gross anatomic and histologic studies. Advanced imaging studies (eg, CT angiography, perfusion MRI) have added to our understanding of the vascular anatomy of the proximal femur and acetabulum, its changes throughout development, and its clinical implications.


Subject(s)
Hip/blood supply , Acetabulum/blood supply , Acetabulum/growth & development , Femoral Artery/anatomy & histology , Femoral Artery/growth & development , Femur Head/blood supply , Femur Head/growth & development , Hip/growth & development , Humans , Round Ligaments/blood supply , Round Ligaments/growth & development
18.
Circulation ; 103(4): 562-9, 2001 Jan 30.
Article in English | MEDLINE | ID: mdl-11157723

ABSTRACT

BACKGROUND: Smooth muscle cell migration, in addition to proliferation, contributes to a large extent to the neointima formed in humans after balloon angioplasty or bypass surgery. Plasminogen activator/plasmin-mediated proteolysis is an important mediator of this smooth muscle cell migration. Here, we report the construction of a novel hybrid protein designed to inhibit the activity of cell surface-bound plasmin, which cannot be inhibited by its natural inhibitors, such as alpha(2)-antiplasmin. This hybrid protein, consisting of the receptor-binding amino-terminal fragment of uPA (ATF), linked to the potent protease inhibitor bovine pancreas trypsin inhibitor (BPTI), can inhibit plasmin activity at the cell surface. METHODS AND RESULTS: The effect of adenovirus-mediated ATF.BPTI expression on neointima formation was tested in human saphenous vein organ cultures. Infection of human saphenous vein segments with Ad.CMV.ATF.BPTI (5x10(9) pfu/mL) resulted in 87.5+/-3.8% (mean+/-SEM, n=10) inhibition of neointima formation after 5 weeks, whereas Ad.CMV.ATF or Ad.CMV.BPTI virus had only minimal or no effect on neointima formation. The efficacy of ATF.BPTI in vivo was demonstrated in a murine model for neointima formation. Neointima formation in the femoral artery of mice, induced by placement of a polyethylene cuff, was strongly inhibited (93.9+/-2%) after infection with Ad.CMV.mATF.BPTI, a variant of ATF.BPTI able to bind specifically to murine uPA receptor; Ad.CMV.mATF and Ad.CMV.BPTI had no significant effect. CONCLUSIONS: These data provide evidence that adenoviral transfer of a hybrid protein that binds selectively to the uPA receptor and inhibits plasmin activity directly on the cell surface is a powerful approach to inhibiting neointima formation and restenosis.


Subject(s)
Aprotinin/physiology , Blood Vessels/physiology , Tunica Intima/growth & development , Urokinase-Type Plasminogen Activator/physiology , Adenoviridae/genetics , Animals , Aprotinin/genetics , CHO Cells , Cattle , Cricetinae , Femoral Artery/growth & development , Femoral Artery/injuries , Femoral Vein/cytology , Femoral Vein/metabolism , Fibrinolysin/metabolism , Gene Expression , Genetic Vectors/genetics , Humans , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Organ Culture Techniques , Peptide Fragments/genetics , Peptide Fragments/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Saphenous Vein/cytology , Transfection , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/genetics
19.
Coron Artery Dis ; 16(7): 451-6, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16205454

ABSTRACT

OBJECTIVES: We previously demonstrated that sphingosine kinase (SPK) increases the level of extracellular sphingosine-1-phosphate and promotes neovascularization in a mouse matrigel model. In this study, we tested the hypothesis that SPK gene transfer using a novel adenoviral 'gutless' vector (AGV) can enhance arteriogenesis in a rabbit hindlimb ischemia model. METHODS: Thirty-five male New Zealand white rabbits were randomized to the AGV-SPK group (n=13), AGV-null group (n=13), and control group (n=9). On day 10, after the induction of unilateral hindlimb ischemia, gene vectors or buffer were introduced and the effect examined on day 30, using calf blood pressure, quantitative angiographic analysis, and histology. RESULTS: Calf systolic blood pressure ratios of the ischemic limb to the normal limb on day 30 were 0.77+/-0.13 in control groups, including the AGV-null group, and 0.91+/-0.14 in the AGV-SPK group (P<0.05). Angiographic vessel counts were significantly increased (8.0+/-2.1 at baseline and 11.8+/-3.2 on day 30, P<0.001) in the AGV-SPK group. Histologic analysis showed that microscopic total vessel counts on day 30 were 3.5+/-1.8/field in the control and AGV-null group and 5.4+/-1.0/field in the AGV-SPK group. Arterioles (AGV-SPK; 3.0+/-0.8 versus control and AGV-null; 2.1+/-1.1, P<0.05) were significantly increased in the AGV-SPK group. CONCLUSIONS: This study shows that SPK promotes arteriogenesis, as evidenced by the maximal improvement in the blood pressure restoration and collateral vessel counts. SPK may be an important angiogenic target to improve perfusion in ischemic tissues.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Genetic Therapy/methods , Ischemia/drug therapy , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Adenoviridae/genetics , Angiogenesis Inducing Agents/therapeutic use , Angiography , Animals , Femoral Artery/drug effects , Femoral Artery/growth & development , Gene Transfer Techniques , Genetic Vectors/biosynthesis , Hindlimb/blood supply , Male , Models, Animal , Neovascularization, Physiologic/drug effects , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/therapeutic use , Rabbits , Regional Blood Flow/drug effects
20.
Atherosclerosis ; 34(2): 167-91, 1979 Oct.
Article in English | MEDLINE | ID: mdl-518734

ABSTRACT

In infants, human femoral arteries display seam-like internal elastic lamina (IEL) covered with endothelium on the luminal side and with smooth muscle cells (SMC) on the medial side. At birth the growth of IEL is finished, correlated with a loss of microfibrils (MF) at the periphery. With the onset of the postnatal vessel growth the joints of IEL seem to be mechanically widened until they have the appearance of gaps with progressing age. After the age of 40 years there are often rod-like crystallites in the IEL, probably composed of cholesterol esters. A small first consecutive lamina (CL) can be seen already in childhood; it enlarges until the 3rd decade of life and is interpreted as a substitute to the "fragmented" IEL. After the 5th decade of life the first CL is arranged within the intima at a certain distance from the IEL and consisting of loosely arranged elastic fibrils. In very old arteries (beyond the 8th decade of life) gaps are rarely seen in the first CL. In individuals over the age of 30 years, the space between IEL and the first CL is occupied by smooth muscle cells (SMC) which are tightly packed. Additional CLs above the first CL can be found in elderly individuals, there CL obviously contribute to the intimal thickening. The ultrastructure of the elastic elements of the vessel wall and their possible function are discussed.


Subject(s)
Aging , Elastin/metabolism , Femoral Artery/growth & development , Adolescent , Adult , Aged , Calcium/metabolism , Child , Child, Preschool , Elastic Tissue/metabolism , Female , Femoral Artery/anatomy & histology , Femoral Artery/metabolism , Humans , Infant , Infant, Newborn , Lipid Metabolism , Male , Middle Aged , Pancreatic Elastase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL