Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 42(2): 175-188, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34879707

RESUMEN

OBJECTIVE: Monocytes, which play an important role in arteriogenesis, can build immunologic memory by a functional reprogramming that modifies their response to a second challenge. This process, called trained immunity, is evoked by insults that shift monocyte metabolism, increasing HIF (hypoxia-inducible factor)-1α levels. Since ischemia enhances HIF-1α, we evaluate whether ischemia can lead to a functional reprogramming of monocytes, which would contribute to arteriogenesis after hindlimb ischemia. METHODS AND RESULTS: Mice exposed to ischemia by 24 hours (24h) of femoral artery occlusion (24h trained) or sham were subjected to hindlimb ischemia one week later; the 24h trained mice showed significant improvement in blood flow recovery and arteriogenesis after hindlimb ischemia. Adoptive transfer using bone marrow-derived monocytes (BM-Mono) from 24h trained or sham donor mice, demonstrated that recipients subjected to hindlimb ischemia who received 24h ischemic-trained monocytes had remarkable blood flow recovery and arteriogenesis. Further, ischemic-trained BM-Mono had increased HIF-1α and GLUT-1 (glucose transporter-1) gene expression during femoral artery occlusion. Circulating cytokines and GLUT-1 were also upregulated during femoral artery occlusion.Transcriptomic analysis and confirmatory qPCR performed in 24h trained and sham BM-Mono revealed that among the 15 top differentially expressed genes, 4 were involved in lipid metabolism in the ischemic-trained monocytes. Lipidomic analysis confirmed that ischemia training altered the cholesterol metabolism of these monocytes. Further, several histone-modifying epigenetic enzymes measured by qPCR were altered in mouse BM-Mono exposed to 24h hypoxia. CONCLUSIONS: Ischemia training in BM-Mono leads to a unique gene profile and improves blood flow and arteriogenesis after hindlimb ischemia.


Asunto(s)
Traslado Adoptivo , Miembro Posterior/irrigación sanguínea , Isquemia/terapia , Monocitos/trasplante , Neovascularización Fisiológica , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Miembro Posterior/inmunología , Miembro Posterior/fisiopatología , Isquemia/inmunología , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología
2.
Am J Physiol Heart Circ Physiol ; 317(4): H867-H876, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31441677

RESUMEN

Atherosclerosis is the most common underlying cause of cardiovascular morbidity and mortality worldwide. c-Kit (CD117) is a member of the receptor tyrosine kinase family, which regulates differentiation, proliferation, and survival of multiple cell types. Recent studies have shown that c-Kit and its ligand stem cell factor (SCF) are present in arterial endothelial cells and smooth muscle cells (SMCs). The role of c-Kit in cardiovascular disease remains unclear. The aim of the current study is to determine the role of c-Kit in atherogenesis. For this purpose, atherosclerotic plaques were quantified in c-Kit-deficient mice (KitMut) after they were fed a high-fat diet (HFD) for 16 wk. KitMut mice demonstrated substantially greater atherosclerosis compared with control (KitWT) littermates (P < 0.01). Transplantation of c-Kit-positive bone marrow cells into KitMut mice failed to rescue the atherogenic phenotype, an indication that increased atherosclerosis was associated with reduced arterial c-Kit. To investigate the mechanism, SMC organization and morphology were analyzed in the aorta by histopathology and electron microscopy. SMCs were more abundant, disorganized, and vacuolated in aortas of c-Kit mutant mice compared with controls (P < 0.05). Markers of the "contractile" SMC phenotype (calponin, SM22α) were downregulated with pharmacological and genetic c-Kit inhibition (P < 0.05). The absence of c-Kit increased lipid accumulation and significantly reduced the expression of the ATP-binding cassette transporter G1 (ABCG1) necessary for lipid efflux in SMCs. Reconstitution of c-Kit in cultured KitMut SMCs resulted in increased spindle-shaped morphology, reduced proliferation, and elevated levels of contractile markers, all indicators of their restored contractile phenotype (P < 0.05).NEW & NOTEWORTHY This study describes the novel vasculoprotective role of c-Kit against atherosclerosis and its function in the preservation of the SMC contractile phenotype.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Hiperlipidemias/complicaciones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Animales , Aorta/metabolismo , Aorta/ultraestructura , Enfermedades de la Aorta/etiología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Aterosclerosis/etiología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Células Espumosas/metabolismo , Células Espumosas/patología , Humanos , Hiperlipidemias/metabolismo , Ratones Noqueados para ApoE , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/ultraestructura , Mutación , Miocitos del Músculo Liso/ultraestructura , Fenotipo , Placa Aterosclerótica , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-kit/genética , Transducción de Señal , Calponinas
3.
Biochem Biophys Res Commun ; 518(2): 227-232, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31416613

RESUMEN

INTRODUCTION: Receptor tyrosine kinases have been implicated in various vascular remodeling processes and cardiovascular disease. However, their role in the regulation of vascular tone is poorly understood. Herein, we evaluate the contribution of c-Kit signaling to vasoactive responses. METHODS: The vascular reactivity of mesenteric arteries was assessed under isobaric conditions in c-Kit deficient (KitW/W-v) and littermate control mice (Kit+/+) using pressure myography. Protein levels of soluble guanylyl cyclase beta 1 (sGCß1) were quantified by Western blot. Mean arterial pressure was measured after high salt (8% NaCl) diet treatment using the tail-cuff method. RESULTS: Smooth muscle cells (SMCs) from c-Kit deficient mice showed a 5-fold downregulation of sGCß1 compared to controls. Endothelium-dependent relaxation of mesenteric arteries demonstrated a predominance of prostanoid vs. nitric oxide (NO) signaling in both animal groups. The dependence on prostanoid-induced dilation was higher in c-Kit mutant mice than in controls, as indicated by a significant impairment in vasorelaxation with indomethacin with respect to the latter. Endothelium-independent relaxation showed significant dysfunction of NO signaling in c-Kit deficient SMCs compared to controls. Mesenteric artery dilation was rescued by addition of a cGMP analog, but not with a NO donor, indicating a deficiency in cGMP production in c-Kit deficient SMCs. Finally, c-Kit deficient mice developed higher blood pressure on an 8% NaCl diet compared to their control littermates. CONCLUSION: c-Kit deficiency inhibits NO signaling in SMCs. The existence of this c-Kit/sGC signaling axis may be relevant for vascular reactivity and remodeling.


Asunto(s)
Miocitos del Músculo Liso/metabolismo , Óxido Nítrico/metabolismo , Proteínas Proto-Oncogénicas c-kit/deficiencia , Transducción de Señal , Animales , Arterias/efectos de los fármacos , Arterias/fisiología , Presión Sanguínea/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Ratones , Prostaglandinas/farmacología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal/efectos de los fármacos , Cloruro de Sodio Dietético , Vasodilatación/efectos de los fármacos
4.
J Surg Res ; 228: 68-76, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29907232

RESUMEN

BACKGROUND: Poor wound healing in critical limb ischemia (CLI) is attributed to impaired neovascularization and reperfusion. Optimizing the ischemic wound with adhesion molecules that enhance stem cell homing may revolutionize treatment. The purpose of this study is to test the efficacy of adhesion molecule E-selectin on wound healing in an ischemic mouse wound. METHODS: Adult FVB/NJ mice underwent unilateral femoral artery and vein ligation to induce CLI. A 4-mm punch biopsy wound was created on the anterior thigh to simulate ischemic wounds. Intramuscular injection of adeno-associated virus (AAV) carrying either E-selectin (E-selectin/AAV, n = 11) or LacZ as control (LacZ/AAV, n = 10) was performed. Gross wound size was measured for 10 d postoperatively. Ischemic hindlimb reperfusion was quantified using laser Doppler imaging. Wound tissue neovascularization was visualized using DiI perfusion and confocal microscopy. E-selectin expression in wounds was verified by immunofluorescence. RESULTS: Immunofluorescence confirmed E-selectin/AAV delivery in treatment versus control limbs. Wounds from E-selectin/AAV mice versus controls revealed surface area healing of 54% versus 20% (P < 0.01) on postoperative day (POD) 1, 78% versus 51% on POD 4 (P < 0.01), and 97% versus 84% on POD 10 (P < 0.01). Laser Doppler imaging revealed greater reperfusion in E-selectin/AAV mice versus controls by POD 10 (0.49 versus 0.27, P < 0.05). DiI perfused ligated hindlimb in E-selectin/AAV versus control mice revealed mean neovascularization intensity score of 30 versus 18 (P < 0.05) on POD 10. CONCLUSIONS: Intramuscularly injected E-selectin/AAV gene therapy in mice with CLI significantly increases wound angiogenesis and limb reperfusion, expediting overall wound healing.


Asunto(s)
Selectina E/genética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Isquemia/terapia , Cicatrización de Heridas/genética , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Células HEK293 , Miembro Posterior/irrigación sanguínea , Miembro Posterior/diagnóstico por imagen , Humanos , Inyecciones Intramusculares , Isquemia/diagnóstico por imagen , Isquemia/genética , Flujometría por Láser-Doppler , Masculino , Ratones , Neovascularización Fisiológica/genética , Flujo Sanguíneo Regional/genética , Piel/irrigación sanguínea , Piel/diagnóstico por imagen
5.
Ann Vasc Surg ; 48: 222-232, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29197603

RESUMEN

BACKGROUND: Lack of a reliable hind limb gangrene animal model limits preclinical studies of gangrene, a severe form of critical limb ischemia. We develop a novel mouse hind limb gangrene model to facilitate translational studies. METHODS: BALB/c, FVB, and C57BL/6 mice underwent femoral artery ligation (FAL) with or without administration of NG-nitro-L-arginine methyl ester (L-NAME), an endothelial nitric oxide synthase inhibitor. Gangrene was assessed using standardized ischemia scores ranging from 0 (no gangrene) to 12 (forefoot gangrene). Laser Doppler imaging (LDI) and DiI perfusion quantified hind limb reperfusion postoperatively. RESULTS: BALB/c develops gangrene with FAL-only (n = 11/11, 100% gangrene incidence), showing mean limb ischemia score of 12 on postoperative days (PODs) 7 and 14 with LDI ranging from 0.08 to 0.12 on respective PODs. Most FVB did not develop gangrene with FAL-only (n = 3/9, 33% gangrene incidence) but with FAL and L-NAME (n = 9/9, 100% gangrene incidence). Mean limb ischemia scores for FVB undergoing FAL with L-NAME were significantly higher than for FVB receiving FAL-only. LDI score and capillary density by POD 28 were significantly lower in FVB undergoing FAL with L-NAME. C57BL/6 did not develop gangrene with FAL-only or FAL and L-NAME. CONCLUSIONS: Reproducible murine gangrene models may elucidate molecular mechanisms for gangrene development, facilitating therapeutic intervention.


Asunto(s)
Arteria Femoral/cirugía , Isquemia/etiología , Músculo Esquelético/irrigación sanguínea , NG-Nitroarginina Metil Éster , Enfermedad Arterial Periférica/etiología , Animales , Velocidad del Flujo Sanguíneo , Modelos Animales de Enfermedad , Gangrena , Miembro Posterior , Isquemia/enzimología , Isquemia/patología , Isquemia/fisiopatología , Ligadura , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Músculo Esquelético/patología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/metabolismo , Enfermedad Arterial Periférica/enzimología , Enfermedad Arterial Periférica/patología , Enfermedad Arterial Periférica/fisiopatología , Flujo Sanguíneo Regional , Especificidad de la Especie , Factores de Tiempo
6.
Biomedicines ; 12(6)2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38927565

RESUMEN

BACKGROUND: Critical limb ischemia (CLI) is the end stage of peripheral artery disease (PAD), and around 30% of CLI patients are ineligible for current treatments. The angiogenic benefits of c-Kit have been reported in the ischemia scenario; however, the present study demonstrates the effects of specific endothelial c-Kit signaling in arteriogenesis during hindlimb ischemia. METHODS: We created conditional knockout mouse models that decrease c-Kit (c-Kit VE-Cadherin CreERT2-c-Kit) or its ligand (SCF VE-Cadherin CreERT2-SCF) specifically in endothelial cells (ECs) after tamoxifen treatment. These mice and a control group (wild-type VE-Cadherin CreERT2-WT) were subjected to hindlimb ischemia or aortic crush to evaluate perfusion/arteriogenesis and endothelial barrier permeability, respectively. RESULTS: Our data confirmed the lower gene expression of c-Kit and SCF in the ECs of c-Kit and SCF mice, respectively. In addition, we confirmed the lower percentage of ECs positive for c-Kit in c-Kit mice. Further, we found that c-Kit and SCF mice had better limb perfusion and arteriogenesis compared to WT mice. We also demonstrated that c-Kit and SCF mice had a preserved endothelial barrier after aortic crush compared to WT. CONCLUSIONS: Our data demonstrate the deleterious effects of endothelial SCF/c-Kit signaling on arteriogenesis and endothelial barrier integrity.

7.
Kidney Int Rep ; 8(4): 837-850, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37069981

RESUMEN

Introduction: The molecular transformation of the human preaccess vein after arteriovenous fistula (AVF) creation is poorly understood. This limits our ability to design efficacious therapies to improve maturation outcomes. Methods: Bulk RNA sequencing (RNA-seq) followed by paired bioinformatic analyses and validation assays were performed in 76 longitudinal vascular biopsies (veins and AVFs) from 38 patients with stage 5 chronic kidney disease or end-stage kidney disease undergoing surgeries for 2-stage AVF creation (19 matured, 19 failed). Results: A total of 3637 transcripts were differentially expressed between veins and AVFs independent of maturation outcomes, with 80% upregulated in fistulas. The postoperative transcriptome demonstrated transcriptional activation of basement membrane and interstitial extracellular matrix (ECM) components, including preexisting and novel collagens, proteoglycans, hemostasis factors, and angiogenesis regulators. A postoperative intramural cytokine storm involved >80 chemokines, interleukins, and growth factors. Postoperative changes in ECM expression were differentially distributed in the AVF wall, with proteoglycans and fibrillar collagens predominantly found in the intima and media, respectively. Interestingly, upregulated matrisome genes were enough to make a crude separation of AVFs that failed from those with successful maturation. We identified 102 differentially expressed genes (DEGs) in association with AVF maturation failure, including upregulation of network collagen VIII in medial smooth muscle cells (SMCs) and downregulation of endothelial-predominant transcripts and ECM regulators. Conclusion: This work delineates the molecular changes that characterize venous remodeling after AVF creation and those relevant to maturation failure. We provide an essential framework to streamline translational models and our search for antistenotic therapies.

8.
Arterioscler Thromb Vasc Biol ; 31(8): 1748-56, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21617137

RESUMEN

OBJECTIVE: Aging is a major risk factor for increased ischemic tissue injury. Whether collateral rarefaction and impaired remodeling contribute to this is unknown. We quantified the number and diameter of native collaterals and their remodeling in 3-, 16-, 24-, and 31-month-old mice. METHODS AND RESULTS: Aging caused an "age-dose-dependent" greater drop in perfusion immediately after femoral artery ligation, followed by a diminished recovery of flow and increase in tissue injury. These effects were associated with a decline in collateral number, diameter, and remodeling. Angiogenesis was also impaired. Mechanistically, these changes were not accompanied by reduced recruitment of T cells or macrophages to remodeling collaterals. However, endothelial nitric oxide synthase signaling was dysfunctional, as indicated by increased protein nitrosylation and less phosphorylated endothelial nitric oxide synthase and vasodilator-stimulated phosphoprotein in collateral wall cells. The cerebral circulation exhibited a similar age-dose-dependent loss of collateral number and diameter and increased tortuosity, resulting in an increase in collateral resistance and infarct volume (eg, 6- and 3-fold, respectively, in 24-month-old mice) after artery occlusion. This was not associated with rarefaction of similarly sized arterioles. Collateral remodeling was also reduced. CONCLUSIONS: Our findings demonstrate that aging causes rarefaction and insufficiency of the collateral circulation in multiple tissues, resulting in more severe ischemic tissue injury.


Asunto(s)
Envejecimiento/patología , Envejecimiento/fisiología , Circulación Colateral , Isquemia/patología , Isquemia/fisiopatología , Animales , Encéfalo/irrigación sanguínea , Infarto Encefálico/patología , Infarto Encefálico/fisiopatología , Modelos Animales de Enfermedad , Arteria Femoral/lesiones , Miembro Posterior/irrigación sanguínea , Masculino , Ratones , Ratones Endogámicos C57BL , Neovascularización Fisiológica , Óxido Nítrico Sintasa de Tipo III/metabolismo , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Transducción de Señal , Resistencia Vascular
9.
Am J Physiol Heart Circ Physiol ; 300(6): H2027-34, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21398592

RESUMEN

Blood flow restoration to ischemic tissue is affected by various risk factors. The aim of this study was to examine gender effects on arteriogenesis and angiogenesis in a mouse ischemic hindlimb model. C57BL/6J mice were subjected to unilateral hindlimb ischemia. Flow recovery was less and hindlimb use impairment was greater in females. No gender difference in vessel number was found at baseline, although 7 days postsurgery females had fewer α-smooth muscle actin-positive vessels in the midpoint of the adductor region. Females had higher hindlimb vascular resistance, were less responsive to vasodilators, and were more sensitive to vasoconstrictors postligation. Western blotting showed that females had higher baseline levels of vascular endothelial growth factor (VEGF) and endothelial nitric oxide synthase (eNOS) in the calf, while 7 days postligation males had higher levels of VEGF, eNOS, and phosphorylated vasodilator stimulated phosphoprotein. Females had less angiogenesis in a Matrigel plug assay and less endothelial cell proliferation in vitro. Females have impaired recovery of flow, a finding presumably caused by multiple factors including decreased collateral remodeling, less angiogenesis, impaired vasodilator response, and increased vasoconstrictor activity; our results also suggest the possibility that new collateral formation, from capillaries, is impaired in females.


Asunto(s)
Arteria Femoral/fisiología , Miembro Posterior/irrigación sanguínea , Isquemia/fisiopatología , Flujo Sanguíneo Regional/fisiología , Caracteres Sexuales , Animales , Femenino , Miembro Posterior/metabolismo , Isquemia/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Neovascularización Fisiológica/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Resistencia Vascular/fisiología
10.
Arterioscler Thromb Vasc Biol ; 30(3): 477-82, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20056912

RESUMEN

OBJECTIVE: In a previous study we identified metallothionein (MT) as a candidate gene potentially influencing collaterogenesis. In this investigation, we determined the effect of MT on collaterogenesis and examined the mechanisms contributing to the effects we found. METHODS AND RESULTS: Collateral blood flow recovery was assessed using laser Doppler perfusion imaging, and angiogenesis was measured using a Matrigel plug assay. Smooth muscle cells were isolated from MT knockout (KO) mice for functional assays. Gene expression of matrix metalloproteinase-9, platelet-derived growth factor, vascular endothelial growth factor, and Fat cadherin in smooth muscle cells was measured by real-time polymerase chain reaction, and protein levels of vascular endothelial growth factor and matrix metalloproteinase-9 were determined using enzyme-linked immunosorbent assay and Western blot. CD11b(+) macrophages were tested for invasiveness using a real-time impedance assay. Both flow recovery and angiogenesis were impaired in MT KO mice. Proliferation, migration, and invasion were decreased in MT KO smooth muscle cells, and matrix metalloproteinase-9, platelet-derived growth factor, and vascular endothelial growth factor expression were also decreased, whereas FAT-1 cadherin expression was elevated. MT KO CD11b(+) cells were more invasive than wild-type cells. CONCLUSIONS: MT plays an important role in collateral flow recovery and angiogenesis, an activity that appears to be mediated, in part, by the effects of MT on the functionality of 3 cell types essential for these processes: endothelial cells, smooth muscle cells, and macrophages.


Asunto(s)
Arterias/crecimiento & desarrollo , Macrófagos/fisiología , Metalotioneína/fisiología , Músculo Liso Vascular/fisiología , Neovascularización Fisiológica/fisiología , Animales , Arterias/citología , Movimiento Celular/fisiología , Proliferación Celular , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/fisiología , Miembro Posterior/irrigación sanguínea , Macrófagos/citología , Masculino , Metalotioneína/genética , Ratones , Ratones Noqueados , Modelos Animales , Músculo Liso Vascular/citología , Flujo Sanguíneo Regional/fisiología
11.
Atherosclerosis ; 324: 133-140, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33781566

RESUMEN

BACKGROUND AND AIMS: Increased receptor tyrosine kinase (RTK) activity has been historically linked to atherosclerosis. Paradoxically, we recently found that global deficiency in c-Kit function increased atherosclerosis in hyperlipidemic mice. This study aimed to investigate if such unusual atheroprotective phenotype depends upon c-Kit's function in smooth muscle cells (SMC). METHODS: We studied atherosclerosis in a SMC-specific conditional knockout mice (KitSMC) and control littermate. Tamoxifen (TAM) and vehicle treated mice were fed high fat diet for 16 weeks before atherosclerosis assessment in the whole aorta using oil red staining. Smooth muscle cells were traced within the aortic sinus of conditional c-Kit tracing mice (KitSMC eYFP) and their control littermates (KitWT eYFP) by immunofluorescent confocal microscopy. We then performed RNA sequencing on primary SMC from c-Kit deficient and control mice, and identified significantly altered genes and pathways as a result of c-Kit deficiency in SMC. RESULTS: Atherosclerosis significantly increased in KitSMC mice with respect to control groups. In addition, the loss of c-Kit in SMC increased plaque size and necrotic core area in the aortic sinus of hyperlipidemic mice. Smooth muscle cells from KitSMC eYFP mice were more prone to migrate and express foam cell markers (e.g., Mac2 and MCAM) than those from control littermate animals. RNAseq analysis showed a significant upregulation in genes associated with cell proliferation, migration, lipid metabolism, and inflammation secondary to the loss of Kit function in primary SMCs. CONCLUSIONS: Loss of c-Kit increases SMC migration, proliferation, and expression of foam cell markers in atherosclerotic plaques from hyperlipidemic mice.


Asunto(s)
Aterosclerosis , Placa Aterosclerótica , Animales , Aorta , Aterosclerosis/genética , Aterosclerosis/prevención & control , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso
12.
JVS Vasc Sci ; 2: 20-32, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34617055

RESUMEN

OBJECTIVE: Novel therapeutic angiogenic concepts for critical limb ischemia are still needed for limb salvage. E-selectin, a cell-adhesion molecule, is vital for recruitment of the stem/progenitor cells necessary for neovascularization in ischemic tissues. We hypothesized that priming ischemic limb tissue with E-selectin/adeno-associated virus (AAV) gene therapy, in a murine hindlimb ischemia and gangrene model, would increase therapeutic angiogenesis and improve gangrene. METHODS: FVB/NJ mice were given intramuscular hindlimb injections of either E-selectin/AAV or LacZ/AAV and then underwent induction of gangrene via femoral artery ligation and concomitant systemic injections of the nitric oxide synthesis inhibitor L-NAME (L-NG-Nitro arginine methyl ester; 40 mg/kg). Gangrene was evaluated via the Faber hindlimb appearance score. The rate of ischemic limb reperfusion and ischemic tissue angiogenesis were evaluated using laser Doppler perfusion imaging and DiI perfusion with confocal laser scanning microscopy of the ischemic footpads, respectively. The treadmill exhaustion test was performed on postoperative day (POD) 8 to determine hindlimb functionality. RESULTS: The E-selectin/AAV-treated mice (n = 10) had decreased Faber ischemia scores compared with those of the LacZ/AAV-treated mice (n = 7) at both PODs 7 and 14 (P < .05 and P < .01, respectively), improved laser Doppler perfusion imaging reperfusion indexes by POD 14 (P < .01), and greater gangrene footpad capillary density (P < .001). E-selectin/AAV-treated mice also had improved exercise tolerance (P < .05) and lower relative muscular atrophy (P < .01). CONCLUSION: We surmised that E-selectin/AAV gene therapy would significantly promote hindlimb angiogenesis, reperfusion, and limb functionality in mice with hindlimb ischemia and gangrene. Our findings highlight the reported novel gene therapy approach to critical limb ischemia as a potential therapeutic option for future clinical studies.

13.
Front Cardiovasc Med ; 8: 826687, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35174227

RESUMEN

AIMS: Novel cell-based therapeutic angiogenic treatments for patients with critical limb ischemia may afford limb salvage. Mesenchymal stem cells (MSCs) do not overexpress E-selectin; however, we have previously demonstrated the cell-adhesion molecule's vital role in angiogenesis and wound healing. Thus, we created a viral vector to overexpress E-selectin on MSCs to increase their therapeutic profile. METHODS AND RESULTS: Femoral artery ligation induced hind limb ischemia in mice and intramuscular injections were administered of vehicle or syngeneic donor MSCs, transduced ex vivo with an adeno-associated viral vector to express either GFP+ (MSCGFP) or E-selectin-GFP+ (MSCE-selectin-GFP). Laser Doppler Imaging demonstrated significantly restored reperfusion in MSCE-selectin-GFP-treated mice vs. controls. After 3 weeks, the ischemic limbs in mice treated with MSCE-selectin-GFP had increased footpad blood vessel density, hematoxylin and eosin stain (H&E) ischemic calf muscle sections revealed mitigated muscular atrophy with restored muscle fiber size, and mice were able to run further before exhaustion. PCR array-based gene profiling analysis identified nine upregulated pro-angiogenic/pro-repair genes and downregulated Tumor necrosis factor (TNF) gene in MSCE-selectin-GFP-treated limb tissues, indicating that the therapeutic effect is likely achieved via upregulation of pro-angiogenic cytokines and downregulation of inflammation. CONCLUSION: This innovative cell therapy confers increased limb reperfusion, neovascularization, improved functional recovery, decreased muscle atrophy, and thus offers a potential therapeutic method for future clinical studies.

14.
Cell Physiol Biochem ; 26(4-5): 577-86, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21063095

RESUMEN

This study tested the hypotheses that the hypertonic environment of the renal medulla regulates the expression of cystic fibrosis transmembrane conductance regulator protein (CFTR) and its natural splice variant, TNR-CFTR. To accomplish this, Madin-Darby canine kidney (MDCK) stable cell lines expressing TNR-CFTR or CFTR were used. The cells were treated with hypertonic medium made with either NaCl or urea or sucrose (480 mOsm/kg or 560 mOsm/kg) to mimic the tonicity of the renal medulla environment. Western blot data showed that CFTR and TNR-CFTR total cell protein is increased by hypertonic medium, but using the surface biotinylation technique, only CFTR was found to be increased in cell plasma membrane. Confocal microscopy showed TNR-CFTR localization primarily at the endoplasmic reticulum and plasma membrane. In conclusion, CFTR and TNR-CFTR have different patterns of distribution in MDCK cells and they are modulated by a hypertonic environment, suggesting their physiological importance in renal medulla.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Soluciones Hipertónicas/farmacología , Animales , Western Blotting , Línea Celular , Membrana Celular/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/análisis , Perros , Retículo Endoplásmico/metabolismo , Microscopía Confocal , Isoformas de Proteínas/análisis , Isoformas de Proteínas/metabolismo , Cloruro de Sodio/farmacología , Sacarosa/farmacología , Urea/farmacología
16.
Surgery ; 163(4): 877-882, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29287914

RESUMEN

BACKGROUND: Arteriogenesis is a process whereby collateral vessels remodel usually in response to increased blood flow and/or wall stress. Remodeling of collaterals can function as a natural bypass to alleviate ischemia during arterial occlusion. Here we used a genetic approach to investigate possible roles of tyrosine receptor c-Kit in arteriogenesis. METHODS: Mutant mice with loss of c-Kit function (KitW/W-v), and controls were subjected to hindlimb ischemia. Blood flow recovery was evaluated pre-, post-, and weekly after ischemia. Foot ischemic damage and function were assessed between days 1 to 14 post-ischemia while collaterals remodeling were measured 28 days post-ischemia. Both groups of mice also were subjected to wild type bone marrow cells transplantation 3 weeks before hindlimb ischemia to evaluate possible contributions of defective bone marrow c-Kit expression on vascular recovery. RESULTS: KitW/W-v mice displayed impaired blood flow recovery, greater ischemic damage and foot dysfunction after ischemia compared to controls. KitW/W-v mice also demonstrated impaired collateral remodeling consistent with flow recovery findings. Because arteriogenesis is a biological process that involves bone marrow-derived cells, we investigated which source of c-Kit signaling (bone marrow or vascular) plays a major role in arteriogenesis. KitW/W-v mice transplanted with bone marrow wild type cells exhibited similar phenotype of impaired blood flow recovery, greater tissue ischemic damage and foot dysfunction as nontransplanted KitW/W-v mice. CONCLUSION: This study provides evidence that c-Kit signaling is required during arteriogenesis. Also, it strongly suggests a vascular role for c-Kit signaling because rescue of systemic c-Kit activity by bone marrow transplantation did not augment the functional recovery of KitW/W-v mouse hindlimbs.


Asunto(s)
Circulación Colateral/fisiología , Miembro Posterior/irrigación sanguínea , Isquemia/fisiopatología , Neovascularización Fisiológica/fisiología , Proteínas Proto-Oncogénicas c-kit/deficiencia , Animales , Biomarcadores/metabolismo , Trasplante de Médula Ósea , Miembro Posterior/fisiología , Miembro Posterior/fisiopatología , Isquemia/metabolismo , Isquemia/terapia , Masculino , Ratones , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal
17.
PeerJ ; 5: e3418, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28626608

RESUMEN

BACKGROUND: c-Kit is a receptor tyrosine kinase present in multiple cell types, including vascular smooth muscle cells (SMC). However, little is known about how c-Kit influences SMC biology and vascular pathogenesis. METHODS: High-throughput microarray assays and in silico pathway analysis were used to identify differentially expressed genes between primary c-Kit deficient (KitW/W-v) and control (Kit+/+) SMC. Quantitative real-time RT-PCR and functional assays further confirmed the differences in gene expression and pro-inflammatory pathway regulation between both SMC populations. RESULTS: The microarray analysis revealed elevated NF-κB gene expression secondary to the loss of c-Kit that affects both the canonical and alternative NF-κB pathways. Upon stimulation with an oxidized phospholipid as pro-inflammatory agent, c-Kit deficient SMC displayed enhanced NF-κB transcriptional activity, higher phosphorylated/total p65 ratio, and increased protein expression of NF-κB regulated pro-inflammatory mediators with respect to cells from control mice. The pro-inflammatory phenotype of mutant cells was ameliorated after restoring c-Kit activity using lentiviral transduction. Functional assays further demonstrated that c-Kit suppresses NF-κB activity in SMC in a TGFß-activated kinase 1 (TAK1) and Nemo-like kinase (NLK) dependent manner. DISCUSSION: Our study suggests a novel mechanism by which c-Kit suppresses NF-κB regulated pathways in SMC to prevent their pro-inflammatory transformation.

18.
J Cardiovasc Transl Res ; 7(8): 749-55, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25315467

RESUMEN

Chronic stress is associated with increased risk of cardiovascular diseases. Aging is also associated with vascular dysfunction. We hypothesize that chronic stress accelerates collateral dysfunction in old mice. Mice were subjected to either chronic social defeat (CSD) or chronic cold stress (CCS). The CSD mice were housed in a box inside an aggressor's cage and exposed to the aggressor. The CCS group was placed in iced water. After chronic stress, mice underwent femoral artery ligation (FAL) and flow recovery was measured. For the CSD group, appearance and use scores of the foot and a behavioral test were performed. CSD impaired collateral flow recovery after FAL. Further, stressed mice had greater ischemic damage, impaired foot function, and altered behavior. The CCS mice also showed impaired collateral flow recovery. Chronic stress causes hind limb collateral dysfunction in old mice, a conclusion reinforced by the fact that two types of stress produced similar changes.


Asunto(s)
Circulación Colateral , Arteria Femoral/fisiopatología , Isquemia/fisiopatología , Estrés Psicológico/fisiopatología , Animales , Conducta Animal , Enfermedad Crónica , Frío , Modelos Animales de Enfermedad , Arteria Femoral/cirugía , Isquemia/etiología , Isquemia/psicología , Ligadura , Masculino , Ratones Endogámicos C57BL , Recuperación de la Función , Flujo Sanguíneo Regional , Conducta Social , Estrés Psicológico/etiología , Estrés Psicológico/psicología , Factores de Tiempo
19.
Dis Model Mech ; 6(2): 323-31, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23324329

RESUMEN

The primary purpose of this investigation was to determine whether ApoE(-/-) mice, when subjected to chronic stress, exhibit lesions characteristic of human vulnerable plaque and, if so, to determine the time course of such changes. We found that the lesions were remarkably similar to human vulnerable plaque, and that the time course of lesion progression raised interesting insights into the process of plaque development. Lard-fed mixed-background ApoE(-/-) mice exposed to chronic stress develop lesions with large necrotic core, thin fibrous cap and a high degree of inflammation. Neovascularization and intraplaque hemorrhage are observed in over 80% of stressed animals at 20 weeks of age. Previously described models report a prevalence of only 13% for neovascularization observed at a much later time point, between 36 and 60 weeks of age. Thus, our new stress-induced model of advanced atherosclerotic plaque provides an improvement over what is currently available. This model offers a tool to further investigate progression of plaque phenotype to a more vulnerable phenotype in humans. Our findings also suggest a possible use of this stress-induced model to determine whether therapeutic interventions have effects not only on plaque burden, but also, and importantly, on plaque vulnerability.


Asunto(s)
Aterosclerosis/etiología , Aterosclerosis/patología , Modelos Animales de Enfermedad , Placa Aterosclerótica/etiología , Placa Aterosclerótica/patología , Estrés Psicológico/complicaciones , Animales , Aterosclerosis/sangre , Aterosclerosis/complicaciones , Presión Sanguínea , Colesterol/sangre , Estenosis Coronaria/complicaciones , Estenosis Coronaria/patología , Corticosterona/sangre , Hemorragia/complicaciones , Hemorragia/patología , Humanos , Inflamación/sangre , Inflamación/complicaciones , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Necrosis , Neovascularización Patológica/complicaciones , Neovascularización Patológica/patología , Neuropéptido Y/sangre , Placa Aterosclerótica/complicaciones , Estrés Psicológico/sangre
20.
J Cardiovasc Transl Res ; 4(6): 779-89, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21538183

RESUMEN

Despite positive animal studies, clinical angiogenesis trials have been disappointing, possibly due to risk factors present in humans but usually unexplored in animals. We recently demonstrated aging causes impaired collateral remodeling and collateral dropout; here, we investigate potential mechanisms responsible for these findings. Four-, 10-, and 18-month-C57BL/6J mice were subjected to femoral artery ligation; flow was measured using laser Doppler perfusion imaging. Endothelial nitric oxide synthase (eNOS) and phosphorylated eNOS were measured in calf muscle. Apoptosis was assessed in endothelial (EC) and smooth muscle (SMC) cells isolated from young and old mice. Angiogenesis was measured using a Matrigel plug assay. Lethally irradiated young and old mice received bone marrow cells (BMC) from either young or old donors and were subjected to femoral artery ligation (FAL). BMC mobilization and homing were assessed. Flow recovery was impaired and less eNOS and phosphorylated eNOS was present in older vs. young mice (p < 0.001 and p = 0.015, respectively). ECs and SMCs from older mice were more sensitive to an apoptotic stimulus, but were rescued by NO-enhancing drugs. In older mice, angiogenesis (Matrigel plug assay) was impaired, as was mobilization and homing of BM progenitor cells following FAL. Although both mobilization and homing improved when older mice received BMC transplantation from young donors, flow recovery failed to improve. Aging impairs BMC mobilization and homing, collateral responsiveness to angiogenic stimuli, and increases EC and SMC susceptibility to apoptosis via dysfunctional eNOS signaling. The latter could contribute to impaired remodeling and collateral dropout. These finding identify potential obstacles to therapeutic interventions in elderly patients.


Asunto(s)
Envejecimiento/metabolismo , Apoptosis , Arteriopatías Oclusivas/fisiopatología , Circulación Colateral , Extremidad Inferior/irrigación sanguínea , Neovascularización Fisiológica , Óxido Nítrico Sintasa de Tipo III/metabolismo , Transducción de Señal , Factores de Edad , Envejecimiento/patología , Animales , Aorta/enzimología , Aorta/patología , Aorta/fisiopatología , Arteriopatías Oclusivas/enzimología , Arteriopatías Oclusivas/patología , Western Blotting , Trasplante de Médula Ósea , Movimiento Celular , Células Cultivadas , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Femenino , Arteria Femoral/cirugía , Flujometría por Láser-Doppler , Ligadura , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/enzimología , Imagen de Perfusión/métodos , Fosforilación , Flujo Sanguíneo Regional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA