Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Biochem Biophys Res Commun ; 409(4): 758-63, 2011 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-21621512

RESUMEN

Uremic toxins such as indoxyl sulfate (IS) accumulate at a high level in end stage renal disease (ESRD) and can exhibit significant systemic endothelial toxicity leading to accelerated cardiovascular events. The precise molecular mechanisms by which IS causes endothelial dysfunction are unknown. We tested the hypothesis that IS negatively influences properties of endothelial cells, such as migration and tube formation, by depleting nitric oxide (NO) bioavailability, and that an NO donor can reverse these inhibitory effects. IS inhibited human umbilical vein endothelial cell (HUVEC) migration and formation of tubes on matrigel. Mechanistically, IS inhibited VEGF-induced NO release from HUVECs. An NO donor, SNAP, reversed IS-mediated inhibition of HUVEC migration as well as tube-formation. IS inhibited ERK 1/2 MAP kinase activity in a dose-dependent manner, but this was preserved by SNAP. Inhibition of ERK 1/2 with a pharmacological inhibitor (U0126) decreased HUVEC migration and tube formation; these effects too were prevented by SNAP. Further, IS stimulated activation of myosin light chain (MLC), potentially stimulating endothelial contractility, while SNAP decreased MLC activation. Thus, we conclude that the negative effects of IS on endothelial cells are prevented, to a major extent, by NO, via its divergent actions on ERK MAP kinase and MLC.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Indicán/antagonistas & inhibidores , Cadenas Ligeras de Miosina/metabolismo , Óxido Nítrico/metabolismo , Uremia/metabolismo , Células Cultivadas , Células Endoteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Humanos , Indicán/metabolismo , Indicán/toxicidad , Cadenas Ligeras de Miosina/antagonistas & inhibidores , Donantes de Óxido Nítrico/farmacología , S-Nitroso-N-Acetilpenicilamina/farmacología
2.
A A Case Rep ; 7(3): 60-2, 2016 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-27258181

RESUMEN

In 2010, our department instituted a nonclinical, administrative rotation in operating room management for anesthesiology residents. Subsequently, we mandated the rotation for all senior anesthesiology residents in 2013. In 2014, under the auspices of the American Society of Anesthesiologists, we developed a web-based module covering the basics of finance, accounting, and operating room management. A multiple-choice test was given to residents at the beginning and end of the rotation, and we compared the mean scores between residents who took the traditional course and residents who took the web-based module. We found no significant difference between the groups of residents, suggesting that the web-based module is as effective as traditional didactics.


Asunto(s)
Anestesiología/educación , Competencia Clínica , Internet , Internado y Residencia/métodos , Sistemas de Información en Quirófanos , Quirófanos/métodos , Citas y Horarios , Humanos , Sistemas de Información en Quirófanos/normas , Quirófanos/normas
3.
J Am Heart Assoc ; 5(1)2016 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-26738788

RESUMEN

BACKGROUND: Circulating angiogenic cells (CACs) are peripheral blood cells whose functional capacity inversely correlates with cardiovascular risk and that have therapeutic benefits in animal models of cardiovascular disease. However, donor age and disease state influence the efficacy of autologous cell therapy. We sought to determine whether age or coronary artery disease (CAD) impairs the therapeutic potential of CACs for myocardial infarction (MI) and whether the use of ex vivo gene therapy to overexpress endothelial nitric oxide (NO) synthase (eNOS) overcomes these defects. METHODS AND RESULTS: We recruited 40 volunteers varying by sex, age (< or ≥45 years), and CAD and subjected their CACs to well-established functional tests. Age and CAD were associated with reduced CAC intrinsic migration (but not specific response to vascular endothelial growth factor, adherence of CACs to endothelial tubes, eNOS mRNA and protein levels, and NO production. To determine how CAC function influences therapeutic potential, we injected the 2 most functional and the 2 least functional CAC isolates into mouse hearts post MI. The high-function isolates substantially improved cardiac function, whereas the low-function isolates led to cardiac function only slightly better than vehicle control. Transduction of the worst isolate with eNOS cDNA adenovirus increased NO production, migration, and cardiac function of post-MI mice implanted with the CACs. Transduction of the best isolate with eNOS small interfering RNA adenovirus reduced all of these capabilities. CONCLUSIONS: Age and CAD impair multiple functions of CACs and limit therapeutic potential for the treatment of MI. eNOS gene therapy in CACs from older donors or those with CAD has the potential to improve autologous cell therapy outcomes.


Asunto(s)
Enfermedad de la Arteria Coronaria/enzimología , Células Progenitoras Endoteliales/enzimología , Células Progenitoras Endoteliales/trasplante , Infarto del Miocardio/cirugía , Neovascularización Fisiológica , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Trasplante de Células Madre/métodos , Adulto , Anciano , Animales , Estudios de Casos y Controles , Movimiento Celular , Células Cultivadas , Técnicas de Cocultivo , Enfermedad de la Arteria Coronaria/diagnóstico , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones SCID , Persona de Mediana Edad , Infarto del Miocardio/enzimología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Óxido Nítrico Sintasa de Tipo III/genética , Fenotipo , Interferencia de ARN , Recuperación de la Función , Regeneración , Transducción de Señal , Factores de Tiempo , Transducción Genética , Transfección
4.
J Appl Physiol (1985) ; 114(6): 752-60, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23349456

RESUMEN

Nitric oxide (NO) exerts a wide range of cellular effects in the cardiovascular system. NO is short lived, but S-nitrosoglutathione (GSNO) functions as a stable intracellular bioavailable NO pool. Accordingly, increased levels can facilitate NO-mediated processes, and conversely, catabolism of GSNO by the regulatory enzyme GSNO reductase (GSNOR) can impair these processes. Because dysregulated GSNOR can interfere with processes relevant to cardiovascular health, it follows that inhibition of GSNOR may be beneficial. However, the effect of GSNOR inhibition on vascular activity is unknown. To study the effects of GSNOR inhibition on endothelial function, we treated rats with a small-molecule inhibitor of GSNOR (N6338) that has vasodilatory effects on isolated aortic rings and assessed effects on arterial flow-mediated dilation (FMD), an NO-dependent process. GSNOR inhibition with a single intravenous dose of N6338 preserved FMD (15.3 ± 5.4 vs. 14.2 ± 6.3%, P = nonsignificant) under partial NO synthase inhibition that normally reduces FMD by roughly 50% (14.1 ± 2.9 vs. 7.6 ± 4.4%, P < 0.05). In hypertensive rats, daily oral administration of N6338 for 14 days reduced blood pressure (170.0 ± 5.3/122.7 ± 6.4 vs. 203.8 ± 1.9/143.7 ± 7.5 mmHg for vehicle, P < 0.001) and vascular resistance index (1.5 ± 0.4 vs. 3.2 ± 1.0 mmHg · min · l(-1) for vehicle, P < 0.001), and restored FMD from an initially impaired state (7.4 ± 1.7%, day 0) to a level (13.0 ± 3.1%, day 14, P < 0.001) similar to that observed in normotensive rats. N6338 also reversed the pathological kidney changes exhibited by the hypertensive rats. GSNOR inhibition preserves FMD under conditions of impaired NO production and protects against both microvascular and conduit artery dysfunction in a model of hypertension.


Asunto(s)
Aldehído Oxidorreductasas/antagonistas & inhibidores , Antihipertensivos/farmacología , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Arteria Femoral/efectos de los fármacos , Hipertensión/tratamiento farmacológico , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Administración Oral , Aldehído Oxidorreductasas/metabolismo , Animales , Antihipertensivos/administración & dosificación , Presión Sanguínea/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Endotelio Vascular/enzimología , Endotelio Vascular/fisiopatología , Inhibidores Enzimáticos/administración & dosificación , Arteria Femoral/enzimología , Arteria Femoral/fisiopatología , Humanos , Hipertensión/enzimología , Hipertensión/etiología , Hipertensión/patología , Hipertensión/fisiopatología , Inyecciones Intravenosas , Riñón/efectos de los fármacos , Riñón/patología , Ratones , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas , Ratas Endogámicas Dahl , Ratas Sprague-Dawley , Cloruro de Sodio Dietético , Factores de Tiempo , Vasodilatadores/administración & dosificación
5.
PLoS One ; 8(4): e61413, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23630585

RESUMEN

Pleiotrophin (PTN) is a growth factor with both pro-angiogenic and limited pro-tumorigenic activity. We evaluated the potential for PTN to be used for safe angiogenic gene therapy using the full length gene and a truncated gene variant lacking the domain implicated in tumorigenesis. Mouse myoblasts were transduced to express full length or truncated PTN (PTN or T-PTN), along with a LacZ reporter gene, and injected into mouse limb muscle and myocardium. In cultured myoblasts, PTN was expressed and secreted via the Golgi apparatus, but T-PTN was not properly secreted. Nonetheless, no evidence of uncontrolled growth was observed in cells expressing either form of PTN. PTN gene delivery to myocardium, and non-ischemic skeletal muscle, did not result in a detectable change in vascularity or function. In ischemic hindlimb at 14 days post-implantation, intramuscular injection with PTN-expressing myoblasts led to a significant increase in skin perfusion and muscle arteriole density. We conclude that (1) delivery of the full length PTN gene to muscle can be accomplished without tumorigenesis, (2) the truncated PTN gene may be difficult to use in a gene therapy context due to inefficient secretion, (3) PTN gene delivery leads to functional benefit in the mouse acute ischemic hindlimb model.


Asunto(s)
Proteínas Portadoras/genética , Citocinas/genética , Terapia Genética , Isquemia/terapia , Músculo Esquelético/irrigación sanguínea , Fragmentos de Péptidos/genética , Animales , Proteínas Portadoras/metabolismo , Movimiento Celular , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Vasos Coronarios/patología , Citocinas/metabolismo , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Músculo Esquelético/patología , Mioblastos/metabolismo , Mioblastos/trasplante , Miocardio/patología , Miocitos del Músculo Liso/fisiología , Neovascularización Fisiológica , Fragmentos de Péptidos/metabolismo
6.
Am J Cardiovasc Dis ; 2(1): 12-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22254209

RESUMEN

BACKGROUND: Chronic or severe acute elevations in plasma glucose are associated with decreases in the number and function of circulating angiogenic cells (CACs). However, less is known about whether fasting plasma glucose levels (FPG) within the normal or pre-diabetic range among healthy individuals are associated with decreased CAC function. Establishing this relationship is an important step in developing a line of research that may ultimately lead to preventative lifestyle interventions intended to maximize endogenous CAC function and reduce cardiometabolic disease risk. OBJECTIVES: 1) To examine whether increases in FPG are associated with decreases in CAC migration among healthy individuals with FPG levels below the threshold for hyperglycemia, and 2) to contrast effect of FPG on CAC migration toward a pro-angiogenic stimulus (vascular endothelial growth factor; VEGF) with effect on intrinsic cell migratory capacity (i.e., random migration with no stimulus). METHODS: 28 men and women ranging from 20-57 years of age and free of cardiovascular disease participated in a pilot study, involving a fasting blood draw for FPG and isolation of peripheral blood mononuclear cells. CAC migration toward VEGF and random cell migration (control) were assessed in vitro. VEGF-induced migration that was normalized to control migration, representing the VEGF-response component of chemotaxis independent of motility, was calculated to determine whether any impairment in migration to VEGF was due to lower specific response to VEGF or to lower non-specific migratory capacity. RESULTS: Increased levels of FPG were associated in a dose-response fashion with a significantly lower random migration under control conditions (CTRL: r= -.408, p=.031), no differences in migration to VEGF (r= -.039, p=.842) and a borderline association with VEGF-induced migration normalized to control migration (VEGF/CTRL: r=.349, p=.069). The relationship between FPG and random migration under control conditions remained significant when controlling for gender and body mass index (p's<.05), and became borderline significant when controlling for age (p=.062). CONCLUSIONS: Among healthy individuals, higher fasting glucose levels, despite falling below the diabetic range, are associated with decreased random CAC migration. These findings suggest a need for further studies investigating the effects of lifestyle or dietary interventions on glucose regulation and CAC function.

7.
J Tissue Sci Eng ; S32011 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-22675670

RESUMEN

Therapeutic results of clinical autologous bone marrow cell (BMC) therapy trials for cardiac disease have been modest compared to results of BMC implantation into rodent hearts post-myocardial infarction (MI). In clinical trials, autologous BMCs are typically harvested from older patients who have recently suffered an MI. In contrast, experimental studies in rodent models typically utilize donor BMCs isolated from young, healthy, inbred mice that are not the recipients. Using unfractionated BMCs from donor mice at ages of young, middle-aged, and old, we discovered that recipient left ventricular function post-MI was significantly improved by young donor BMC implantation but was only preserved by middle-aged donor BMCs. Notably, old donor BMCs did not slow the decline in recipient post-MI cardiac function, suggesting BMC impairment by advanced donor age. Furthermore, we also show here that BMCs that are therapeutically impaired by donor age can be further impaired by concurrent donor MI. In conclusion, our findings suggest that therapeutic impairment of BMCs by advanced age is one of the important factors that can limit the success of clinical autologous BMC-based therapy.

8.
Sci Transl Med ; 3(100): 100ra90, 2011 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-21918107

RESUMEN

Delivery of bone marrow cells (BMCs) to the heart has substantially improved cardiac function in most rodent models of myocardial infarction (MI), but clinical trials of BMC therapy have led to only modest improvements. Rodent models typically involve intramyocardial injection of BMCs from distinct donor individuals who are healthy. In contrast, autologous BMCs from individuals after MI are used for clinical trials. Using BMCs from donor mice after MI, we discovered that recent MI impaired BMC therapeutic efficacy. MI led to myocardial inflammation and an increased inflammatory state in the bone marrow, changing the BMC composition and reducing their efficacy. Injection of a general anti-inflammatory drug or a specific interleukin-1 inhibitor to donor mice after MI prevented this impairment. Our findings offer an explanation of why human trials have not matched the success of rodent experiments and suggest potential strategies to improve the success of clinical autologous BMC therapy.


Asunto(s)
Trasplante de Médula Ósea/métodos , Interleucina-1/metabolismo , Infarto del Miocardio/inmunología , Infarto del Miocardio/terapia , Donantes de Tejidos , Animales , Ecocardiografía , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA