Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Am J Respir Crit Care Med ; 203(5): 614-627, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33021405

RESUMEN

Rationale: Pulmonary arterial hypertension (PAH) is a life-threatening condition characterized by abnormally elevated pulmonary pressures and right ventricular failure. Excessive proliferation and resistance to apoptosis of pulmonary artery smooth muscle cells (PASMCs) is one of the most important drivers of vascular remodeling in PAH, for which available treatments have limited effectiveness.Objectives: To gain insights into the mechanisms leading to the development of the disease and identify new actionable targets.Methods: Protein expression profiling was conducted by two-dimensional liquid chromatography coupled to tandem mass spectrometry in isolated PASMCs from controls and patients with PAH. Multiple molecular, biochemical, and pharmacologic approaches were used to decipher the role of NUDT1 (nudrix hyrolase 1) in PAH.Measurements and Main Results: Increased expression of the detoxifying DNA enzyme NUDT1 was detected in cells and tissues from patients with PAH and animal models. In vitro, molecular or pharmacological inhibition of NUDT1 in PAH-PASMCs induced accumulation of oxidized nucleotides in the DNA, irresolvable DNA damage (comet assay), disruption of cellular bioenergetics (Seahorse), and cell death (terminal deoxynucleotidyl transferase dUTP nick end labeling assay). In two animal models with established PAH (i.e., monocrotaline and Sugen/hypoxia-treated rats), pharmacological inhibition of NUDT1 using (S)-Crizotinib significantly decreased pulmonary vascular remodeling and improved hemodynamics and cardiac function.Conclusions: Our results indicate that, by overexpressing NUDT1, PAH-PASMCs hijack persistent oxidative stress in preventing incorporation of oxidized nucleotides into DNA, thus allowing the cell to escape apoptosis and proliferate. Given that NUDT1 inhibitors are under clinical investigation for cancer, they may represent a new therapeutic option for PAH.


Asunto(s)
Enzimas Reparadoras del ADN/genética , ADN/metabolismo , Estrés Oxidativo/genética , Monoéster Fosfórico Hidrolasas/genética , Hipertensión Arterial Pulmonar/genética , Arteria Pulmonar/metabolismo , Remodelación Vascular/genética , 8-Hidroxi-2'-Desoxicoguanosina/metabolismo , Adulto , Anciano , Animales , Apoptosis/genética , Western Blotting , Estudios de Casos y Controles , Proliferación Celular/genética , Cromatografía Liquida , Ensayo Cometa , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Enzimas Reparadoras del ADN/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína Forkhead Box M1/metabolismo , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Oxidación-Reducción , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/metabolismo , Hipertensión Arterial Pulmonar/metabolismo , Pirofosfatasas/antagonistas & inhibidores , Pirofosfatasas/genética , Pirofosfatasas/metabolismo , ARN Mensajero/metabolismo , Ratas , Espectrometría de Masas en Tándem , Regulación hacia Arriba
2.
Arterioscler Thromb Vasc Biol ; 40(3): 783-801, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31969012

RESUMEN

OBJECTIVE: Pulmonary arterial hypertension (PAH) is a fatal disease characterized by the narrowing of pulmonary arteries (PAs). It is now established that this phenotype is associated with enhanced PA smooth muscle cells (PASMCs) proliferation and suppressed apoptosis. This phenotype is sustained in part by the activation of several DNA repair pathways allowing PASMCs to survive despite the unfavorable environmental conditions. PIM1 (Moloney murine leukemia provirus integration site) is an oncoprotein upregulated in PAH and involved in many prosurvival pathways, including DNA repair. The objective of this study was to demonstrate the implication of PIM1 in the DNA damage response and the beneficial effect of its inhibition by pharmacological inhibitors in human PAH-PASMCs and in rat PAH models. Approach and Results: We found in vitro that PIM1 inhibition by either SGI-1776, TP-3654, siRNA (silencer RNA) decreased the phosphorylation of its newly identified direct target KU70 (lupus Ku autoantigen protein p70) resulting in the inhibition of double-strand break repair (Comet Assay) by the nonhomologous end-joining as well as reduction of PAH-PASMCs proliferation (Ki67-positive cells) and resistance to apoptosis (Annexin V positive cells) of PAH-PASMCs. In vivo, SGI-1776 and TP-3654 given 3× a week, improved significantly pulmonary hemodynamics (right heart catheterization) and vascular remodeling (Elastica van Gieson) in monocrotaline and Fawn-Hooded rat models of PAH. CONCLUSIONS: We demonstrated that PIM1 phosphorylates KU70 and initiates DNA repair signaling in PAH-PASMCs and that PIM1 inhibitors represent a therapeutic option for patients with PAH.


Asunto(s)
Daño del ADN , Reparación del ADN por Unión de Extremidades , Hipertensión Pulmonar/enzimología , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Animales , Antihipertensivos/farmacología , Apoptosis , Proliferación Celular , Células Cultivadas , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Histonas/metabolismo , Humanos , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Autoantígeno Ku/metabolismo , Masculino , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Fosfoproteínas/metabolismo , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-pim-1/genética , Arteria Pulmonar/enzimología , Arteria Pulmonar/patología , Ratas Sprague-Dawley , Remodelación Vascular
3.
Arterioscler Thromb Vasc Biol ; 39(8): 1667-1681, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31092016

RESUMEN

OBJECTIVE: Pulmonary arterial hypertension (PAH) is a debilitating disease associated with progressive vascular remodeling of distal pulmonary arteries leading to elevation of pulmonary artery pressure, right ventricular hypertrophy, and death. Although presenting high levels of DNA damage that normally jeopardize their viability, pulmonary artery smooth muscle cells (PASMCs) from patients with PAH exhibit a cancer-like proproliferative and apoptosis-resistant phenotype accounting for vascular lumen obliteration. In cancer cells, overexpression of the serine/threonine-protein kinase CHK1 (checkpoint kinase 1) is exploited to counteract the excess of DNA damage insults they are exposed to. This study aimed to determine whether PAH-PASMCs have developed an orchestrated response mediated by CHK1 to overcome DNA damage, allowing cell survival and proliferation. Approach and Results: We demonstrated that CHK1 expression is markedly increased in isolated PASMCs and distal PAs from patients with PAH compared with controls, as well as in multiple complementary animal models recapitulating the disease, including monocrotaline rats and the simian immunodeficiency virus-infected macaques. Using a pharmacological and molecular loss of function approach, we showed that CHK1 promotes PAH-PASMCs proliferation and resistance to apoptosis. In addition, we found that inhibition of CHK1 induces downregulation of the DNA repair protein RAD 51 and severe DNA damage. In vivo, we provided evidence that pharmacological inhibition of CHK1 significantly reduces vascular remodeling and improves hemodynamic parameters in 2 experimental rat models of PAH. CONCLUSIONS: Our results show that CHK1 exerts a proproliferative function in PAH-PASMCs by mitigating DNA damage and suggest that CHK1 inhibition may, therefore, represent an attractive therapeutic option for patients with PAH.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada/fisiología , Células Cultivadas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/fisiología , Daño del ADN , Modelos Animales de Enfermedad , Humanos , Masculino , MicroARNs/fisiología , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/fisiología , Ratas , Ratas Sprague-Dawley
4.
J Mol Med (Berl) ; 96(2): 223-235, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29290032

RESUMEN

Pulmonary arterial hypertension (PAH) is a progressive vascular remodeling disease characterized by a persistent elevation of pulmonary artery pressure, leading to right heart failure and premature death. Exaggerated proliferation and resistance to apoptosis of pulmonary artery smooth muscle cells (PASMCs) is a key component of vascular remodeling. Despite major advances in the field, current therapies for PAH remain poorly effective in reversing the disease or significantly improving long-term survival. Because the transcription factor FOXM1 is necessary for PASMC proliferation during lung morphogenesis and its overexpression stimulates proliferation and evasion of apoptosis in cancer cells, we thus hypothesized that upregulation of FOXM1 in PAH-PASMCs promotes cell expansion and vascular remodeling. Our results showed that FOXM1 was markedly increased in distal pulmonary arteries and isolated PASMCs from PAH patients compared to controls as well as in two preclinical models. In vitro, we showed that miR-204 expression regulates FOXM1 levels and that inhibition of FOXM1 reduced cell proliferation and resistance to apoptosis through diminished DNA repair mechanisms and decreased expression of the pro-remodeling factor survivin. Accordingly, inhibition of FOXM1 with thiostrepton significantly improved established PAH in two rat models. Thus, we show for the first time that FOXM1 is implicated in PAH development and represents a new promising target. KEY MESSAGES: FOXM1 is overexpressed in human PAH-PASMCs and PAH animal models. FOXM1 promotes PAH-PASMC proliferation and resistance to apoptosis. Pharmacological inhibition of FOXM1 improves established PAH in the MCT and Su/Hx rat models. FOXM1 may be a novel therapeutic target in PAH.


Asunto(s)
Proteína Forkhead Box M1/fisiología , Hipertensión Pulmonar/metabolismo , Miocitos del Músculo Liso/fisiología , Arteria Pulmonar/fisiología , Animales , Línea Celular , Proliferación Celular , Proteína Forkhead Box M1/antagonistas & inhibidores , Humanos , Hipertensión Pulmonar/tratamiento farmacológico , Hipertrofia Ventricular Derecha/metabolismo , Masculino , MicroARNs/metabolismo , Arteria Pulmonar/citología , Ratas Sprague-Dawley , Tioestreptona/uso terapéutico , Remodelación Vascular
5.
Pulm Circ ; 8(1): 2045893217741429, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29064353

RESUMEN

Dysregulated metabolism and rarefaction of the capillary network play a critical role in pulmonary arterial hypertension (PAH) etiology. They are associated with a decrease in perfusion of the lungs, skeletal muscles, and right ventricle (RV). Previous studies suggested that endothelin-1 (ET-1) modulates both metabolism and angiogenesis. We hypothesized that dual ETA/ETB receptors blockade improves PAH by improving cell metabolism and promoting angiogenesis. Five weeks after disease induction, Sugen/hypoxic rats presented severe PAH with pulmonary artery (PA) remodeling, RV hypertrophy and capillary rarefaction in the lungs, RV, and skeletal muscles (microCT angiogram, lectin perfusion, CD31 staining). Two-week treatment with dual ETA/ETB receptors antagonist macitentan (30 mg/kg/d) significantly improved pulmonary hemodynamics, PA vascular remodeling, and RV function and hypertrophy compared to vehicle-treated animals (all P = 0.05). Moreover, macitentan markedly increased lung, RV and quadriceps perfusion, and microvascular density (all P = 0.05). In vitro, these effects were associated with increases in oxidative phosphorylation (oxPhox) and markedly reduced cell proliferation of PAH-PA smooth muscle cells (PASMCs) treated with macitentan without affecting apoptosis. While macitentan did not affect oxPhox, proliferation, and apoptosis of PAH-PA endothelial cells (PAECs), it significantly improved their angiogenic capacity (tube formation assay). Exposure of control PASMC and PAEC to ET-1 fully mimicked the PAH cells phenotype, thus confirming that ET-1 is implicated in both metabolism and angiogenesis abnormalities in PAH. Dual ETA/ETB receptor blockade improved the metabolic changes involved in PAH-PASMCs' proliferation and the angiogenic capacity of PAH-PAEC leading to an increased capillary density in lungs, RV, and skeletal muscles.

6.
Sci Rep ; 7(1): 4546, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28674407

RESUMEN

Pulmonary arterial hypertension (PAH) is a vascular remodeling disease with limited therapeutic options. Although exposed to stressful conditions, pulmonary artery (PA) smooth muscle cells (PASMCs) exhibit a "cancer-like" pro-proliferative and anti-apoptotic phenotype. HDAC6 is a cytoplasmic histone deacetylase regulating multiple pro-survival mechanisms and overexpressed in response to stress in cancer cells. Due to the similarities between cancer and PAH, we hypothesized that HDAC6 expression is increased in PAH-PASMCs to face stress allowing them to survive and proliferate, thus contributing to vascular remodeling in PAH. We found that HDAC6 is significantly up-regulated in lungs, distal PAs, and isolated PASMCs from PAH patients and animal models. Inhibition of HDAC6 reduced PAH-PASMC proliferation and resistance to apoptosis in vitro sparing control cells. Mechanistically, we demonstrated that HDAC6 maintains Ku70 in a hypoacetylated state, blocking the translocation of Bax to mitochondria and preventing apoptosis. In vivo, pharmacological inhibition of HDAC6 improved established PAH in two experimental models and can be safely given in combination with currently approved PAH therapies. Moreover, Hdac6 deficient mice were partially protected against chronic hypoxia-induced pulmonary hypertension. Our study shows for the first time that HDAC6 is implicated in PAH development and represents a new promising target to improve PAH.


Asunto(s)
Histona Desacetilasa 6/genética , Histona Desacetilasa 6/metabolismo , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Acetilación , Animales , Apoptosis/genética , Línea Celular , Movimiento Celular/genética , Proliferación Celular/genética , Modelos Animales de Enfermedad , Expresión Génica , Regulación de la Expresión Génica , Proteínas HSP90 de Choque Térmico/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/fisiopatología , Inmunohistoquímica , Autoantígeno Ku/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Miocitos del Músculo Liso/metabolismo , Ratas , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
7.
Am J Respir Crit Care Med ; 194(10): 1273-1285, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27149112

RESUMEN

RATIONALE: Pulmonary arterial hypertension (PAH) is characterized by excessive proliferation of pulmonary artery smooth muscle cells (PASMCs). This is sustained in time by the down-regulation of microRNA (miR)-204. In systemic vascular diseases, reduced miR-204 expression promotes vascular biomineralization by augmenting the expression of the transcription factor Runt-related transcription factor 2 (RUNX2). Implication of RUNX2 in PAH-related vascular remodeling and presence of calcified lesions in PAH remain unexplored. OBJECTIVES: We hypothesized that RUNX2 is up-regulated in lungs of patients with PAH, contributing to vascular remodeling and calcium-related biomineralization. METHODS: We harvested human lung tissues in which we assessed calcification lesions and RUNX2 expression. We also isolated PASMCs from these tissues for in vitro analyses. Using a bidirectional approach, we investigated the role for RUNX2 in cell proliferation, apoptosis, and calcification capacity. Ectopic delivery of small interfering RNA against RUNX2 was used in an animal model of PAH to evaluate the therapeutic potential of RUNX2 inhibition in this disease. MEASUREMENTS AND MAIN RESULTS: Patients with PAH display features of calcified lesions within the distal pulmonary arteries (PAs). We show that RUNX2 is up-regulated in lungs, distal PAs, and primary cultured human PASMCs isolated from PAH and compared with patients without PAH. RUNX2 expression histologically correlates with vascular remodeling and calcification. Using in vitro gain- and loss-of-function approaches, we mechanistically demonstrate that miR-204 diminution promotes RUNX2 up-regulation and that sustained RUNX2 expression activates hypoxia-inducible factor-1α, leading to aberrant proliferation, resistance to apoptosis, and subsequent transdifferentiation of PAH-PASMCs into osteoblast-like cells. In the PAH Sugen/hypoxia rat model, molecular RUNX2 inhibition reduces PA remodeling and prevents calcification, thus improving pulmonary hemodynamic parameters and right ventricular function. CONCLUSIONS: RUNX2 plays a pivotal role in the pathogenesis of PAH, contributing to the development of proliferative and calcified PA lesions. Inhibition of RUNX2 may therefore represent an attractive therapeutic strategy for PAH.


Asunto(s)
Proliferación Celular/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/fisiopatología , Calcificación Vascular/genética , Calcificación Vascular/fisiopatología , Adulto , Proliferación Celular/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad
8.
Circ Res ; 117(6): 525-35, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26224795

RESUMEN

RATIONALE: Pulmonary arterial hypertension (PAH) is a vasculopathy characterized by enhanced pulmonary artery (PA) smooth muscle cell (PASMC) proliferation and suppressed apoptosis. Decreased expression of microRNA-204 has been associated to this phenotype. By a still elusive mechanism, microRNA-204 downregulation promotes the expression of oncogenes, including nuclear factor of activated T cells, B-cell lymphoma 2, and Survivin. In cancer, increased expression of the epigenetic reader bromodomain-containing protein 4 (BRD4) sustains cell survival and proliferation. Interestingly, BRD4 is a predicted target of microRNA-204 and has binding sites on the nuclear factor of activated T cells promoter region. OBJECTIVE: To investigate the role of BRD4 in PAH pathogenesis. METHODS AND RESULTS: BRD4 is upregulated in lungs, distal PAs, and PASMCs of patients with PAH compared with controls. With mechanistic in vitro experiments, we demonstrated that BRD4 expression in PAH is microRNA-204 dependent. We further studied the molecular downstream targets of BRD4 by inhibiting its activity in PAH-PASMCs using a clinically available inhibitor JQ1. JQ1 treatment in PAH-PASMCs increased p21 expression, thus triggering cell cycle arrest. Furthermore, BRD4 inhibition, by JQ1 or siBRD4, decreased the expression of 3 major oncogenes, which are overexpressed in PAH: nuclear factor of activated T cells, B-cell lymphoma 2, and Survivin. Blocking this oncogenic signature led to decreased PAH-PASMC proliferation and increased apoptosis in a BRD4-dependent manner. Indeed, pharmacological JQ1 or molecular (siRNA) inhibition of BRD4 reversed this pathological phenotype in addition to restoring mitochondrial membrane potential and to increasing cells spare respiratory capacity. Moreover, BRD4 inhibition in vivo reversed established PAH in the Sugen/hypoxia rat model. CONCLUSIONS: BRD4 plays a key role in the pathological phenotype in PAH, which could offer new therapeutic perspectives for patients with PAH.


Asunto(s)
Epigénesis Genética/fisiología , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Proteínas Nucleares/biosíntesis , Arteria Pulmonar/metabolismo , Factores de Transcripción/biosíntesis , Adulto , Anciano , Animales , Proteínas de Ciclo Celular , Células Cultivadas , Femenino , Humanos , Hipertensión Pulmonar/patología , Masculino , Persona de Mediana Edad , Arteria Pulmonar/patología , Ratas
9.
Circulation ; 132(10): 932-43, 2015 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-26162916

RESUMEN

BACKGROUND: Right ventricular (RV) failure is the most important factor of both morbidity and mortality in pulmonary arterial hypertension (PAH). However, the underlying mechanisms resulting in the failed RV in PAH remain unknown. There is growing evidence that angiogenesis and microRNAs are involved in PAH-associated RV failure. We hypothesized that microRNA-126 (miR-126) downregulation decreases microvessel density and promotes the transition from a compensated to a decompensated RV in PAH. METHODS AND RESULTS: We studied RV free wall tissues from humans with normal RV (n=17), those with compensated RV hypertrophy (n=8), and patients with PAH with decompensated RV failure (n=14). Compared with RV tissues from patients with compensated RV hypertrophy, patients with decompensated RV failure had decreased miR-126 expression (quantitative reverse transcription-polymerase chain reaction; P<0.01) and capillary density (CD31(+) immunofluorescence; P<0.001), whereas left ventricular tissues were not affected. miR-126 downregulation was associated with increased Sprouty-related EVH1 domain-containing protein 1 (SPRED-1), leading to decreased activation of RAF (phosphorylated RAF/RAF) and mitogen-activated protein kinase (MAPK); (phosphorylated MAPK/MAPK), thus inhibiting the vascular endothelial growth factor pathway. In vitro, Matrigel assay showed that miR-126 upregulation increased angiogenesis of primary cultured endothelial cells from patients with decompensated RV failure. Furthermore, in vivo miR-126 upregulation (mimic intravenous injection) improved cardiac vascular density and function of monocrotaline-induced PAH animals. CONCLUSIONS: RV failure in PAH is associated with a specific molecular signature within the RV, contributing to a decrease in RV vascular density and promoting the progression to RV failure. More importantly, miR-126 upregulation in the RV improves microvessel density and RV function in experimental PAH.


Asunto(s)
Regulación hacia Abajo/fisiología , Insuficiencia Cardíaca/metabolismo , Hipertensión Pulmonar/metabolismo , MicroARNs/metabolismo , Disfunción Ventricular Derecha/metabolismo , Adulto , Anciano , Animales , Células Cultivadas , Femenino , Insuficiencia Cardíaca/diagnóstico , Humanos , Hipertensión Pulmonar/diagnóstico , Masculino , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley , Disfunción Ventricular Derecha/diagnóstico
10.
J Comp Neurol ; 522(12): 2707-28, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24610630

RESUMEN

Transplantation of human neural progenitor cells (NPCs) into the brain or spinal cord to replace lost cells, modulate the injury environment, or create a permissive milieu to protect and regenerate host neurons is a promising therapeutic strategy for neurological diseases. Deriving NPCs from human fetal tissue is feasible, although problematic issues include limited sources and ethical concerns. Here we describe a new and abundant source of NPCs derived from human induced pluripotent stem cells (iPSCs). A novel chopping technique was used to transform adherent iPSCs into free-floating spheres that were easy to maintain and were expandable (EZ spheres) (Ebert et al. [2013] Stem Cell Res 10:417-427). These EZ spheres could be differentiated towards NPC spheres with a spinal cord phenotype using a combination of all-trans retinoic acid (RA) and epidermal growth factor (EGF) and fibroblast growth factor-2 (FGF-2) mitogens. Suspension cultures of NPCs derived from human iPSCs or fetal tissue have similar characteristics, although they were not similar when grown as adherent cells. In addition, iPSC-derived NPCs (iNPCs) survived grafting into the spinal cord of athymic nude rats with no signs of overgrowth and with a very similar profile to human fetal-derived NPCs (fNPCs). These results suggest that human iNPCs behave like fNPCs and could thus be a valuable alternative for cellular regenerative therapies of neurological diseases.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Células-Madre Neurales/fisiología , Médula Espinal/citología , Análisis de Varianza , Animales , Acuaporina 4/metabolismo , Astrocitos/fisiología , Humanos , Antígeno Ki-67/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/trasplante , Neuronas/metabolismo , Ratas , Ratas Desnudas , Médula Espinal/cirugía , Transcriptoma/fisiología
11.
PLoS One ; 8(5): e64957, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23705021

RESUMEN

Type 2 diabetes is characterized by both insulin resistance and progressive deterioration of ß-cell function. The forkhead transcription factor FoxO1 is a prominent mediator of insulin signaling in ß-cells. We reasoned that identification of FoxO1 target genes in ß-cells could reveal mechanisms linking ß-cell dysfunction to insulin resistance. In this study, we report the characterization of Nov/Ccn3 as a novel transcriptional target of FoxO1 in pancreatic ß-cells. FoxO1 binds to an evolutionarily conserved response element in the Ccn3 promoter to regulate its expression. Accordingly, CCN3 levels are elevated in pancreatic islets of mice with overexpression of a constitutively active form of FoxO1 or insulin resistance. Our functional studies reveal that CCN3 impairs ß-cell proliferation concomitantly with a reduction in cAMP levels. Moreover, CCN3 decreases glucose oxidation, which translates into inhibition of glucose-stimulated Ca(2+) entry and insulin secretion. Our results identify CCN3, a novel transcriptional target of FoxO1 in pancreatic ß-cells, as a potential target for therapeutic intervention in the treatment of diabetes.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Células Secretoras de Insulina/metabolismo , Proteína Hiperexpresada del Nefroblastoma/metabolismo , Transcripción Genética , Animales , Secuencia de Bases , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Secuencia Conservada/genética , Modelos Animales de Enfermedad , Proteína Forkhead Box O1 , Glucosa/farmacología , Humanos , Insulina/metabolismo , Resistencia a la Insulina/genética , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/patología , Ratones , Proteína Hiperexpresada del Nefroblastoma/genética , Regiones Promotoras Genéticas , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
12.
PLoS One ; 7(4): e34932, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22523565

RESUMEN

Proliferation of glia and immune cells is a common pathological feature of many neurodegenerative diseases including amyotrophic lateral sclerosis (ALS). Here, to investigate the role of proliferating cells in motor neuron disease, SOD1(G93A) transgenic mice were treated intracerebroventicularly (i.c.v.) with the anti-mitotic drug cytosine arabinoside (Ara-C). I.c.v. delivery of Ara-C accelerated disease progression in SOD1(G93A) mouse model of ALS. Ara-C treatment caused substantial decreases in the number of microglia, NG2+ progenitors, Olig2+ cells and CD3+ T cells in the lumbar spinal cord of symptomatic SOD1(G93A) transgenic mice. Exacerbation of disease was also associated with significant alterations in the expression inflammatory molecules IL-1ß, IL-6, TGF-ß and the growth factor IGF-1.


Asunto(s)
Enfermedad de la Neurona Motora/complicaciones , Superóxido Dismutasa/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Proliferación Celular/efectos de los fármacos , Citarabina/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Interleucina-6/metabolismo , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
13.
Optom Vis Sci ; 84(10): 954-61, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18049361

RESUMEN

PURPOSE: To validate the biocompatibility and transmittance properties of contact lenses bearing intact liposomes. These liposomal lenses loaded with therapeutics can be used as ophthalmic drug delivery systems. METHODS: The biocompatibility of soft contact lenses, coated with liposomes was evaluated through in vitro direct and indirect cytocompatibility assays on human corneal epithelial cells, on reconstructed human corneas and on ex vivo rabbit corneas. The direct and indirect transmission spectra of liposome-covered lenses were also evaluated to test if they transmit all wavelengths of the ultraviolet-visible spectrum, to thereby fulfill their optical function, without gross alteration of the colors perception and with a minimum of light dispersion. RESULTS: Contact lenses bearing layers of stable liposomes did not induce any significant changes in cell viability and in cell growth, compared with lenses bearing no liposome. Elution assays revealed that no cytotoxic compound leaks from the lenses whether bearing liposomes or not. Histological analyses of reconstructed human corneas and ex vivo rabbit corneas directly exposed to liposomal lenses revealed neither alteration to the cell nor to the tissue structures. Contact lenses bearing layers of liposomes did not significantly affect light transmission compared with control lenses without liposome at the wavelength of maximal photopic sensitivity, i.e., 550 nm. In addition, the contact lenses afford more eye protection in the ultraviolet spectrum, compared with the control lenses. CONCLUSIONS: Liposomal contact lenses are biocompatible and their transmittance properties are not affected in the visible light range.


Asunto(s)
Lentes de Contacto Hidrofílicos , Luz , Liposomas , Ensayo de Materiales , Animales , División Celular , Supervivencia Celular , Células Cultivadas , Epitelio Corneal/citología , Epitelio Corneal/fisiología , Diseño de Equipo , Humanos , Óptica y Fotónica , Conejos
14.
Anticancer Res ; 24(4): 2169-77, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15330157

RESUMEN

BACKGROUND: Three-dimensional (3-D) culture systems that simulate the tumor extracellular microenvironment may be appropriate to test cancer cell potential for invasion and tumor cell sensitivity to anticancer drugs. MATERIALS AND METHODS: Human PC-3 prostate, A549 colon, HT-29 lung and MCF-7 and MDA-MB231 breast cancer cells were embedded and grown in collagen gel surrounded by a fibrin clot. Increasing concentrations of cisplatin, doxorubicin, paclitaxel and 5-fluorouracil were comparatively evaluated for their ability to inhibit tumor cell proliferation and colony formation in vitro. RESULTS: All cells, except MDA, formed colonies in collagen. PC-3, A549 and HT-29 cells massively invaded fibrin forming migratory fronts. Cell colonies were also formed in fibrin (secondary tumor-like structures) apart from migratory fronts; HT-29 cells were the most aggressive in this regard MDA cells were particularly sensitive to doxorubicin, while MCF-7 cells showed sensitivity to all anticancer regimens tested. A549 cells were the tumor cell type with greatest potential for invasion and were sensitive mostly to cisplatin. PC-3 cells were primarily sensitive to cisplatin and doxorubicin, while HT-29 cells were sensitive to fluorouracil and doxorubicin. CONCLUSION: 3-D collagen cell culture systems can be used to study cancer cell potential for invasion and their relative sensitivity/resistance to anticancer drugs.


Asunto(s)
Ensayos de Selección de Medicamentos Antitumorales/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Cisplatino/farmacología , Colágeno , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Doxorrubicina/farmacología , Fibrina , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Masculino , Invasividad Neoplásica , Paclitaxel/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...