Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Annu Rev Pharmacol Toxicol ; 58: 83-103, 2018 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-28992430

RESUMEN

Billions of US dollars are invested every year by the pharmaceutical industry in drug development, with the aim of introducing new drugs that are effective and have minimal side effects. Thirty percent of in-pipeline drugs are excluded in an early phase of preclinical and clinical screening owing to cardiovascular safety concerns, and several lead molecules that pass the early safety screening make it to market but are later withdrawn owing to severe cardiac side effects. Although the current drug safety screening methodologies can identify some cardiotoxic drug candidates, they cannot accurately represent the human heart in many aspects, including genomics, transcriptomics, and patient- or population-specific cardiotoxicity. Despite some limitations, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are a powerful and evolving technology that has been shown to recapitulate many attributes of human cardiomyocytes and their drug responses. In this review, we discuss the potential impact of the inclusion of the hiPSC-CM platform in premarket candidate drug screening.


Asunto(s)
Cardiotoxicidad/etiología , Evaluación Preclínica de Medicamentos/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Corazón/efectos de los fármacos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Animales , Humanos , Miocitos Cardíacos/efectos de los fármacos
2.
Circulation ; 136(7): 664-679, 2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28588076

RESUMEN

BACKGROUND: Fibrosis is the pathological consequence of stress-induced tissue remodeling and matrix accumulation. Increased levels of plasminogen activator inhibitor type I (PAI-1) have been shown to promote fibrosis in multiple organ systems. Paradoxically, homozygous genetic deficiency of PAI-1 is associated with spontaneous age-dependent, cardiac-selective fibrosis in mice. We have identified a novel PAI-1-dependent mechanism that regulates cardiomyocyte-derived fibrogenic signals and cardiac transcriptional pathways during injury. METHODS: Cardiac fibrosis in subjects with homozygous mutation in SERPINE-1 was evaluated with late gadolinium-enhanced cardiac magnetic resonance imaging. A murine cardiac injury model was performed by subcutaneous infusion of either saline or Angiotensin II by osmotic minipumps. We evaluated blood pressure, cardiac function (by echocardiography), fibrosis (with Masson Trichrome staining), and apoptosis (with TUNEL staining), and we performed transcriptome analysis (with RNA sequencing). We further evaluated fibrotic signaling in isolated murine primary ventricular myocytes. RESULTS: Cardiac fibrosis was detected in 2 otherwise healthy humans with complete PAI-1 deficiency because of a homozygous frameshift mutation in SERPINE-1. In addition to its suppressive role during spontaneous cardiac fibrosis in multiple species, we hypothesized that PAI-1 also regulates fibrosis during cardiac injury. Treatment of young PAI-1-/- mice with Angiotensin II induced extensive hypertrophy and fibrotic cardiomyopathy, with increased cardiac apoptosis and both reactive and replacement fibrosis. Although Angiotensin II-induced hypertension was blunted in PAI-1-/- mice, cardiac hypertrophy was accelerated. Furthermore, ventricular myocytes were found to be an important source of cardiac transforming growth factor-ß (TGF-ß) and PAI-1 regulated TGF-ß synthesis by cardiomyocytes in vitro as well as in vivo during cardiac injury. Transcriptome analysis of ventricular RNA after Angiotensin II treatment confirmed that PAI-1 deficiency significantly enhanced multiple TGF-ß signaling elements and transcriptional targets, including genes for extracellular matrix components, mediators of extracellular matrix remodeling, matricellular proteins, and cardiac integrins compared with wild-type mice. CONCLUSIONS: PAI-1 is an essential repressor of cardiac fibrosis in mammals. We define a novel cardiomyocyte-specific regulatory mechanism for TGF-ß production by PAI-1, which explains the paradoxical effect of PAI-1 deficiency in promoting cardiac-selective fibrosis. Thus, PAI-1 is a molecular switch that controls the cardiac TGF-ß axis and its early transcriptional effects that lead to myocardial fibrosis.


Asunto(s)
Cardiomegalia/patología , Miocitos Cardíacos/metabolismo , Inhibidor 1 de Activador Plasminogénico/genética , Factor de Crecimiento Transformador beta/metabolismo , Angiotensina II/farmacología , Angiotensina II/uso terapéutico , Animales , Proteína Morfogenética Ósea 7/farmacología , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/metabolismo , Células Cultivadas , Femenino , Mutación del Sistema de Lectura , Humanos , Imagen por Resonancia Cinemagnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Inhibidor 1 de Activador Plasminogénico/deficiencia , Inhibidor 1 de Activador Plasminogénico/metabolismo , ARN/química , ARN/metabolismo , Análisis de Secuencia de ARN , Proteína smad6/antagonistas & inhibidores , Proteína smad6/genética , Proteína smad6/metabolismo , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología
3.
Commun Biol ; 1: 155, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30302399

RESUMEN

Kv7.1 (KCNQ1) coassembles with KCNE1 to generate the cardiac IKs -channel. Gain- and loss-of-function mutations in KCNQ1 are associated with cardiac arrhthymias, highlighting the importance of modulating IKs activity for cardiac function. Here, we report proteolysis of Kv7.1 as an irreversible posttranslational modification. The identification of two C-terminal fragments of Kv7.1 led us to identify an aspartate critical for the generation of one of the fragments and caspases as responsible for mediating proteolysis. Activating caspases reduces Kv7.1/KCNE1 currents, which is abrogated in cells expressing caspase-resistant channels. Enhanced cleavage of Kv7.1 can be detected for the LQT mutation G460S, which is located adjacent to the cleavage site, whereas a calmodulin-binding-deficient mutation impairs cleavage. Application of apoptotic stimuli or doxorubicin-induced cardiotoxicity provokes caspase-mediated cleavage of endogenous IKs in human cardiomyocytes. In summary, caspases are novel regulatory components of IKs channels that may have important implications for the molecular mechanism of doxorubicin-induced cardiotoxicity.

4.
J Electrocardiol ; 40(6 Suppl): S199-201, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17993323

RESUMEN

Heart failure survival after diagnosis has barely changed for more than half a century. Recently, investigation has focused on differentiation of stem cells in vitro and their delivery for use in vivo as replacement cardiac contractile elements. Here we report preliminary results using mesenchymal stem cells partially differentiated to a cardiac lineage in vitro. When delivered to the canine heart on an extracellular matrix patch to replace a full-thickness ventricular defect in vivo, they improve regional mechanical function. The delivered cells were also tracked, and some became myocytes with mature sarcomeres.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Infarto del Miocardio/patología , Infarto del Miocardio/cirugía , Animales , Perros , Proyectos Piloto , Resultado del Tratamiento
5.
Circulation ; 110(6): 713-7, 2004 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-15289373

RESUMEN

BACKGROUND: Neuregulins are required for maintenance of acetylcholine receptor-inducing activity of nicotinic receptors in neurons and skeletal muscle, but effects of neuregulins on muscarinic receptors are not known. In the normal heart, parasympathetic activation counterbalances beta-adrenergic activation. To test the hypothesis that neuregulins modify parasympathetic function in the heart, we studied cardiomyocytes from mice heterozygous for neuregulin-1 gene deletion (NRG-1+/-) and examined the effects of beta-adrenergic stimulation on contractility in the presence and absence of the muscarinic agonist carbachol. METHODS AND RESULTS: We evaluated contraction and intracellular Ca2+ transients ([Ca2+]i) in left ventricular (LV) myocytes loaded with Fluo-3 from NRG-1+/- and wild-type (WT) mice. Under baseline conditions (0.5 Hz, 1.5 mmol/L [Ca2+]o, 25 degrees C), characteristics of myocyte contraction/relengthening and systolic/diastolic [Ca2+]i were not different between WT and NRG-1+/- mice. The steady-state increases in fractional shortening (FS) and peak-systolic [Ca2+]i in response to isoproterenol were similar in both groups. In WT myocytes stimulated with isoproterenol, carbachol decreased FS, peak-systolic [Ca2+]i, and cAMP levels. In NRG-1+/- myocytes, carbachol did not attenuate either FS or peak-systolic [Ca2+]i, associated with the failure to decrease cAMP levels. Investigation of muscarinic receptor signaling showed no difference of LV protein levels of muscarinic M2 receptors or G protein Galpha(i1,2), Galpha(i3), and Galpha(o) subunits. CONCLUSIONS: Cardiomyocytes deficient in neuregulin signaling are unable to adequately counterbalance beta-adrenergic activation by inhibitory parasympathetic activity. This mechanism may contribute to the known increased risk of heart failure in injured human hearts when neuregulin signaling is suppressed.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Neurregulina-1/fisiología , Receptor Muscarínico M2/fisiología , Animales , Señalización del Calcio/efectos de los fármacos , Carbacol/farmacología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/fisiología , AMP Cíclico/fisiología , Eliminación de Gen , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/fisiología , Heterocigoto , Isoproterenol/farmacología , Ratones , Ratones Transgénicos , Agonistas Muscarínicos/farmacología , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Neurregulina-1/deficiencia , Neurregulina-1/genética , Sistema Nervioso Parasimpático/efectos de los fármacos , Sistema Nervioso Parasimpático/fisiología , Receptores Nicotínicos/efectos de los fármacos , Receptores Nicotínicos/fisiología
6.
Exp Hematol ; 31(11): 1112-8, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14585377

RESUMEN

To block development of progressive childhood diseases, in utero transplantation (IUTx) requires immediate and significant donor peripheral blood (PB) cell amplification. To date, negligible and nontherapeutic donor PB cell levels have been observed postnatally, except in patients with immunodeficiency diseases. Donor cell fate in utero still is not clear. Ease of identifying and quantifying beta-glucuronidase (GUSB)-expressing donor cells in GUSB-null mucopolysaccharidosis type VII (MPSVII) mouse recipients allowed us to evaluate temporal donor cell engraftment and amplification post-IUTx. Like humans, MPSVII mice are unable to catabolize lysosomal glycosaminoglycans and progressively develop severe storage disease unless they are treated early in life.IUTx recipients were nonablated MPSVII fetuses and genetically stem cell-deficient, and hence myeloablated, W(41)/W(41) MPSVII fetuses. Donor GUSB+ cells were identified and counted in histochemical tissue sections. Quantitative results were confirmed by flow cytometry, enzyme analysis, and histopathology. Whereas GUSB+ cells engraft in most tissues in utero, significant amplification does not occur until the first postnatal week in the nonablated MPSVII hosts. In contrast, genetically myeloablated MPSVII recipients display widely distributed donor cell replacement accompanied by extensive amplification in utero. In both models, storage is alleviated in adult tissues with significant donor cell repopulation. To become therapeutic, IUTx must overcome the limitations of donor cell expansion in the highly competitive fetal environment. Fortunately, nonablative mechanisms to amplify cells in utero are coming on line.


Asunto(s)
Enfermedades Fetales/terapia , Glucuronidasa/análisis , Mucopolisacaridosis VII/terapia , Trasplante de Células Madre/métodos , Animales , Recuento de Células , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factores de Tiempo
7.
Tissue Eng Part A ; 15(8): 2189-201, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19231971

RESUMEN

During the past few years, studies involving the implantation of stem cells, chemical factors, and scaffolds have demonstrated the ability to augment the mammalian heart's native regenerative capacity. Scaffolds comprised of extracellular matrix (ECM) have been used to repair myocardial defects. These scaffolds become populated with myocytes and provide regional contractile function, but quantification of the myocyte population has not yet been conducted. The purpose of this study was to quantitate the myocyte content within the ECM bioscaffold and to correlate this cell population with the regional mechanical function over time. Xenogenic ECM scaffolds derived from porcine urinary bladder were implanted into a full-thickness, surgically induced, right ventricular-free wall defect in a dog model. Zero, 2, and 8 weeks following implantation, regional function and myocyte content were determined in each patch region. Regional function did not significantly increase from 0 to 2 weeks. At 8 weeks, however, regional stroke work increased to 3.7 +/- 0.7% and systolic contraction increased to 4.4 +/- 1.2%. The myocyte content also significantly increased during that period generating a linear relationship between regional function and myocyte content. In conclusion, ECM used as a myocardial patch increases both the regional function and the myocyte content over time. The mechanical function generated in the patch region is correlated with the quantity of local tissue myocytes.


Asunto(s)
Fenómenos Mecánicos , Células Musculares/citología , Miocardio/metabolismo , Implantación de Prótesis , Ingeniería de Tejidos , Animales , Ciclo Celular , Proliferación Celular , Perros , Matriz Extracelular/trasplante , Células Musculares/metabolismo , Miocardio/patología , Regeneración , Coloración y Etiquetado , Sus scrofa , Factores de Tiempo , Andamios del Tejido , Vejiga Urinaria/trasplante , Presión Ventricular
8.
Curr Treat Options Cardiovasc Med ; 10(1): 59-72, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18325308

RESUMEN

Cellular cardiomyoplasty has raised hopes of regenerating mechanical function in the heart. Several cell sources have been investigated for their ability to repair the damaged heart, providing reason for optimism. Multiple mechanisms have been proposed for the beneficial effects of the delivered cells; however, true reversal of cardiac damage implies the generation of new contractile myocytes. The assessment of a cell's ability to regenerate contractile cells requires a defined set of criteria that, if met, define success. Here we review data from the four primary players in cellular cardiomyoplasty (skeletal myoblasts, bone marrow cells, embryonic stem cells, and resident cardiac stem cells) and assess their potential to differentiate into contractile myocytes as indicated by their ability to meet such specified milestones. Both animal studies and clinical trials suggest that current experimental approaches to cellular cardiomyoplasty yield short-term improvement, although it may be independent of cell type used. However, the mechanisms underlying this salutary effect, as well as its persistence in the longer term, have remained elusive.

9.
Am J Physiol Heart Circ Physiol ; 294(3): H1274-81, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18178728

RESUMEN

The role of the angiotensin II type 2 (AT2) receptor in cardiac hypertrophy remains controversial. We studied the effects of AT2 receptors on chronic pressure overload-induced cardiac hypertrophy in transgenic mice selectively overexpressing AT2 receptors in ventricular myocytes. Left ventricular (LV) hypertrophy was induced by ascending aorta banding (AS). Transgenic mice overexpressing AT2 (AT2TG-AS) and nontransgenic mice (NTG-AS) were studied after 70 days of aortic banding. Nonbanded NTG mice were used as controls. LV function was determined by catheterization via LV puncture and cardiac magnetic resonance imaging. LV myocyte diameter and interstitial collagen were determined by confocal microscopy. Atrial natriuretic polypeptide (ANP) and brain natriuretic peptide (BNP) were analyzed by Northern blot. Sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA)2, inducible nitric oxide synthase (iNOS), endothelial NOS, ERK1/2, p70S6K, Src-homology 2 domain-containing protein tyrosine phosphatase-1, and protein serine/threonine phosphatase 2A were analyzed by Western blot. LV myocyte diameter and collagen were significantly reduced in AT2TG-AS compared with NTG-AS mice. LV anterior and posterior wall thickness were not different between AT2TG-AS and NTG-AS mice. LV systolic and diastolic dimensions were significantly higher in AT2TG-AS than in NTG-AS mice. LV systolic pressure and end-diastolic pressure were lower in AT2TG-AS than in NTG-AS mice. ANP, BNP, and SERCA2 were not different between AT2TG-AS and NTG-AS mice. Phospholamban (PLB) and the PLB-to-SERCA2 ratio were significantly higher in AT2TG-AS than in NTG-AS mice. iNOS was higher in AT2TG-AS than in NTG-AS mice but not significantly different. Our results indicate that AT2 receptor overexpression modified the pathological hypertrophic response to aortic banding in transgenic mice.


Asunto(s)
Presión Sanguínea/fisiología , Cardiomegalia/genética , Cardiomegalia/patología , Hipertensión/patología , Miocitos Cardíacos/patología , Receptor de Angiotensina Tipo 2/genética , Animales , Factor Natriurético Atrial/metabolismo , Northern Blotting , Western Blotting , Colágeno/metabolismo , Hipertensión/genética , Masculino , Ratones , Ratones Transgénicos , Microscopía Confocal , Miocardio/metabolismo , Péptido Natriurético Encefálico/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor de Angiotensina Tipo 2/fisiología , Análisis de Supervivencia , Función Ventricular Izquierda/fisiología
10.
Am J Physiol Heart Circ Physiol ; 295(6): H2257-63, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18835924

RESUMEN

The need to regenerate tissue is paramount, especially for the heart that lacks the ability to regenerate after injury. The urinary bladder extracellular matrix (ECM), when used to repair a right ventricular defect, successfully regenerated some mechanical function. The objective of the current study was to determine whether the regenerative effect of ECM could be improved by seeding the patch with human mesenchymal stem cells (hMSCs) enhanced to differentiate down a cardiac linage. hMSCs were used to form three-dimensional spheroids. The expression of cardiac proteins was determined in cells exposed to the spheroid formation and compared with nonmanipulated hMSCs. To determine whether functional calcium channels were present, the cells were patch clamped. To evaluate the ability of these cells to regenerate mechanical function, the spheroids were seeded on ECM and then implanted into the canine heart to repair a full-thickness right ventricular defect. As a result, many of the cells spreading from the spheroids expressed cardiac-specific proteins, including sarcomeric alpha-actinin, cardiotin, and atrial natriuretic peptide, as well as the cell cycle markers cyclin D1 and proliferating cell nuclear antigen. A calcium current similar in amplitude to that of ventricular myocytes was present in 16% of the cells. The cardiogenic cell-seeded scaffolds increased the regional mechanical function in the canine heart compared with the unmanipulated hMSC-seeded scaffolds. In addition, the cells prelabeled with fluorescent markers demonstrated myocyte-specific actinin staining with sarcomere spacing similar to that of normal myocytes. In conclusion, the spheroid-derived cells express cardiac-specific proteins and demonstrate a calcium current similar to adult ventricular myocytes. When these cells are implanted into the canine heart, some of these cells appear striated and mechanical function is improved compared with the unmanipulated hMSCs. Further investigation will be required to determine whether the increased mechanical function is due to a differentiation of the cardiogenic cells to myocytes or to other effects.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Matriz Extracelular/metabolismo , Cardiopatías/cirugía , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Miocitos Cardíacos/metabolismo , Andamios del Tejido , Animales , Canales de Calcio Tipo L/metabolismo , Modelos Animales de Enfermedad , Perros , Cardiopatías/metabolismo , Cardiopatías/fisiopatología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/cirugía , Humanos , Potenciales de la Membrana , Proteínas Musculares/metabolismo , Contracción Miocárdica , Regeneración , Sarcómeros/metabolismo , Esferoides Celulares , Porcinos , Vejiga Urinaria/metabolismo , Función Ventricular Derecha
11.
Stem Cells ; 25(8): 2128-38, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17495112

RESUMEN

Stem cells show promise for repair of damaged cardiac tissue. Little is known with certainty, however, about the distribution of these cells once introduced in vivo. Previous attempts at tracking delivered stem cells have been hampered by the autofluorescence of host tissue and limitations of existing labeling techniques. We have developed a novel loading approach to stably label human mesenchymal stem cells with quantum dot (QD) nanoparticles. We report the optimization and validation of this long-term tracking technique and highlight several important biological applications by delivering labeled cells to the mammalian heart. The bright QD crystals illuminate exogenous stem cells in histologic sections for at least 8 weeks following delivery and permit, for the first time, the complete three-dimensional reconstruction of the locations of all stem cells following injection into the heart. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Imagenología Tridimensional , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Microscopía Fluorescente , Miocardio/citología , Puntos Cuánticos , Coloración y Etiquetado/métodos , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Perros , Endocitosis/fisiología , Colorantes Fluorescentes/farmacología , Corazón/fisiología , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratas , Regeneración , Transfección
12.
Am J Physiol Heart Circ Physiol ; 289(2): H660-6, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15833803

RESUMEN

Neuregulins and their erbB receptors are essential for cardiac development and postulated to be cardioprotective in the presence of injury in the postnatal heart. We tested the hypothesis that the development of doxorubicin-induced cardiotoxicity in vivo is more severe in mice with heterozygous knockout of the neuregulin-1 gene (NRG-1(+/-)) compared with wild-type mice (WT). Three-month old NRG-1(+/-) and WT mice were injected with a single dose of doxorubicin (20 mg/kg ip). Survival was analyzed by the Kaplan-Meier approach. Left ventricular (LV) function and signaling pathways were analyzed 4 days after treatment. Fifteen days after treatment, survival was significantly lower in doxorubicin-treated NRG-1(+/-) mice (NRG-1(+/-)-Dox) compared with doxorubicin-treated WT mice (WT-Dox) (15% vs. 33%, P < 0.01). LV mass was significantly lower in NRG-1(+/-)-Dox but not in WT-Dox compared with nontreated animals. LV systolic pressure and LV midwall fractional shortening were significantly lower in NRG-1(+/-)-Dox compared with WT-Dox mice. LV protein levels of NRG-1, erbB2, and erbB4 receptors were similar in WT-Dox and NRG-1(+/-)-Dox mice. However, levels of phosphorylated erbB2, Akt, and ERK-1/2 were significantly decreased in NRG-1(+/-)-Dox compared with WT-Dox mice. A significant decrease in phosphorylated P70S6K levels was also observed in NRG-1(+/-)-Dox compared with nontreated NRG-1(+/-) mice. These results demonstrate that heterozygous knockout of the neuregulin-1 gene worsens survival and LV function in the presence of doxorubicin-induced cardiac injury in vivo. This is associated with the depression of activation of the erbB2 receptor as well as Akt, p70S6K, and ERK-1/2 pathways.


Asunto(s)
Gasto Cardíaco Bajo/inducido químicamente , Gasto Cardíaco Bajo/fisiopatología , Doxorrubicina , Neurregulina-1/deficiencia , Animales , Gasto Cardíaco Bajo/diagnóstico por imagen , Gasto Cardíaco Bajo/patología , Ecocardiografía , Receptores ErbB/metabolismo , Hemodinámica , Heterocigoto , Etiquetado Corte-Fin in Situ , Ratones , Ratones Noqueados , Miocardio/patología , Neurregulina-1/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-4 , Índice de Severidad de la Enfermedad , Transducción de Señal , Análisis de Supervivencia
13.
J Immunol ; 171(6): 3270-7, 2003 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12960357

RESUMEN

A significant number of nonmalignant, progressive childhood disorders respond to bone marrow transplantation (BMT). Toxic myeloablative pretreatment regimens, graft failure, and graft-vs-host disease complicate the utility of BMT for neonatal treatment. We recently demonstrated high-dose BMT in neonatal animals enables chimeric engraftment without toxic myeloablation. Reagents that block T cell costimulation (anti-CD40L mAb and/or CTLA-4Ig) establish tolerant allogeneic engraftment in adult recipients. Donor lymphocyte infusion (DLI) re-establishes failing grafts and treats malignant relapse via a graft-vs-leukemia response. In this study, we tested the hypothesis that combining these approaches would allow tolerant allogeneic engraftment devoid of myeloablation in neonatal normal and mutant mice with lysosomal storage disease. Tolerant chimeric allogeneic engraftment was achieved before DLI only in the presence of both anti-CD40L mAb and CTLA-4Ig. DLI amplified allografts to full donor engraftment long-term. DLI-treated mice either maintained long-term tolerance or developed late-onset chronic graft-vs-host disease. This combinatorial approach provides a nontoxic method to establish tolerant allogeneic engraftment for treatment of progressive childhood diseases.


Asunto(s)
Animales Recién Nacidos/inmunología , Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Trasplante de Médula Ósea/inmunología , Ligando de CD40/inmunología , Inmunoconjugados/administración & dosificación , Activación de Linfocitos/inmunología , Acondicionamiento Pretrasplante/métodos , Abatacept , Animales , Animales Recién Nacidos/genética , Trasplante de Médula Ósea/métodos , Trasplante de Médula Ósea/patología , Ligando de CD40/fisiología , Células Cultivadas , Quimera/inmunología , Enfermedad Crónica , Quimioterapia Combinada , Femenino , Refuerzo Inmunológico de Injertos/métodos , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Antígenos H-2/genética , Humanos , Tolerancia Inmunológica/genética , Activación de Linfocitos/genética , Transfusión de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Mutantes , Trasplante Homólogo
14.
J Card Fail ; 10(6): 511-8, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15599842

RESUMEN

BACKGROUND: Clinical and experimental studies suggest that aldosterone modulates myocardial hypertrophy. From in vivo studies, it is not possible to distinguish between direct actions on myocyte growth and effects of mechanical load. In this study we tested the hypothesis that aldosterone induces myocyte hypertrophy in low-density, serum-free cultures of neonatal rat ventricular myocytes. METHODS AND RESULTS: Hypertrophy was quantified by [(14)C]-phenylalanine incorporation and confocal microscopic assessment of myocyte surface area. Aldosterone caused a 27% increase in protein incorporation (EC(50) = 40 nmol/L) and a 29% increase in myocyte surface area compared with the vehicle control. This response was associated with increased mRNA levels of atrial natriuretic factor, alpha- and beta-myosin heavy chain measured by RNase protection assay, and it was suppressed by the mineralocorticoid receptor blocker spironolactone. Analysis of early signaling events showed that aldosterone stimulation acutely translocated protein kinase C (PKC)-alpha to the membrane fraction and increased the levels of phosphorylated ERK1/2 and JNK. PD 98059, an inhibitor of the ERK activator MEK (mitogen-activated protein kinase kinase) and bisindolylmaleimide I, an inhibitor of PKC activation, each blocked aldosterone-stimulated hypertrophy. CONCLUSION: Aldosterone directly stimulates hypertrophy in neonatal rat ventricular myocytes. The growth response is dependent on the mineralocorticoid receptor and is associated with activation of ERK, JNK, and PKC-alpha.


Asunto(s)
Aldosterona/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Aldosterona/administración & dosificación , Animales , Animales Recién Nacidos , Células Cultivadas , Medio de Cultivo Libre de Suero , Dexametasona/farmacología , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Glucocorticoides/farmacología , Antagonistas de Hormonas/farmacología , Hipertrofia , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Microscopía Confocal , Mifepristona/farmacología , Antagonistas de Receptores de Mineralocorticoides/farmacología , Proteínas Quinasas Activadas por Mitógenos/fisiología , Proteína Quinasa C/fisiología , Ratas , Ratas Wistar , Receptores de Mineralocorticoides/fisiología , Espironolactona/farmacología
15.
Blood Cells Mol Dis ; 32(1): 199-213, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14757436

RESUMEN

Lysosomal storage diseases (LSD) respond to bone marrow (BM) transplantation when donor-derived cells deliver needed enzyme. Hypothetically, the ubiquitous resident macrophages (MPhi) are the primary delivery vehicle of therapeutic protein. In mucopolysaccharidosis type VII (MPS VII) mice with LSD, transplanted mature MPhi reduce undegraded glycosaminoglycans (GAG) in the lysosome but are incapable of self-renewal, leading to return of storage after 1 month. We show here that a population of early BM-derived myeloid progenitors devoid of long-term hematopoietic stem cells (LT-HSC) engrafted MPS VII BM, released monocytes into peripheral blood (PBL), and engrafted tissues at known sites of resident MPhi. These primitive Mac-1- cells were sorted from normal whole BM and were defined by ER-MP12hi20-58med/hi labeling. Lysosomal storage was reduced in liver, spleen, thymus, heart, kidney, and bone. Cells persisted for 3 months, suggesting self-renewal capacity or a long half-life. Cells sorted from BM by ER-MP12-20hi marker expression (which are maturer myeloid cells that express Mac-1) engrafted tissues instead of BM and quantitatively repopulated less than cells derived from the ER-MP12hi20-58med/hi population. Also, reduction of lysosomal storage was variable and generally less when compared to that following transplantation of immature ER-MP12hi20-58med/hi cells. We conclude that primitive myeloid progenitors are more therapeutic for LSD than mature myeloid cells due to their greater longevity and increased capacity to seed tissues. The ability of cells derived from these primitive precursors to seed deep within tissues make them excellent candidates for both cellular therapy and gene transfer techniques to cure a wide range of metabolic diseases.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Enfermedades por Almacenamiento Lisosomal/terapia , Macrófagos/citología , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/trasplante , Animales , Células de la Médula Ósea , Movimiento Celular , Separación Celular , Supervivencia de Injerto , Antígeno de Macrófago-1/análisis , Ratones , Monocitos/citología , Mucopolisacaridosis VII/terapia , Especificidad de Órganos
16.
Am J Physiol Heart Circ Physiol ; 285(5): H2179-87, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12869376

RESUMEN

The angiotensin II type 2 (AT2) receptor is upregulated in the left ventricle in heart failure, but its pathophysiological roles in vivo are not understood. In the present study, AT2 receptors were expressed in transgenic (TG) mice using the ventricular-specific myosin light-chain (MLC-2v) promoter. In TG compared with nontransgenic (NTG) mice, in vivo left ventricular (LV) systolic pressure and peak +dP/dt were depressed while LV diastolic pressure was elevated (P < 0.05). Echocardiography showed severely depressed LV fractional shortening, increased systolic and diastolic dimensions, and wall thinning (P < 0.05). Confocal and electron microscopy studies revealed an increase in the size of myocytes and interstitial spaces as well as an increase in interstitial collagen, disruption of the Z-band, and changes in cytochrome c localization. The changes were most prominent in the highest-expressing TG line, which implies a dose-response relationship. AT2 overexpression was also directly associated with the increase of phosphorylated protein levels of PKC-alpha, PKC-beta, and p70S6 kinase. These data demonstrate that ventricular myocyte-specific expression of AT2 receptors promotes the development of dilated cardiomyopathy and heart failure in vivo.


Asunto(s)
Cardiomiopatía Dilatada/fisiopatología , Insuficiencia Cardíaca/fisiopatología , Receptor de Angiotensina Tipo 2/genética , Animales , Cardiomiopatía Dilatada/etiología , Cardiomiopatía Dilatada/patología , Expresión Génica/fisiología , Corazón/fisiología , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Microscopía Electrónica , Miocitos Cardíacos/patología , Miocitos Cardíacos/ultraestructura , Cadenas Ligeras de Miosina/genética , Regiones Promotoras Genéticas , Función Ventricular , Función Ventricular Izquierda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA