Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 571(7764): 284-288, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31263273

RESUMEN

Hedgehog signalling is fundamental to embryonic development and postnatal tissue regeneration1. Aberrant postnatal Hedgehog signalling leads to several malignancies, including basal cell carcinoma and paediatric medulloblastoma2. Hedgehog proteins bind to and inhibit the transmembrane cholesterol transporter Patched-1 (PTCH1), which permits activation of the seven-transmembrane transducer Smoothened (SMO) via a mechanism that is poorly understood. Here we report the crystal structure of active mouse SMO bound to both the agonist SAG21k and to an intracellular binding nanobody that stabilizes a physiologically relevant active state. Analogous to other G protein-coupled receptors, the activation of SMO is associated with subtle motions in the extracellular domain, and larger intracellular changes. In contrast to recent models3-5, a cholesterol molecule that is critical for SMO activation is bound deep within the seven-transmembrane pocket. We propose that the inactivation of PTCH1 by Hedgehog allows a transmembrane sterol to access this seven-transmembrane site (potentially through a hydrophobic tunnel), which drives the activation of SMO. These results-combined with signalling studies and molecular dynamics simulations-delineate the structural basis for PTCH1-SMO regulation, and suggest a strategy for overcoming clinical resistance to SMO inhibitors.


Asunto(s)
Membrana Celular/química , Proteínas Hedgehog/agonistas , Transducción de Señal/efectos de los fármacos , Receptor Smoothened/agonistas , Receptor Smoothened/metabolismo , Esteroles/farmacología , Animales , Sitios de Unión , Técnicas Biosensibles , Dominio Catalítico/efectos de los fármacos , Membrana Celular/metabolismo , Colesterol/química , Colesterol/metabolismo , Colesterol/farmacología , Proteínas Hedgehog/metabolismo , Ligandos , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Receptor Patched-1/antagonistas & inhibidores , Receptor Patched-1/metabolismo , Conformación Proteica , Estabilidad Proteica , Anticuerpos de Cadena Única/inmunología , Receptor Smoothened/antagonistas & inhibidores , Receptor Smoothened/química , Esteroles/química , Esteroles/metabolismo , Proteínas de Xenopus/química
2.
J Biol Chem ; 298(12): 102705, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36400200

RESUMEN

The Sonic Hedgehog (SHh) precursor protein undergoes biosynthetic autoprocessing to cleave off and covalently attach cholesterol to the SHh signaling ligand, a vital morphogen and oncogenic effector protein. Autoprocessing is self-catalyzed by SHhC, the SHh precursor's C-terminal enzymatic domain. A method to screen for small molecule regulators of this process may be of therapeutic value. Here, we describe the development and validation of the first cellular reporter to monitor human SHhC autoprocessing noninvasively in high-throughput compatible plates. The assay couples intracellular SHhC autoprocessing using endogenous cholesterol to the extracellular secretion of the bioluminescent nanoluciferase enzyme. We developed a WT SHhC reporter line for evaluating potential autoprocessing inhibitors by concentration response-dependent suppression of extracellular bioluminescence. Additionally, a conditional mutant SHhC (D46A) reporter line was developed for identifying potential autoprocessing activators by a concentration response-dependent gain of extracellular bioluminescence. The D46A mutation removes a conserved general base that is critical for the activation of the cholesterol substrate. Inducibility of the D46A reporter was established using a synthetic sterol, 2-α carboxy cholestanol, designed to bypass the defect through intramolecular general base catalysis. To facilitate direct nanoluciferase detection in the cell culture media of 1536-well plates, we designed a novel anionic phosphonylated coelenterazine, CLZ-2P, as the nanoluciferase substrate. This new reporter system offers a long-awaited resource for small molecule discovery for cancer and for developmental disorders where SHh ligand biosynthesis is dysregulated.


Asunto(s)
Proteínas Hedgehog , Humanos , Colesterol/metabolismo , Proteínas Hedgehog/agonistas , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/metabolismo , Ligandos , Proteínas Oncogénicas , Esteroles
3.
Cancer Sci ; 112(5): 1798-1810, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33629464

RESUMEN

The G-protein-coupled receptor 126 (GPR126) may play an important role in tumor development, although its role remains poorly understood. We found that GPR126 had higher expression in most colorectal cancer cell lines than in normal colon epithelial cell lines, and higher expression levels in colorectal cancer tissues than in normal adjacent colon tissues. GPR126 knockdown induced by shRNA inhibited cell viability and colony formation in HT-29, HCT116, and LoVo cells, decreased BrdU incorporation into newly synthesized proliferating HT-29 cells, led to an arrest of cell cycle progression at the G1 phase in HCT-116 and HT-29 cells, and suppressed tumorigenesis of HT-29, HCT116, and LoVo cells in nude mouse xenograft models. GPR126 knockdown engendered decreased transcription and translation of histone deacetylase 2 (HDAC2), previously implicated in the activation of GLI1 and GLI2 in the Hedgehog signaling pathway. Ectopic expression of HDAC2 in GPR126-silenced cells restored cell viability and proliferation, GLI2 luciferase reporter activity, partially recovered GLI2 expression, and reduced the cell cycle arrest. HDAC2 regulated GLI2 expression and, along with GLI2, it bound to the PTCH1 promoter, as evidenced by a chip assay with HT-29 cells. Purmorphamine, a hedgehog agonist, largely restored the cell viability and expression of GLI2 proteins in GPR126-silenced HT-29 cells, whereas GANT61, a hedgehog inhibitor, further enhanced the GPR126 knockdown-induced inhibitory effects. Our findings demonstrate that GPR126 regulates colorectal cancer cell proliferation by mediating the expression of HDAC2 and GLI2, therefore it may represent a suitable therapeutic target for colorectal cancer treatment.


Asunto(s)
Proliferación Celular/fisiología , Neoplasias Colorrectales/metabolismo , Histona Desacetilasa 2/metabolismo , Proteínas Nucleares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteína Gli2 con Dedos de Zinc/metabolismo , Animales , Bromodesoxiuridina/metabolismo , Ciclo Celular/genética , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Supervivencia Celular/fisiología , Colon/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/terapia , ADN/biosíntesis , Fase G1 , Técnicas de Silenciamiento del Gen , Células HT29 , Proteínas Hedgehog/agonistas , Proteínas Hedgehog/antagonistas & inhibidores , Proteínas Hedgehog/metabolismo , Xenoinjertos , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Desnudos , Morfolinas/farmacología , Proteínas de Neoplasias/metabolismo , Trasplante de Neoplasias , Receptor Patched-1/metabolismo , Purinas/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , ARN Mensajero/metabolismo , Receptores Acoplados a Proteínas G/genética
4.
J Cell Physiol ; 233(11): 8962-8970, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29856487

RESUMEN

Early loss of up to 50% of cells is common for in vitro chondrogenesis of mesenchymal stromal cells (MSC) in pellet culture, reducing the efficacy and the tissue yield for cartilage engineering. Enhanced proliferation could compensate for this unwanted effect, but relevant signaling pathways remain largely unknown. The aim of this study was to identify the contribution of bone morphogenetic protein (BMP), fibroblast growth factor (FGF), insulin-like growth factor (IGF), and hedgehog (HH) signaling toward cell proliferation during chondrogenesis and investigate whether a further mitogenic stimulation is possible and promising. Human MSC were subjected to chondrogenesis in the presence or absence of pathway inhibitors or activators up to Day 14 or from Days 14 to 28, before proliferation, DNA and proteoglycan content were quantified. [3H]-thymidine incorporation revealed arrest of proliferation on Day 3, after which cell division was reinitiated. Although BMP signaling was essential for proliferation throughout chondrogenesis, IGF signaling was relevant only up to Day 14. In contrast, FGF and HH signaling drove proliferation only from Day 14 onward. Early BMP4, IGF-1, or FGF18 treatment neither prevented early cell loss nor allowed further mitogenic stimulation. However, application of the HH-agonist purmorphamine from Day 14 increased proliferation 1.44-fold (p < 0.05) and late BMP4-application enhanced the DNA and proteoglycan content, with significant effects on tissue yield. Conclusively, a differential and phase-dependent contribution of the four pathways toward proliferation was uncovered and BMP4 treatment was promising to enhance tissue yield. Culture forms less prone to size limitations by nutrient/oxygen gradients and a focus on early apoptosis prevention may be considered as the next steps to further enhance chondrocyte formation from MSC.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Condrogénesis/genética , Células Madre Mesenquimatosas/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteína Morfogenética Ósea 4/genética , Cartílago/efectos de los fármacos , Cartílago/crecimiento & desarrollo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Proteínas Hedgehog/agonistas , Proteínas Hedgehog/genética , Humanos , Factor I del Crecimiento Similar a la Insulina/agonistas , Factor I del Crecimiento Similar a la Insulina/genética , Células Madre Mesenquimatosas/efectos de los fármacos , Morfolinas/farmacología , Purinas/farmacología , Transducción de Señal/efectos de los fármacos
5.
Cerebellum ; 17(2): 213-227, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29134361

RESUMEN

The cerebellum undergoes rapid growth during the third trimester and is vulnerable to injury and deficient growth in infants born prematurely. Factors associated with preterm cerebellar hypoplasia include chronic lung disease and postnatal glucocorticoid administration. We modeled chronic hypoxemia and glucocorticoid administration in neonatal mice to study whole cerebellar and cell type-specific effects of dual exposure. Chronic neonatal hypoxia resulted in permanent cerebellar hypoplasia. This was compounded by administration of prednisolone as shown by greater volume loss and Purkinje cell death. In the setting of hypoxia and prednisolone, administration of a small molecule Smoothened-Hedgehog agonist (SAG) preserved cerebellar volume and protected against Purkinje cell death. Such protective effects were observed even when SAG was given as a one-time dose after dual insult. To model complex injury and determine cell type-specific roles for the hypoxia inducible factor (HIF) pathway, we performed conditional knockout of von Hippel Lindau (VHL) to hyperactivate HIF1α in cerebellar granule neuron precursors (CGNP) or Purkinje cells. Surprisingly, HIF activation in either cell type resulted in no cerebellar deficit. However, in mice administered prednisolone, HIF overactivation in CGNPs resulted in significant cerebellar hypoplasia, whereas HIF overactivation in Purkinje cells caused cell death. Together, these findings indicate that HIF primes both cell types for injury via glucocorticoids, and that hypoxia/HIF + postnatal glucocorticoid administration act on distinct cellular pathways to cause cerebellar injury. They further suggest that SAG is neuroprotective in the setting of complex neonatal cerebellar injury.


Asunto(s)
Antiinflamatorios/uso terapéutico , Cerebelo/anomalías , Ciclohexilaminas/uso terapéutico , Proteínas Hedgehog/agonistas , Proteínas Hedgehog/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Tiofenos/uso terapéutico , Aminoácidos Dicarboxílicos/farmacología , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Cerebelo/efectos de los fármacos , Discapacidades del Desarrollo/etiología , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Glucocorticoides/farmacología , Hipoxia Encefálica/complicaciones , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Malformaciones del Sistema Nervioso/etiología , Prednisolona/uso terapéutico , Células de Purkinje/efectos de los fármacos , Células de Purkinje/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
6.
Cell Mol Life Sci ; 74(19): 3481-3490, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28523343

RESUMEN

Myocardial infarction (MI) is caused by the occlusion of a coronary artery due to underlying atherosclerosis complicated by localized thrombosis. The blockage of blood flow leads to cardiomyocyte (CM) death in the infarcted area. Adult mammalian cardiomyocytes have little capacity to proliferate in response to injury; however, some pathways active during embryogenesis and silent during adult life are recruited in response to tissue injury. One such example is hedgehog (Hh) signaling. Hh is involved in the embryonic development of the heart and coronary vascular system. Pathological conditions including ischemia activate Hh signaling in adult tissues. This review highlights the involvement of Hh signaling in ischemic tissue regeneration with a particular emphasis on heart regeneration and discusses its potential role as a therapeutic agent.


Asunto(s)
Corazón/fisiología , Proteínas Hedgehog/metabolismo , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/terapia , Regeneración , Transducción de Señal , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Técnicas de Reprogramación Celular/métodos , Descubrimiento de Drogas , Corazón/efectos de los fármacos , Proteínas Hedgehog/agonistas , Humanos , Terapia Molecular Dirigida , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Regeneración/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
7.
Proc Natl Acad Sci U S A ; 112(15): 4678-83, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25825734

RESUMEN

Hedgehog (Hh) signaling is essential for osteoblast differentiation in the endochondral skeleton during embryogenesis. However, the molecular mechanism underlying the osteoblastogenic role of Hh is not completely understood. Here, we report that Hh markedly induces the expression of insulin-like growth factor 2 (Igf2) that activates the mTORC2-Akt signaling cascade during osteoblast differentiation. Igf2-Akt signaling, in turn, stabilizes full-length Gli2 through Serine 230, thus enhancing the output of transcriptional activation by Hh. Importantly, genetic deletion of the Igf signaling receptor Igf1r specifically in Hh-responding cells diminishes bone formation in the mouse embryo. Thus, Hh engages Igf signaling in a positive feedback mechanism to activate the osteogenic program.


Asunto(s)
Diferenciación Celular , Proteínas Hedgehog/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Osteoblastos/metabolismo , Animales , Western Blotting , Línea Celular , Retroalimentación Fisiológica/efectos de los fármacos , Femenino , Proteínas Hedgehog/agonistas , Proteínas Hedgehog/genética , Hibridación in Situ , Factor II del Crecimiento Similar a la Insulina/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones Noqueados , Ratones Transgénicos , Morfolinas/farmacología , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Osteoblastos/citología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Purinas/farmacología , Interferencia de ARN , Receptores de Somatomedina/genética , Receptores de Somatomedina/metabolismo , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteína con Dedos de Zinc GLI1
8.
Perfusion ; 33(3): 209-218, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29065787

RESUMEN

OBJECTIVE: The present study was been designed to investigate the role and pharmacological potential of hedgehog in oestrogen-deficient rat heart. METHODS: Oestrogen deficiency was produced in female Wistar rats by the surgical removal of both ovaries and these animals were used four weeks later. Isolated rat heart was subjected to 30 min ischaemia followed by 120 min of reperfusion (I/R). The heart was subjected to pharmacological preconditioning with the hedgehog agonist purmorphamine (1µM) and GDC-0449, a hedgehog antagonist, in the last episode of reperfusion before I/R. Myocardial infarction was assessed in terms of the increase in lactate dehydrogenase (LDH), creatinine kinase-MB (CK-MB), myeloperoxidase (MPO) level and infarct size (triphenyltetrazolium chloride staining). Immunohistochemistry analysis was done for the assessment of tumour necrosis factor (TNF)-α level in cardiac tissue. eNOS expression was estimated by rt-PCR. RESULTS: Pharmacological preconditioning with purmorphamine significantly attenuated I/R-induced myocardial infarction, TNF-α, MPO level and release of LDH and CK-MB compared to the I/R control group. However, GDC-0449 prevented the ameliorative preconditioning effect of estradiol. CONCLUSION: It may be concluded that the hedgehog agonist purmorphamine prevents the ovariectomised heart from ischaemic reperfusion injury.


Asunto(s)
Cardiotónicos/uso terapéutico , Corazón/efectos de los fármacos , Proteínas Hedgehog/agonistas , Morfolinas/uso terapéutico , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Purinas/uso terapéutico , Animales , Femenino , Corazón/fisiopatología , Infarto del Miocardio/fisiopatología , Daño por Reperfusión Miocárdica/fisiopatología , Ovariectomía , Ratas Wistar
9.
Stroke ; 48(6): 1636-1645, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28487338

RESUMEN

BACKGROUND AND PURPOSE: Because of the limitation in treatment window of the r-tPA (recombinant tissue-type plasminogen activator), the development of delayed treatment for stroke is needed. In this study, we examined the efficacy of delayed poststroke treatment (post 3-8 days) of the sonic hedgehog pathway agonist on functional recovery and the underlying mechanisms. METHODS: We evaluated functional recovery at 1 month after stroke using locomotion analysis and Barnes maze test for cognitive function. We used a genetically inducible neural stem cell-specific reporter mouse line (nestin-CreERT2-R26R-YFP) to label and track their proliferation, survival, and differentiation in ischemic brain. Brain tissue damage, angiogenesis, and cerebral blood flow recovery was evaluated using magnetic resonance imaging techniques and immunostaining. RESULTS: Our results show that delayed treatment of sonic hedgehog pathway agonist in stroke mice results in enhanced functional recovery both in locomotor function and in cognitive function at 1 month after stroke. Furthermore, using the Nestincre-ERT2-YFP mice, we showed that poststroke sonic hedgehog pathway agonist treatment increased surviving newly born cells derived from both subventricular zone and subgranular zone neural stem cells, total surviving DCX+ (Doublecortin) neuroblast cells, and neurons (NeuN+/YFP+) in the ischemic brain. Sonic hedgehog pathway agonist treatment also improved the brain tissue repair in ischemic region supported by our T2-weighted magnetic resonance imaging, cerebral blood flow map by arterial spin labeling, and immunohistochemistry (α-smooth muscle actin and CD31 immunostaining). CONCLUSIONS: These data confirm an important role for the hedgehog pathway in poststroke brain repair and functional recovery, suggesting a prolonged treatment window for potential treatment strategy to modulate sonic hedgehog pathway after stroke.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Proteínas Hedgehog/agonistas , Proteínas Hedgehog/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Recuperación de la Función/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Conducta Animal , Modelos Animales de Enfermedad , Proteína Doblecortina , Proteínas Hedgehog/administración & dosificación , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Actividad Motora , Nestina , Células-Madre Neurales
10.
Cerebellum ; 16(2): 450-461, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27600805

RESUMEN

Granule neuron precursors (GNPs) proliferate under the influence of Sonic hedgehog (Shh) that is secreted by Purkinje neurons during early postnatal cerebellar development. To investigate microRNA (miRNA) function in this developmental process, we conditionally deleted the Dicer1 gene under the activity of human glial fibrillary acidic protein (hGFAP) promoter. We report that Dicer1-ablated GNPs display decreased proliferation and survival at early postnatal stages and that the proliferation defect of mutant GNPs cannot be rescued by treatment of an Shh agonist in vitro as assayed by 5-bromo-2'-deoxyuridine (BrdU) pulse labeling and Shh target gene expression detection. Further analysis reveals that the expression of distinct cell cycle regulator genes including cell cycle inhibitor, CDKN1a (p21), selectively increases in Dicer1-ablated GNPs. Subsequently, we demonstrate that miR-17-5p exhibits high expression level in the developing cerebellum and that transfection of a synthetic miR-17-5p mimic downregulates p21 protein expression in GNPs and promotes proliferation of GNPs in culture. Therefore, Dicer1 ablation impairs Shh-induced GNP proliferation by disrupting the expression of distinct cell cycle regulator genes that are targets of miR-17∼92 cluster members. This study establishes a molecular link between miRNAs and cell cycle progression in the proliferating GNPs during normal cerebellar development and may facilitate miRNA application in treating medulloblastoma.


Asunto(s)
Proliferación Celular/fisiología , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , ARN Helicasas DEAD-box/deficiencia , Proteínas Hedgehog/metabolismo , Células-Madre Neurales/metabolismo , Ribonucleasa III/deficiencia , Animales , Ciclo Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Cerebelo/patología , ARN Helicasas DEAD-box/genética , Regulación del Desarrollo de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Hedgehog/agonistas , Humanos , Ratones Transgénicos , MicroARNs/metabolismo , Células-Madre Neurales/patología , Neurogénesis/fisiología , Neuronas/metabolismo , Neuronas/patología , Regiones Promotoras Genéticas , Ribonucleasa III/genética
11.
J Mol Cell Cardiol ; 93: 57-66, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26906632

RESUMEN

Sonic hedgehog (Shh) signaling plays an important role for early heart development, such as heart looping and cardiomyogenesis of pluripotent stem cells. A multifunctional receptor Cdo functions as a Shh coreceptor together with Boc and Gas1 to activate Shh signaling and these coreceptors seem to play compensatory roles in early heart development. Thus in this study, we examined the role of Cdo in cardiomyogenesis by utilizing an in vitro differentiation of pluripotent stem cells. Here we show that Cdo is required for efficient cardiomyogenesis of pluripotent stem cells by activation of Shh signaling. Cdo is induced concurrently with Shh signaling activation upon induction of cardiomyogenesis of P19 embryonal carcinoma (EC) cells. Cdo-depleted P19 EC and Cdo(-/-) mouse embryonic stem (ES) cells display decreased expression of key cardiac regulators, including Gata4, Nkx2.5 and Mef2c and this decrease coincides with reduced Shh signaling activities. Furthermore Cdo deficiency causes a stark reduction in formation of mature contractile cardiomyocytes. This defect in cardiomyogenesis is overcome by reactivation of Shh signaling at the early specification stage of cardiomyogenesis. The Shh agonist treatment restores differentiation capacities of Cdo-deficient ES cells into contractile cardiomyocytes by recovering both the expression of early cardiac regulators and structural genes such as cardiac troponin T and Connexin 43. Therefore Cdo is required for efficient cardiomyogenesis of pluripotent stem cells and an excellent target to improve the differentiation potential of stem cells for generation of transplantable cells to treat cardiomyopathies.


Asunto(s)
Moléculas de Adhesión Celular/genética , Corazón/embriología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Organogénesis , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Diferenciación Celular , Conexina 43/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Técnicas de Inactivación de Genes , Proteínas Hedgehog/agonistas , Proteínas Hedgehog/metabolismo , Ratones , Contracción Miocárdica/genética , Organogénesis/genética , Transducción de Señal
12.
Cell Physiol Biochem ; 39(3): 1129-40, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27576501

RESUMEN

BACKGROUND/AIMS: Lipid accumulation, inflammatory responses and oxidative stress have been implicated in the pathology of alcoholic liver disease (ALD). Targeting inhibition of these features may provide a promising therapeutic strategy for ALD. Baicalin, a flavonoid isolated from Scutellaria baicalensis Georgi, has been shown to exert a hepatoprotective effect. However, its effects on ALD remain obscure. This study was aimed to investigate the effects of baicalin on alcohol-induced liver injury and its related mechanisms. METHODS: For in vivo experiments, rats were supplied intragastrical administration of alcohol continuously for 4 or 8 weeks, and then received baicalin treatment in the latter 4 weeks in the presence / absence of alcohol intake. Liver histology and function, inflammatory cytokines, oxidative mediators, and the components of the Sonic hedgehog pathway were evaluated. For in vitro experiments, alcohol-stimulated human normal liver cells LO2 were used. RESULTS: Baicalin treatment significantly alleviated alcoholic liver injury, improved liver function impaired by alcohol, and inhibited hepatocytes apoptosis. In addition, baicalin decreased the expression levels of proinflammatory cytokines TNF-α, IL-1ß, IL-6) and malonyldialdehyde (MDA), and increased the activities of antioxidant enzymes SOD and GSH-Px. Furthermore, baicalin modulated the activation of Sonic hedgehog (Shh) pathway. Administration of baicalin upregulated the expression of sonic hedgehog (Shh), patched (Ptc), Smoothened (Smo), and Glioblastoma-1(Gli-1). Blockade of the Shh pathway in cyclopamine abolished the effects of baicalin in vitro. CONCLUSION: Both in vivo and in vitro experimental results indicate that baicalin exerts hepatoprotective roles in alcohol-induced liver injury through inhibiting oxidative stress, inflammatory response, and the regulation of the Shh pathway.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Flavonoides/farmacología , Proteínas Hedgehog/agonistas , Hepatopatías Alcohólicas/tratamiento farmacológico , Hígado/efectos de los fármacos , Sustancias Protectoras/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Modelos Animales de Enfermedad , Etanol , Regulación de la Expresión Génica , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Hígado/patología , Hepatopatías Alcohólicas/genética , Hepatopatías Alcohólicas/metabolismo , Hepatopatías Alcohólicas/patología , Pruebas de Función Hepática , Masculino , Malondialdehído/metabolismo , Receptor Patched-1/genética , Receptor Patched-1/metabolismo , Ratas , Ratas Wistar , Scutellaria baicalensis/química , Transducción de Señal , Receptor Smoothened/genética , Receptor Smoothened/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
13.
Biochem Biophys Res Commun ; 479(4): 772-778, 2016 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-27693795

RESUMEN

Bone fracture healing is processed through multiple biological stages including the transition from cartilaginous callus to bony callus formation. Because of its specific, temporal and indispensable functions demonstrated by mouse genetic studies, Hedgehog (Hh) signaling is one of the most potent signaling pathways involved in these processes, but the effect of Hh-signaling activation by small compounds on the repair process had not yet been addressed. Here we examined therapeutic effects of local and one shot-administration of the Hh agonist known as smoothened agonist (SAG) on bone fracture healing in a mouse model. A quantitative analysis with three-dimensional micro-computed tomography showed that SAG administration increased the size of both the cartilaginous callus and bony callus at 14 days after the surgery. A histological analysis showed that SAG administration increased the number of cells expressing a proliferation marker and a chondrocyte marker in cartilaginous callus as well as the cells expressing an osteoblast marker in bony callus. These results indicate that the SAG administration resulted in an enhancement of callus formation during bone fracture healing, which is at least in part mediated by an increase in chondrocyte proliferation in cartilaginous callus and the promotion of bone formation in bony callus. Therapeutic strategies with a SAG-mediated protocol may thus be useful for the treatment of bone fractures.


Asunto(s)
Ciclohexilaminas/administración & dosificación , Curación de Fractura/efectos de los fármacos , Proteínas Hedgehog/agonistas , Tiofenos/administración & dosificación , Animales , Densidad Ósea/efectos de los fármacos , Callo Óseo/efectos de los fármacos , Callo Óseo/metabolismo , Callo Óseo/patología , Condrocitos/efectos de los fármacos , Condrocitos/patología , Modelos Animales de Enfermedad , Curación de Fractura/fisiología , Imagenología Tridimensional , Masculino , Ratones , Ratones Endogámicos C57BL , Fracturas de la Tibia/diagnóstico por imagen , Fracturas de la Tibia/tratamiento farmacológico , Fracturas de la Tibia/patología , Microtomografía por Rayos X
14.
Development ; 140(3): 660-6, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23293297

RESUMEN

Adult mammalian cardiomyocytes have little capacity to proliferate in response to injury, a deficiency that underlies the poor regenerative ability of human hearts after myocardial infarction. By contrast, zebrafish regenerate heart muscle after trauma by inducing proliferation of spared cardiomyocytes, providing a model for identifying manipulations that block or enhance these events. Although direct genetic or chemical screens of heart regeneration in adult zebrafish present several challenges, zebrafish embryos are ideal for high-throughput screening. Here, to visualize cardiomyocyte proliferation events in live zebrafish embryos, we generated transgenic zebrafish lines that employ fluorescent ubiquitylation-based cell cycle indicator (FUCCI) technology. We then performed a chemical screen and identified several small molecules that increase or reduce cardiomyocyte proliferation during heart development. These compounds act via Hedgehog, Insulin-like growth factor or Transforming growth factor ß signaling pathways. Direct examination of heart regeneration after mechanical or genetic ablation injuries indicated that these pathways are activated in regenerating cardiomyocytes and that they can be pharmacologically manipulated to inhibit or enhance cardiomyocyte proliferation during adult heart regeneration. Our findings describe a new screening system that identifies molecules and pathways with the potential to modify heart regeneration.


Asunto(s)
Proliferación Celular , Corazón/fisiología , Ensayos Analíticos de Alto Rendimiento/métodos , Miocitos Cardíacos/citología , Regeneración , Animales , Animales Modificados Genéticamente/embriología , Animales Modificados Genéticamente/metabolismo , Animales Modificados Genéticamente/fisiología , Biomarcadores/metabolismo , Catecoles/farmacología , Recuento de Células , Ciclohexilaminas/farmacología , Embrión no Mamífero/citología , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/fisiología , Femenino , Corazón/embriología , Proteínas Hedgehog/agonistas , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Isoquinolinas/farmacología , Masculino , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Tiofenos/farmacología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Transgenes , Ubiquitinación , Pez Cebra/genética , Pez Cebra/lesiones , Pez Cebra/fisiología
15.
Bull Exp Biol Med ; 161(5): 674-678, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27709388

RESUMEN

We studied the effect of an activator (ShTh) and an inhibitor (cyclopamine) of the Hedgehog signaling pathway on proliferation of human glioma cell lines U87-MG and U251-MG and cultured human astrocytes. The Hedgehog signaling pathway is activated in glioma cells, but not in cultured human astrocytes. Experiments with Shh and cyclopamine can serve as an additional criterion for assessing activity of Hedgehog signaling in known cell lines and primary cultured cells.


Asunto(s)
Proliferación Celular , Glioblastoma/metabolismo , Transducción de Señal , Astrocitos/fisiología , Línea Celular Tumoral , Glioblastoma/patología , Proteínas Hedgehog/agonistas , Proteínas Hedgehog/metabolismo , Humanos , Ligandos , Alcaloides de Veratrum/farmacología
16.
J Cell Physiol ; 230(4): 922-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25215620

RESUMEN

Hedgehog (Hh) family members are involved in multiple cellular processes including proliferation, migration, differentiation, and cell fate determination. Recently, the novel Hh agonists Hh-Ag 1.3 and 1.7 were identified in a high-throughput screening of small molecule compounds that activate the expression of Gli1, a target of Hh signaling. This study demonstrates that Hh-Ag 1.3 and 1.7 strongly activate the expression of endogenous Gli1 and promote osteoblast differentiation in the mesenchymal stem cell line C3H10T1/2. Both compounds stimulated alkaline phosphatase activity in a dose-dependent manner, and induced osteoblast marker gene expression in C3H10T1/2 cells, which indicated that they had acquired an osteoblast identity. Of the markers, the expression of osterix/Sp7, a downstream target of runt-related transcription factor (Runx)2, was induced by Hh-Ag 1.7, which also rescued the osteoblast differentiation defect of RD-127, a mesenchymal cell line from Runx2-deficient mice. Hh-Ags also activated canonical Wnt signaling and synergized with low doses of BMP-2 to enhance osteoblastic potential. Thus, Hh-Ag 1.7 could be useful for bone healing in individuals with abnormalities in osteogenesis, such as osteoporosis patients and the elderly, and can contribute to the development of novel therapeutics for the treatment of bone fractures and defects.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proteínas Hedgehog/agonistas , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Animales , Proteína Morfogenética Ósea 2/efectos de los fármacos , Células Cultivadas , Células Madre Mesenquimatosas/citología , Ratones , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Receptores de Superficie Celular/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/efectos de los fármacos
17.
J Neurosci ; 33(39): 15408-13, 2013 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-24068809

RESUMEN

Humans with Down syndrome (DS) and Ts65Dn mice both show a reduced volume of the cerebellum due to a significant reduction in the density of granule neurons. Recently, cerebellar hypoplasia in Ts65Dn mice was rescued by a single treatment with SAG, an agonist of the Sonic hedgehog pathway, administered on the day of birth. In addition to normalizing cerebellar morphology, this treatment restored the ability to learn a spatial navigation task, which is associated with hippocampal function. It is not clear to what extent this improved performance results from restoration of the cerebellar architecture or a yet undefined role of Sonic hedgehog (Shh) in perinatal hippocampal development. The absence of a clearly demonstrated deficit in cerebellar function in trisomic mice exacerbates the problem of discerning how SAG acts to improve learning and memory. Here we show that phase reversal adaptation and consolidation of the vestibulo-ocular reflex is significantly impaired in Ts65Dn mice, providing for the first time a precise characterization of cerebellar functional deficits in this murine model of DS. However, these deficits do not benefit from the normalization of cerebellar morphology following treatment with SAG. Together with the previous observation that the synaptic properties of Purkinje cells are also unchanged by SAG treatment, this lack of improvement in a region-specific behavioral assay supports the possibility that a direct effect of Shh pathway stimulation on the hippocampus might explain the benefits of this potential approach to the improvement of cognition in DS.


Asunto(s)
Cerebelo/fisiopatología , Ciclohexilaminas/uso terapéutico , Síndrome de Down/tratamiento farmacológico , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Tiofenos/uso terapéutico , Animales , Cerebelo/patología , Ciclohexilaminas/farmacología , Modelos Animales de Enfermedad , Proteínas Hedgehog/agonistas , Ratones , Células de Purkinje/patología , Reflejo Vestibuloocular/efectos de los fármacos , Sinapsis/patología , Tiofenos/farmacología
18.
Hepatology ; 58(3): 995-1010, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23504944

RESUMEN

UNLABELLED: Hedgehog (Hh) signaling plays an important role in embryonic development and in the regulation of a variety of cellular functions. Aberrant activation of Hh signaling has been implicated in several human cancers including hepatocellular carcinoma (HCC). In this study we examined the pathobiological functions and molecular mechanisms of the Hh signaling pathway in HCC cells. Treatment of cultured human HCC cells (Huh7, Hep3B, and HepG2) with the Hh signaling ligand (recombinant Shh) or agonist, SAG and purmorphamine, prevented the induction of autophagy. In contrast, GANT61 (a small molecule inhibitor of Gli1 and Gli2) induced autophagy, as determined by immunoblotting for microtubule-associated protein light chain 3 (LC3) and p62, GFP-LC3 puncta, monodansylcadaverine (MDC) staining, and transmission electron microscopy. Hh inhibition-induced autophagy was associated with up-regulation of Bnip3, as determined by immunoblotting and real-time polymerase chain reaction (PCR) assay. Knockdown of Bnip3 by RNAi impaired GANT61-induced autophagy. Additionally, Hh inhibition-induced autophagy was associated with Bnip3-mediated displacement of Bcl-2 from Beclin-1, as determined by immunoblotting and immunoprecipitation assays. Furthermore, inhibition of Hh signaling increased HCC cell apoptosis and decreased cell viability, as determined by caspase and WST-1 assays. Pharmacological or genetic inhibition of autophagy by 3-methyladenine (3-MA) or Beclin-1 small interfering RNA (siRNA) partially suppressed GANT61-induced cell apoptosis and cytotoxicity. In a tumor xenograft model using SCID mice inoculated with Huh7 cells, administration of GANT61 inhibited tumor formation and decreased tumor volume; this effect was partially blocked by the autophagy inhibitor, 3-MA. CONCLUSION: These findings provide novel evidence that Hh inhibition induces autophagy through up-regulation of Bnip3 and that this mechanism contributes to apoptosis. Therefore, the status of autophagy is a key factor that determines the therapeutic response to Hh-targeted therapies.


Asunto(s)
Autofagia/fisiología , Carcinoma Hepatocelular/metabolismo , Proteínas Hedgehog/metabolismo , Neoplasias Hepáticas/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/efectos de los fármacos , Beclina-1 , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Células Cultivadas , Ciclohexilaminas/farmacología , Proteínas Hedgehog/agonistas , Humanos , Neoplasias Hepáticas/patología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones SCID , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Piridinas/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Tiofenos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cells ; 13(8)2024 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-38667291

RESUMEN

Both Hedgehog and androgen signaling pathways are known to promote myelin regeneration in the central nervous system. Remarkably, the combined administration of agonists of each pathway revealed their functional cooperation towards higher regeneration in demyelination models in males. Since multiple sclerosis, the most common demyelinating disease, predominates in women, and androgen effects were reported to diverge according to sex, it seemed essential to assess the existence of such cooperation in females. Here, we developed an intranasal formulation containing the Hedgehog signaling agonist SAG, either alone or in combination with testosterone. We show that SAG promotes myelin regeneration and presumably a pro-regenerative phenotype of microglia, thus mimicking the effects previously observed in males. However, unlike in males, the combined molecules failed to cooperate in the demyelinated females, as shown by the level of functional improvement observed. Consistent with this observation, SAG administered in the absence of testosterone amplified peripheral inflammation by presumably activating NK cells and thus counteracting a testosterone-induced reduction in Th17 cells when the molecules were combined. Altogether, the data uncover a sex-dependent effect of the Hedgehog signaling agonist SAG on the peripheral innate immune system that conditions its ability to cooperate or not with androgens in the context of demyelination.


Asunto(s)
Enfermedades Desmielinizantes , Testosterona , Animales , Femenino , Masculino , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/tratamiento farmacológico , Ratones , Testosterona/farmacología , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/agonistas , Ratones Endogámicos C57BL , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Sistema Nervioso Central/metabolismo , Receptor Smoothened/metabolismo , Receptor Smoothened/agonistas , Vaina de Mielina/metabolismo , Modelos Animales de Enfermedad , Transducción de Señal/efectos de los fármacos , Sistema Inmunológico/efectos de los fármacos , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/inmunología , Caracteres Sexuales
20.
Neuroreport ; 35(11): 679-686, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-38874950

RESUMEN

Intracerebral hemorrhage (ICH) is a significant public health matter that has no effective treatment. ICH-induced destruction of the blood-brain barrier (BBB) leads to neurological deterioration. Astrocytic sonic hedgehog (SHH) alleviates brain injury by maintaining the integrity of the BBB after ICH. Silent information regulator 1 (SIRT1) is neuroprotective in several central nervous system diseases via BBB regulation. It is also a possible influential factor of the SHH signaling pathway. Nevertheless, the role of SIRT1 on BBB and the underlying pathological process associated with the SHH signaling pathway after ICH remain unclear. We established an intracerebral hemorrhagic mouse model by collagenase injection. SRT1720 (a selective agonist of SIRT1) was used to evaluate the effect of SIRT1 on BBB integrity after ICH. SIRT1 expression was reduced in the mouse brain after ICH. SRT1720 attenuated neurobehavioral impairments and brain edema of ICH mouse. After ICH induction, SRT1720 improved BBB integrity and tight junction expressions in the mouse brain. The SHH signaling pathway-related factors smoothened and glioma-associated oncogene homolog-1 were increased with the intervention of SRT1720, while cyclopamine (a specific inhibitor of the SHH signaling pathway) reversed these effects. These findings suggest that SIRT1 protects from ICH by altering BBB permeability and tight junction expression levels. This process is associated with the SHH signaling pathway, suggesting that SIRT1 may be a potential therapeutic target for ICH.


Asunto(s)
Barrera Hematoencefálica , Hemorragia Cerebral , Compuestos Heterocíclicos de 4 o más Anillos , Sirtuina 1 , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Sirtuina 1/metabolismo , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/tratamiento farmacológico , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Masculino , Ratones , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/agonistas , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA