Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Eur J Pharm Biopharm ; 184: 83-91, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36693545

RESUMEN

Nanomedicine offers great potential for the treatment of cardiovascular disease and particulate systems have the capacity to markedly improve bioavailability of therapeutics. The delivery of pro-angiogenic hepatocyte growth factor (HGF) and pro-survival and pro-myogenic insulin-like growth factor (IGF-1) encapsulated in Alginate-Sulfate nanoparticles (AlgS-NP) might improve left ventricular (LV) functional recovery after myocardial infarction (MI). In a porcine ischemia-reperfusion model, MI is induced by 75 min balloon occlusion of the mid-left anterior descending coronary artery followed by reperfusion. After 1 week, pigs (n = 12) with marked LV-dysfunction (LV ejection fraction, LVEF < 45%) are randomized to fusion imaging-guided intramyocardial injections of 8 mg AlgS-NP prepared with 200 µg HGF and IGF-1 (HGF/IGF1-NP) or PBS (Control). Intramyocardial injection is safe and pharmacokinetic studies of Cy5-labeled NP confirm superior cardiac retention compared to intracoronary infusion. Seven weeks after intramyocardial-injection of HGF/IGF1-NP, infarct size, measured using magnetic resonance imaging, is significantly smaller than in controls and is associated with increased coronary flow reserve. Importantly, HGF/IGF1-NP-treated pigs show significantly increased LVEF accompanied by improved myocardial remodeling. These findings demonstrate the feasibility and efficacy of using AlgS-NP as a delivery system for growth factors and offer the prospect of innovative treatment for refractory ischemic cardiomyopathy.


Asunto(s)
Infarto del Miocardio , Daño por Reperfusión Miocárdica , Nanopartículas , Animales , Modelos Animales de Enfermedad , Factor de Crecimiento de Hepatocito , Factor I del Crecimiento Similar a la Insulina , Sulfatos , Porcinos
2.
Cardiovasc Res ; 118(5): 1262-1275, 2022 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33909875

RESUMEN

AIMS: Hepatic capillaries are lined with specialized liver sinusoidal endothelial cells (LSECs) which support macromolecule passage to hepatocytes and prevent fibrosis by keeping hepatic stellate cells (HSCs) quiescent. LSEC specialization is co-determined by transcription factors. The zinc-finger E-box-binding homeobox (Zeb)2 transcription factor is enriched in LSECs. Here, we aimed to elucidate the endothelium-specific role of Zeb2 during maintenance of the liver and in liver fibrosis. METHODS AND RESULTS: To study the role of Zeb2 in liver endothelium we generated EC-specific Zeb2 knock-out (ECKO) mice. Sequencing of liver EC RNA revealed that deficiency of Zeb2 results in prominent expression changes in angiogenesis-related genes. Accordingly, the vascular area was expanded and the presence of pillars inside ECKO liver vessels indicated that this was likely due to increased intussusceptive angiogenesis. LSEC marker expression was not profoundly affected and fenestrations were preserved upon Zeb2 deficiency. However, an increase in continuous EC markers suggested that Zeb2-deficient LSECs are more prone to dedifferentiation, a process called 'capillarization'. Changes in the endothelial expression of ligands that may be involved in HSC quiescence together with significant changes in the expression profile of HSCs showed that Zeb2 regulates LSEC-HSC communication and HSC activation. Accordingly, upon exposure to the hepatotoxin carbon tetrachloride (CCl4), livers of ECKO mice showed increased capillarization, HSC activation, and fibrosis compared to livers from wild-type littermates. The vascular maintenance and anti-fibrotic role of endothelial Zeb2 was confirmed in mice with EC-specific overexpression of Zeb2, as the latter resulted in reduced vascularity and attenuated CCl4-induced liver fibrosis. CONCLUSION: Endothelial Zeb2 preserves liver angioarchitecture and protects against liver fibrosis. Zeb2 and Zeb2-dependent genes in liver ECs may be exploited to design novel therapeutic strategies to attenuate hepatic fibrosis.


Asunto(s)
Células Endoteliales , Cirrosis Hepática , Animales , Biomarcadores/metabolismo , Células Endoteliales/metabolismo , Endotelio , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Hígado/metabolismo , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Cirrosis Hepática/prevención & control , Ratones
3.
JACC Basic Transl Sci ; 6(8): 631-646, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34466750

RESUMEN

The bacterial C-type lectin domain family 4 member E (CLEC4E) has an important role in sterile inflammation, but its role in myocardial repair is unknown. Using complementary approaches in porcine, murine, and human samples, we show that CLEC4E expression levels in the myocardium and in blood correlate with the extent of myocardial injury and left ventricular (LV) functional impairment. CLEC4E expression is markedly increased in the vasculature, cardiac myocytes, and infiltrating leukocytes in the ischemic heart. Loss of Clec4e signaling is associated with reduced acute cardiac injury, neutrophil infiltration, and infarct size. Reduced myocardial injury in Clec4e -/- translates into significantly improved LV structural and functional remodeling at 4 weeks' follow-up. The early transcriptome of LV tissue from Clec4e -/- mice versus wild-type mice reveals significant upregulation of transcripts involved in myocardial metabolism, radical scavenging, angiogenesis, and extracellular matrix organization. Therefore, targeting CLEC4E in the early phase of ischemia-reperfusion injury is a promising therapeutic strategy to modulate myocardial inflammation and enhance repair after ischemia-reperfusion injury.

4.
Circ Heart Fail ; 14(1): e006979, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33464950

RESUMEN

BACKGROUND: Chronic pressure overload predisposes to heart failure, but the pathogenic role of microvascular endothelial cells (MiVEC) remains unknown. We characterized transcriptional, metabolic, and functional adaptation of cardiac MiVEC to pressure overload in mice and patients with aortic stenosis (AS). METHODS: In Tie2-Gfp mice subjected to transverse aortic constriction or sham surgery, we performed RNA sequencing of isolated cardiac Gfp+-MiVEC and validated the signature in freshly isolated MiVEC from left ventricle outflow tract and right atrium of patients with AS. We next compared their angiogenic and metabolic profiles and finally correlated molecular and pathological signatures with clinical phenotypes of 42 patients with AS (50% women). RESULTS: In mice, transverse aortic constriction induced progressive systolic dysfunction, fibrosis, and reduced microvascular density. After 10 weeks, 25 genes predominantly involved in matrix-regulation were >2-fold upregulated in isolated MiVEC. Increased transcript levels of Cartilage Intermediate Layer Protein (Cilp), Thrombospondin-4, Adamtsl-2, and Collagen1a1 were confirmed by quantitative reverse transcription polymerase chain reaction and recapitulated in left ventricle outflow tract-derived MiVEC of AS (P<0.05 versus right atrium-MiVEC). Fatty acid oxidation increased >2-fold in left ventricle outflow tract-MiVEC, proline content by 130% (median, IQR, 58%-474%; P=0.008) and procollagen secretion by 85% (mean [95% CI, 16%-154%]; P<0.05 versus right atrium-MiVEC for all). The altered transcriptome in left ventricle outflow tract-MiVEC was associated with impaired 2-dimensional-vascular network formation and 3-dimensional-spheroid sprouting (P<0.05 versus right atrium-MiVEC), profibrotic ultrastructural changes, and impaired diastolic left ventricle function, capillary density and functional status, especially in female AS. CONCLUSIONS: Pressure overload induces major transcriptional and metabolic adaptations in cardiac MiVEC resulting in excess interstitial fibrosis and impaired angiogenesis. Molecular rewiring of MiVEC is worse in women, compromises functional status, and identifies novel targets for intervention.


Asunto(s)
Estenosis de la Válvula Aórtica/genética , Vasos Coronarios/metabolismo , Células Endoteliales/metabolismo , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/metabolismo , Microvasos/metabolismo , Proteínas ADAMTS/genética , Anciano , Animales , Aorta , Estenosis de la Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/cirugía , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Constricción Patológica , Vasos Coronarios/patología , Modelos Animales de Enfermedad , Células Endoteliales/patología , Proteínas de la Matriz Extracelular/genética , Ácidos Grasos/metabolismo , Femenino , Perfilación de la Expresión Génica , Atrios Cardíacos/patología , Implantación de Prótesis de Válvulas Cardíacas , Ventrículos Cardíacos/patología , Humanos , Masculino , Ratones , Ratones Transgénicos , Densidad Microvascular , Microvasos/patología , Procolágeno/metabolismo , Prolina/metabolismo , Pirofosfatasas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ARN , Trombospondinas/genética
5.
Stem Cell Res Ther ; 11(1): 389, 2020 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-32894199

RESUMEN

BACKGROUND: Cell therapy has been evaluated pre-clinically and clinically as a means to improve wound vascularization and healing. While translation of this approach to clinical practice ideally requires the availability of clinical grade xenobiotic-free cell preparations, studies proving the pre-clinical efficacy of the latter are mostly lacking. Here, the potential of xenobiotic-free human multipotent adult progenitor cell (XF-hMAPC®) preparations to promote vascularization was evaluated. METHODS: The potential of XF-hMAPC cells to support blood vessel formation was first scored in an in vivo Matrigel assay in mice. Next, a dose-response study was performed with XF-hMAPC cells in which they were tested for their ability to support vascularization and (epi) dermal healing in a physiologically relevant splinted wound mouse model. RESULTS: XF-hMAPC cells supported blood vessel formation in Matrigel by promoting the formation of mature (smooth muscle cell-coated) vessels. Furthermore, XF-hMAPC cells dose-dependently improved wound vascularization associated with increasing wound closure and re-epithelialization, granulation tissue formation, and dermal collagen organization. CONCLUSIONS: Here, we demonstrated that the administration of clinical-grade XF-hMAPC cells in mice represents an effective approach for improving wound vascularization and healing that is readily applicable for translation in humans.


Asunto(s)
Células Madre Adultas , Neovascularización Fisiológica , Animales , Tejido de Granulación , Ratones , Repitelización , Cicatrización de Heridas
6.
Cell Death Differ ; 27(5): 1520-1538, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31654035

RESUMEN

Defective cell migration causes delayed wound healing (WH) and chronic skin lesions. Autologous micrograft (AMG) therapies have recently emerged as a new effective and affordable treatment able to improve wound healing capacity. However, the precise molecular mechanism through which AMG exhibits its beneficial effects remains unrevealed. Herein we show that AMG improves skin re-epithelialization by accelerating the migration of fibroblasts and keratinocytes. More specifically, AMG-treated wounds showed improvement of indispensable events associated with successful wound healing such as granulation tissue formation, organized collagen content, and newly formed blood vessels. We demonstrate that AMG is enriched with a pool of WH-associated growth factors that may provide the starting signal for a faster endogenous wound healing response. This work links the increased cell migration rate to the activation of the extracellular signal-regulated kinase (ERK) signaling pathway, which is followed by an increase in matrix metalloproteinase expression and their extracellular enzymatic activity. Overall we reveal the AMG-mediated wound healing transcriptional signature and shed light on the AMG molecular mechanism supporting its potential to trigger a highly improved wound healing process. In this way, we present a framework for future improvements in AMG therapy for skin tissue regeneration applications.


Asunto(s)
Movimiento Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Trasplante de Piel , Cicatrización de Heridas , Animales , Movimiento Celular/genética , Células Cultivadas , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Queratinocitos/citología , Queratinocitos/enzimología , Sistema de Señalización de MAP Quinasas/genética , Metaloproteinasas de la Matriz/metabolismo , Ratones Endogámicos C57BL , Solubilidad , Transcripción Genética , Trasplante Autólogo , Cicatrización de Heridas/genética
7.
Nat Commun ; 8(1): 1249, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29093487

RESUMEN

Muscular dystrophies (MDs) are often characterized by impairment of both skeletal and cardiac muscle. Regenerative strategies for both compartments therefore constitute a therapeutic avenue. Mesodermal iPSC-derived progenitors (MiPs) can regenerate both striated muscle types simultaneously in mice. Importantly, MiP myogenic propensity is influenced by somatic lineage retention. However, it is still unknown whether human MiPs have in vivo potential. Furthermore, methods to enhance the intrinsic myogenic properties of MiPs are likely needed, given the scope and need to correct large amounts of muscle in the MDs. Here, we document that human MiPs can successfully engraft into the skeletal muscle and hearts of dystrophic mice. Utilizing non-invasive live imaging and selectively induced apoptosis, we report evidence of striated muscle regeneration in vivo in mice by human MiPs. Finally, combining RNA-seq and miRNA-seq data, we define miRNA cocktails that promote the myogenic potential of human MiPs.


Asunto(s)
Corazón/crecimiento & desarrollo , Células Madre Pluripotentes Inducidas/citología , Mesodermo/citología , MicroARNs/genética , Desarrollo de Músculos/genética , Músculo Esquelético/crecimiento & desarrollo , Distrofia Muscular Animal/patología , Miocardio/citología , Animales , Diferenciación Celular , Ecocardiografía , Corazón/diagnóstico por imagen , Humanos , Ratones , Músculo Esquelético/citología , Músculo Esquelético/diagnóstico por imagen , Músculo Esquelético/patología , Distrofia Muscular Animal/diagnóstico por imagen , Miocardio/patología , Regeneración
8.
J Cardiovasc Transl Res ; 10(4): 348-358, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28397162

RESUMEN

Angiogenic growth factor therapy for ischemic cardiovascular disease carries a risk of stimulating atherosclerotic plaque growth. We evaluated risk benefit ratio of sustained administration of recombinant human placental growth factor (rhPlGF)-2 in mice with advanced atherosclerosis and chronic ischemic cardiomyopathy. We maintained apolipoprotein E-deficient mice on a high cholesterol diet and induced myocardial infarction by transient ligation at 4 weeks. At 8 weeks, we assessed left ventricular (LV) function and randomized mice to receive rhPlGF-2 or vehicle (VEH) subcutaneously for 28 days. Administration of rhPlGF-2 significantly increased PlGF plasma levels without adverse hemodynamic or systemic inflammatory effects. RhPlGF-2 did not increase plaque area, composition, or vulnerability in the aortic arch. RhPlGF-2 significantly improved contractile function and reduced LV end-systolic and end-diastolic volume indices with a concomitant increase in capillary and arteriolar density in ischemic myocardium. RhPlGF-2 may represent a promising therapeutic strategy in chronic ischemic cardiomyopathy.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Aorta/efectos de los fármacos , Enfermedades de la Aorta/tratamiento farmacológico , Aterosclerosis/tratamiento farmacológico , Cardiomiopatías/tratamiento farmacológico , Infarto del Miocardio/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , Factor de Crecimiento Placentario/administración & dosificación , Función Ventricular Izquierda/efectos de los fármacos , Inductores de la Angiogénesis/toxicidad , Animales , Aorta/patología , Aorta/fisiopatología , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/fisiopatología , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/fisiopatología , Colesterol en la Dieta , Enfermedad Crónica , Modelos Animales de Enfermedad , Infusiones Subcutáneas , Masculino , Ratones Noqueados para ApoE , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/fisiopatología , Factor de Crecimiento Placentario/toxicidad , Placa Aterosclerótica , Proteínas Recombinantes/administración & dosificación , Recuperación de la Función , Volumen Sistólico/efectos de los fármacos , Factores de Tiempo , Rigidez Vascular/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
9.
Antioxid Redox Signal ; 26(4): 153-164, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27505125

RESUMEN

AIMS: The use of doxorubicin, a potent chemotherapeutic agent, is limited by cardiotoxicity. We tested the hypothesis that decreased soluble guanylate cyclase (sGC) enzyme activity contributes to the development of doxorubicin-induced cardiotoxicity. RESULTS: Doxorubicin administration (20 mg/kg, intraperitoneally [IP]) reduced cardiac sGC activity in wild-type (WT) mice. To investigate whether decreased sGC activity contributes to doxorubicin-induced cardiotoxicity, we studied mice with cardiomyocyte-specific deficiency of the sGC α1-subunit (mice with cardiomyocyte-specific deletion of exon 6 of the sGCα1 allele [sGCα1-/-CM]). After 12 weeks of doxorubicin administration (2 mg/kg/week IP), left ventricular (LV) systolic dysfunction was greater in sGCα1-/-CM than WT mice. To further assess whether reduced sGC activity plays a pathogenic role in doxorubicin-induced cardiotoxicity, we studied a mouse model in which decreased cardiac sGC activity was induced by cardiomyocyte-specific expression of a dominant negative sGCα1 mutant (DNsGCα1) upon doxycycline removal (Tet-off). After 8 weeks of doxorubicin administration, DNsGCα1tg/+, but not WT, mice displayed LV systolic dysfunction and dilatation. The difference in cardiac function and remodeling between DNsGCα1tg/+ and WT mice was even more pronounced after 12 weeks of treatment. Further impairment of cardiac function was attenuated when DNsGCα1 gene expression was inhibited (beginning at 8 weeks of doxorubicin treatment) by administering doxycycline. Furthermore, doxorubicin-associated reactive oxygen species generation was higher in sGCα1-deficient than WT hearts. Innovation and Conclusion: These data demonstrate that a reduction in cardiac sGC activity worsens doxorubicin-induced cardiotoxicity in mice and identify sGC as a potential therapeutic target. Various pharmacological sGC agonists are in clinical development or use and may represent a promising approach to limit doxorubicin-associated cardiotoxicity. Antioxid. Redox Signal. 26, 153-164.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Doxorrubicina/efectos adversos , Cardiopatías/etiología , Cardiopatías/metabolismo , Guanilil Ciclasa Soluble/sangre , Animales , Antibióticos Antineoplásicos/administración & dosificación , Cardiotoxicidad , Modelos Animales de Enfermedad , Doxorrubicina/administración & dosificación , Activación Enzimática/efectos de los fármacos , Expresión Génica , Cardiopatías/fisiopatología , Ratones , Ratones Noqueados , Mutación , Miocitos Cardíacos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Guanilil Ciclasa Soluble/deficiencia , Disfunción Ventricular
10.
J Am Heart Assoc ; 5(4): e002288, 2016 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-27091182

RESUMEN

BACKGROUND: Blood outgrowth endothelial cells (BOECs) mediate therapeutic neovascularization in experimental models, but outgrowth characteristics and functionality of BOECs from patients with ischemic cardiomyopathy (ICMP) are unknown. We compared outgrowth efficiency and in vitro and in vivo functionality of BOECs derived from ICMP with BOECs from age-matched (ACON) and healthy young (CON) controls. METHODS AND RESULTS: We isolated 3.6±0.6 BOEC colonies/100×10(6) mononuclear cells (MNCs) from 60-mL blood samples of ICMP patients (n=45; age: 66±1 years; LVEF: 31±2%) versus 3.5±0.9 colonies/100×10(6) MNCs in ACON (n=32; age: 60±1 years) and 2.6±0.4 colonies/100×10(6) MNCs in CON (n=55; age: 34±1 years), P=0.29. Endothelial lineage (VEGFR2(+)/CD31(+)/CD146(+)) and progenitor (CD34(+)/CD133(-)) marker expression was comparable in ICMP and CON. Growth kinetics were similar between groups (P=0.38) and not affected by left ventricular systolic dysfunction, maladaptive remodeling, or presence of cardiovascular risk factors in ICMP patients. In vitro neovascularization potential, assessed by network remodeling on Matrigel and three-dimensional spheroid sprouting, did not differ in ICMP from (A)CON. Secretome analysis showed a marked proangiogenic profile, with highest release of angiopoietin-2 (1.4±0.3×10(5) pg/10(6) ICMP-BOECs) and placental growth factor (5.8±1.5×10(3) pg/10(6) ICMP BOECs), independent of age or ischemic disease. Senescence-associated ß-galactosidase staining showed comparable senescence in BOECs from ICMP (5.8±2.1%; n=17), ACON (3.9±1.1%; n=7), and CON (9.0±2.8%; n=13), P=0.19. High-resolution microcomputed tomography analysis in the ischemic hindlimb of nude mice confirmed increased arteriogenesis in the thigh region after intramuscular injections of BOECs from ICMP (P=0.025; n=8) and CON (P=0.048; n=5) over vehicle control (n=8), both to a similar extent (P=0.831). CONCLUSIONS: BOECs can be successfully culture-expanded from patients with ICMP. In contrast to impaired functionality of ICMP-derived bone marrow MNCs, BOECs retain a robust proangiogenic profile, both in vitro and in vivo, with therapeutic potential for targeting ischemic disease.


Asunto(s)
Endotelio Vascular/fisiopatología , Isquemia Miocárdica/fisiopatología , Neovascularización Fisiológica/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Estudios de Casos y Controles , Proliferación Celular/fisiología , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/trasplante , Femenino , Humanos , Masculino , Ratones Desnudos , Persona de Mediana Edad , Estrés Oxidativo/fisiología , Adulto Joven
11.
Int J Cardiol ; 203: 534-42, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26569359

RESUMEN

OBJECTIVES: We investigated whether sustained infusion of recombinant human placental growth factor-2 (rhPlGF-2) improves myocardial perfusion and left ventricular (LV) function in a porcine model of ischemic cardiomyopathy (ICM). METHODS: We induced myocardial ischemia using a flow-limiting stent in the LAD. Four weeks later, we randomized pigs with confirmed myocardial dysfunction to blinded rhPlGF-2 administration (PlGF2, 15 µg/kg/day, 14 days) or PBS (CON). At 8 weeks, we measured hemodynamics, contractile function and regional perfusion at rest and during stress using MRI and microspheres. We evaluated neovascularization post mortem. RESULTS: RhPlGF-2 administration increased PlGF serum levels more than 63-fold (83 3 ± 361 versus 11 ± 5 pg/ml CON, P<0.05) without adverse effects. After 4weeks, rhPlGF-2 significantly enhanced perfusion in the ischemic region at rest (0.83 ± 0.32 versus 0.58 ± 0.21 ml/min/g CON, P<0.05) and during hyperemia (1.50 ± 0.50 versus 1.02 ± 0.46 ml/min/g CON, P<0.05). Consequently, regional contractile function in rhPlGF-2-treated pigs improved at rest (37 ± 15% versus 23 ± 9% CON, P<0.05) and during high dose dobutamine stress (53 ± 31% versus 27 ± 16% CON, P<0.05). Enhanced perfusion translated into a greater improvement in LV ejection fraction and in preload-recruitable stroke work in rhPlGF-2-treated animals than in CON (52 ± 11 versus 41 ± 9%, and 76 ± 24 versus 41 ± 21 mmHg, respectively, P<0.05 for both), which was associated with significantly greater vascular density in the ischemic region. CONCLUSIONS: In chronic ICM, systemic rhPlGF-2 administration significantly enhances regional myocardial perfusion, contractile function at rest and during stress, and induces a prominent recovery of global cardiac function. PlGF-2 protein infusion is safe and may represent a promising therapy in chronic ICM.


Asunto(s)
Proteínas de la Membrana/uso terapéutico , Isquemia Miocárdica/tratamiento farmacológico , Guías de Práctica Clínica como Asunto , Proteínas Recombinantes/uso terapéutico , Animales , Enfermedad Crónica , Cricetinae , Modelos Animales de Enfermedad , Femenino , Estudios de Seguimiento , Masculino , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/metabolismo , Miocardio/patología , Porcinos , Factores de Tiempo , Resultado del Tratamiento
12.
J Pharmacol Exp Ther ; 356(2): 284-92, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26621143

RESUMEN

Enhanced cyclic guanosine monophosphate (cGMP) signaling may attenuate myocardial ischemia-reperfusion injury (I/R) and improve left ventricular (LV) functional recovery after myocardial infarction (MI). We investigated the cardioprotection afforded by inhaled NO (iNO), the phosphodiesterase 5 (PDE5)-specific inhibitor tadalafil (TAD), or their combination (iNO+TAD) in C57Bl6J mice subjected to 6-minute left anterior descending artery ligation followed by reperfusion. We measured plasma and cardiac concentrations of cGMP during early reperfusion, quantified myocardial necrosis and inflammation by serial troponin-I (TnI) and myeloperoxidase-positive cell infiltration at day 3, and evaluated LV function and remodeling after 4 weeks using echocardiography and pressure-conductance catheterization. Administration of iNO, TAD, or both during I/R was safe and hemodynamically well tolerated. Compared with untreated mice (CON), only iNO+TAD increased plasma and cardiac-cGMP levels during early reperfusion (80 ± 12 versus 36 ± 6 pmol/ml and 0.15 ± 0.02 versus 0.05 ± 0.01 pmol/mg protein, P < 0.05 for both). Moreover, iNO+TAD reduced TnI at 4 hours to a greater extent (P < 0.001 versus CON) than either alone (P < 0.05 versus CON) and was associated with significantly less myocardial inflammatory cell infiltration at day 3. After 4 weeks and compared with CON, iNO+TAD was associated with increased fractional shortening (43 ± 1 versus 33 ± 2%, P < 0.01), larger stroke volumes (14.9 ± 1.2 versus 10.2 ± 0.9 µl, P < 0.05), enhanced septal and posterior wall thickening (P < 0.05 and P < 0.001, respectively), and attenuated LV dilatation (P < 0.001), whereas iNO or TAD alone conferred less benefit. Thus, iNO+TAD has superior efficacy to limit early reperfusion injury and attenuate adverse LV remodeling. Combination of inhaled NO with a long-acting PDE5 inhibitor may represent a promising strategy to reduce ischemic damage following reperfusion and better preserve LV function.


Asunto(s)
Cardiotónicos/administración & dosificación , Daño por Reperfusión Miocárdica/diagnóstico por imagen , Daño por Reperfusión Miocárdica/prevención & control , Óxido Nítrico/administración & dosificación , Inhibidores de Fosfodiesterasa 5/administración & dosificación , Administración por Inhalación , Animales , Quimioterapia Combinada , Masculino , Ratones , Ratones Endogámicos C57BL , Ultrasonografía
13.
J Clin Invest ; 125(12): 4463-82, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26571398

RESUMEN

Conditions such as muscular dystrophies (MDs) that affect both cardiac and skeletal muscles would benefit from therapeutic strategies that enable regeneration of both of these striated muscle types. Protocols have been developed to promote induced pluripotent stem cells (iPSCs) to differentiate toward cardiac or skeletal muscle; however, there are currently no strategies to simultaneously target both muscle types. Tissues exhibit specific epigenetic alterations; therefore, source-related lineage biases have the potential to improve iPSC-driven multilineage differentiation. Here, we determined that differential myogenic propensity influences the commitment of isogenic iPSCs and a specifically isolated pool of mesodermal iPSC-derived progenitors (MiPs) toward the striated muscle lineages. Differential myogenic propensity did not influence pluripotency, but did selectively enhance chimerism of MiP-derived tissue in both fetal and adult skeletal muscle. When injected into dystrophic mice, MiPs engrafted and repaired both skeletal and cardiac muscle, reducing functional defects. Similarly, engraftment into dystrophic mice of canine MiPs from dystrophic dogs that had undergone TALEN-mediated correction of the MD-associated mutation also resulted in functional striatal muscle regeneration. Moreover, human MiPs exhibited the same capacity for the dual differentiation observed in murine and canine MiPs. The findings of this study suggest that MiPs should be further explored for combined therapy of cardiac and skeletal muscles.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Mesodermo/metabolismo , Músculo Esquelético/fisiología , Miocardio , Regeneración , Animales , Perros , Humanos , Células Madre Pluripotentes Inducidas/citología , Mesodermo/citología , Ratones , Distrofias Musculares/metabolismo , Distrofias Musculares/terapia , Ratas
14.
J Appl Physiol (1985) ; 117(5): 563-71, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25059236

RESUMEN

Transverse aortic constriction (TAC) and angiotensin II (ANG II) subcutaneous osmotic pump infusion are frequently used murine models of pressure overload hypertrophy. The aim of this paper is to investigate time- and stressor-dependent functional and structural changes using echocardiographic B-mode, M-mode, and Doppler characterization. Ten-week-old male C57BL6/J wild-type mice received 4-wk ANG II (1.5 mg·kg(-1)·day(-1), n = 19) or saline (n = 10) infusion followed by echocardiography (Vevo2100, Visual Sonics), or underwent TAC (n = 63) or a sham operation (n = 30). In the TAC protocol, echocardiography was performed after 2 wk (n = 22 TAC, n = 10 sham), after 4 wk (n = 20 TAC, n = 10 sham), and after 10 wk (n = 21 TAC, n = 10 sham). ANG II infusion was associated with a mixed pressure and volume overload, with a variable contribution of volume overload caused by aortic valve insufficiency (grade 0.5-3.5/4). The degree of aortic valve insufficiency correlated with the degree of left ventricular dilation (r(2) = 0.671, P < 0.001). After TAC, all hypertrophic remodeling patterns known in human disease were observed: 1) low-flow, low-gradient with preserved ejection fraction (EF); 2) concentric hypertrophy with normal EF and flow; 3) concentric hypertrophy with moderately decreased EF and/or flow; 4) eccentric hypertrophy with normal EF and flow; 5) eccentric hypertrophy with moderately decreased EF and/or flow; and 6) eccentric hypertrophy with severely depressed EF. Eccentric remodeling was time dependent, with 5% of mice developing this phenotype at 2 wk, 39% at 4 wk, and 59% at 10 wk. Comprehensive echocardiographic analysis allows identification of homogeneous subgroups of mice subjected to hypertrophic stress, reducing variability in experimental results and facilitating clinical translation.


Asunto(s)
Cardiomegalia/diagnóstico por imagen , Cardiomegalia/fisiopatología , Angiotensina II , Animales , Aorta Torácica , Cardiomegalia/inducido químicamente , Constricción Patológica , Interpretación Estadística de Datos , Modelos Animales de Enfermedad , Ecocardiografía/métodos , Ecocardiografía Doppler/métodos , Ratones , Ratones Endogámicos C57BL , Presión , Vasoconstrictores
15.
Ultrasound Med Biol ; 39(10): 1848-60, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23830981

RESUMEN

Ultrasound assessment of myocardial strain can provide valuable information on regional cardiac function. However, Doppler-based methods often used in practice for strain estimation suffer from angle dependency. In this study, a partial solution to that fundamental limitation is presented. We have previously reported using simulated data sets that spatial compounding of axial velocities obtained at three steering angles can theoretically outperform 2-D speckle tracking for 2-D strain estimation in the mouse heart. In this study, the feasibility of the method was analyzed in vivo using spatial compounding of Doppler velocities on six mice with myocardial infarction and five controls, and results were compared with those of tagged microscopic magnetic resonance imaging (µMRI). Circumferential estimates quantified by means of both ultrasound and µMRI could detect regional dysfunction. Between echocardiography and µMRI, a good regression coefficient was obtained for circumferential strain estimates (r = 0.69), whereas radial strain estimates correlated only moderately (r = 0.37). A second echocardiography was performed after µMRI to test the reproducibility of the compounding method. This yielded a higher correlation coefficient for the circumferential component than for the radial component (r = 0.74 circumferentially, r = 0.49 radially).


Asunto(s)
Algoritmos , Ecocardiografía Doppler/métodos , Diagnóstico por Imagen de Elasticidad/métodos , Ventrículos Cardíacos/diagnóstico por imagen , Interpretación de Imagen Asistida por Computador/métodos , Contracción Miocárdica/fisiología , Función Ventricular Izquierda/fisiología , Animales , Módulo de Elasticidad/fisiología , Estudios de Factibilidad , Aumento de la Imagen/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Análisis Espacio-Temporal , Estrés Mecánico , Resistencia a la Tracción/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...