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










Base de datos
Intervalo de año de publicación
1.
Elife ; 132024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38591541

RESUMEN

Collective cell migration is fundamental for the development of organisms and in the adult for tissue regeneration and in pathological conditions such as cancer. Migration as a coherent group requires the maintenance of cell-cell interactions, while contact inhibition of locomotion (CIL), a local repulsive force, can propel the group forward. Here we show that the cell-cell interaction molecule, N-cadherin, regulates both adhesion and repulsion processes during Schwann cell (SC) collective migration, which is required for peripheral nerve regeneration. However, distinct from its role in cell-cell adhesion, the repulsion process is independent of N-cadherin trans-homodimerisation and the associated adherens junction complex. Rather, the extracellular domain of N-cadherin is required to present the repulsive Slit2/Slit3 signal at the cell surface. Inhibiting Slit2/Slit3 signalling inhibits CIL and subsequently collective SC migration, resulting in adherent, nonmigratory cell clusters. Moreover, analysis of ex vivo explants from mice following sciatic nerve injury showed that inhibition of Slit2 decreased SC collective migration and increased clustering of SCs within the nerve bridge. These findings provide insight into how opposing signals can mediate collective cell migration and how CIL pathways are promising targets for inhibiting pathological cell migration.


Asunto(s)
Cadherinas , Movimiento Celular , Inhibición de Contacto , Péptidos y Proteínas de Señalización Intercelular , Proteínas de la Membrana , Regeneración Nerviosa , Proteínas del Tejido Nervioso , Células de Schwann , Células de Schwann/metabolismo , Células de Schwann/fisiología , Animales , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Ratones , Cadherinas/metabolismo , Cadherinas/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Regeneración Nerviosa/fisiología , Locomoción/fisiología , Adhesión Celular , Transducción de Señal
2.
J Med Chem ; 64(16): 12109-12131, 2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-34382808

RESUMEN

Aberrant activation of Wnt/ß-catenin signaling is strongly associated with many diseases including cancer invasion and metastasis. Small-molecule targeting of the central signaling node of this pathway, ß-catenin, is a biologically rational approach to abolish hyperactivation of ß-catenin signaling but has been demonstrated to be a difficult task. Herein, we report a drug-like small molecule, ZW4864, that binds with ß-catenin and selectively disrupts the protein-protein interaction (PPI) between B-cell lymphoma 9 (BCL9) and ß-catenin while sparing the ß-catenin/E-cadherin PPI. ZW4864 dose-dependently suppresses ß-catenin signaling activation, downregulates oncogenic ß-catenin target genes, and abrogates invasiveness of ß-catenin-dependent cancer cells. More importantly, ZW4864 shows good pharmacokinetic properties and effectively suppresses ß-catenin target gene expression in the patient-derived xenograft mouse model. This study offers a selective chemical probe to explore ß-catenin-related biology and a drug-like small-molecule ß-catenin/BCL9 disruptor for future drug development.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Piperidinas/uso terapéutico , Unión Proteica/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores , beta Catenina/antagonistas & inhibidores , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Diseño de Fármacos , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones SCID , Estructura Molecular , Piperidinas/síntesis química , Piperidinas/farmacocinética , Relación Estructura-Actividad , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/metabolismo
3.
Mol Cancer Ther ; 19(8): 1623-1635, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32430484

RESUMEN

Although gemcitabine is the cornerstone of care for pancreatic ductal adenocarcinoma (PDA), patients lack durable responses and relapse is inevitable. While the underlying mechanisms leading to gemcitabine resistance are likely to be multifactorial, there is a strong association between activating gemcitabine metabolism pathways and clinical outcome. This study evaluated casein kinase 1 delta (CK1δ) as a potential therapeutic target for PDA and bladder cancer, in which CK1δ is frequently overexpressed. We assessed the antitumor effects of genetically silencing or pharmacologically inhibiting CK1δ using our in-house CK1δ small-molecule inhibitor SR-3029, either alone or in combination with gemcitabine, on the proliferation and survival of pancreatic and bladder cancer cell lines and orthotopic mouse models. Genetic studies confirmed that silencing CK1δ or treatment with SR-3029 induced a significant upregulation of deoxycytidine kinase (dCK), a rate-limiting enzyme in gemcitabine metabolite activation. The combination of SR-3029 with gemcitabine induced synergistic antiproliferative activity and enhanced apoptosis in both pancreatic and bladder cancer cells. Furthermore, in an orthotopic pancreatic tumor model, we observed improved efficacy with combination treatment concomitant with increased dCK expression. This study demonstrates that CK1δ plays a role in gemcitabine metabolism, and that the combination of CK1δ inhibition with gemcitabine holds promise as a future therapeutic option for metastatic PDA as well as other cancers with upregulated CK1δ expression.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Quinasa Idelta de la Caseína/antagonistas & inhibidores , Desoxicitidina Quinasa/metabolismo , Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Antimetabolitos Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Desoxicitidina/farmacología , Desoxicitidina Quinasa/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/enzimología , Neoplasias de la Vejiga Urinaria/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina , Neoplasias Pancreáticas
4.
Cell Death Differ ; 27(4): 1200-1213, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31506606

RESUMEN

Cellular DNA is constantly under threat from internal and external insults, consequently multiple pathways have evolved to maintain chromosomal fidelity. Our previous studies revealed that chronic stress, mediated by continuous stimulation of the ß2-adrenergic-ßarrestin-1 signaling axis suppresses activity of the tumor suppressor p53 and impairs genomic integrity. In this pathway, ßarrestin-1 (ßarr1) acts as a molecular scaffold to promote the binding and degradation of p53 by the E3-ubiquitin ligase, MDM2. We sought to determine whether ßarr1 plays additional roles in the repair of DNA damage. Here we demonstrate that in mice ßarr1 interacts with p53-binding protein 1 (53BP1) with major consequences for the repair of DNA double-strand breaks. 53BP1 is a principle component of the DNA damage response, and when recruited to the site of double-strand breaks in DNA, 53BP1 plays an important role coordinating repair of these toxic lesions. Here, we report that ßarr1 directs 53BP1 degradation by acting as a scaffold for the E3-ubiquitin ligase Rad18. Consequently, knockdown of ßarr1 stabilizes 53BP1 augmenting the number of 53BP1 DNA damage repair foci following exposure to ionizing radiation. Accordingly, ßarr1 loss leads to a marked increase in irradiation resistance both in cells and in vivo. Thus, ßarr1 is an important regulator of double strand break repair, and disruption of the ßarr1/53BP1 interaction offers an attractive strategy to protect cells against high levels of exposure to ionizing radiation.


Asunto(s)
Reparación del ADN , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , beta-Arrestina 1/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Daño del ADN , Reparación del ADN/efectos de la radiación , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Humanos , Intestinos/patología , Ratones Endogámicos C57BL , Unión Proteica/efectos de la radiación , Procesamiento Proteico-Postraduccional/efectos de la radiación , Tolerancia a Radiación/efectos de la radiación , Radiación Ionizante
5.
Cancer Cell ; 36(5): 545-558.e7, 2019 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-31668947

RESUMEN

Epigenetic regulation enables tumors to respond to changing environments during tumor progression and metastases and facilitates treatment resistance. Targeting chromatin modifiers or catalytic effectors of transcription is an emerging anti-cancer strategy. The cyclin-dependent kinases (CDKs) 12 and 13 phosphorylate the C-terminal domain of RNA polymerase II, regulating transcription and co-transcriptional processes. Here we report the development of SR-4835, a highly selective dual inhibitor of CDK12 and CDK13, which disables triple-negative breast cancer (TNBC) cells. Mechanistically, inhibition or loss of CDK12/CDK13 triggers intronic polyadenylation site cleavage that suppresses the expression of core DNA damage response proteins. This provokes a "BRCAness" phenotype that results in deficiencies in DNA damage repair, promoting synergy with DNA-damaging chemotherapy and PARP inhibitors.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proteína Quinasa CDC2/metabolismo , Línea Celular Tumoral , Cisplatino/farmacología , Cisplatino/uso terapéutico , Quinasas Ciclina-Dependientes/metabolismo , Daño del ADN/efectos de los fármacos , Sinergismo Farmacológico , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Recombinación Homóloga/efectos de los fármacos , Humanos , Intrones/efectos de los fármacos , Intrones/genética , Ratones , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Poliadenilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cell Rep ; 25(10): 2755-2765.e5, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30517863

RESUMEN

The formation of myelinating Schwann cells (mSCs) involves the remarkable biogenic process, which rapidly generates the myelin sheath. Once formed, the mSC transitions to a stable homeostatic state, with loss of this stability associated with neuropathies. The histone deacetylases histone deacetylase 1 (HDAC1) and HDAC2 are required for the myelination transcriptional program. Here, we show a distinct role for HDAC3, in that, while dispensable for the formation of mSCs, it is essential for the stability of the myelin sheath once formed-with loss resulting in progressive severe neuropathy in adulthood. This is associated with the prior failure to downregulate the biogenic program upon entering the homeostatic state leading to hypertrophy and hypermyelination of the mSCs, progressing to the development of severe myelination defects. Our results highlight distinct roles of HDAC1/2 and HDAC3 in controlling the differentiation and homeostatic states of a cell with broad implications for the understanding of this important cell-state transition.


Asunto(s)
Histona Desacetilasas/metabolismo , Homeostasis , Vaina de Mielina/metabolismo , Células de Schwann/citología , Células de Schwann/enzimología , Envejecimiento/metabolismo , Animales , Ratones Endogámicos C57BL , Vaina de Mielina/ultraestructura , Ratas , Nervio Ciático/metabolismo , Nervio Ciático/ultraestructura , Transcripción Genética
7.
SLAS Discov ; 23(8): 842-849, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29750582

RESUMEN

Glioblastoma (GBM) is the most aggressive primary brain cancer with an average survival time after diagnosis of only 12-14 months, with few (<5%) long-term survivors. A growing body of work suggests that GBMs contain a small population of glioma stem cells (GSCs) that are thought to be major contributors to treatment resistance and disease relapse. Identifying compounds that modulate GSC proliferation would provide highly valuable molecular probes of GSC-directed signaling. However, targeting GSCs pharmacologically has been challenging. Patient-derived GSCs can be cultured as neurospheres, and in vivo these cells functionally recapitulate the heterogeneity of the original tumor. Using patient-derived GSC-enriched cultures, we have developed a 1536-well spheroid-based proliferation assay and completed a pilot screen, testing ~3300 compounds comprising approved drugs. This cytotoxic and automation-friendly assay yielded a signal-to-background (S/B) ratio of 161.3 ± 7.5 and Z' of 0.77 ± 0.02, demonstrating its robustness. Importantly, compounds were identified with anti-GSC activity, demonstrating the applicability of this assay for large-scale high-throughput screening (HTS).


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Glioma/metabolismo , Glioma/patología , Ensayos Analíticos de Alto Rendimiento , Células Madre Neoplásicas/metabolismo , Animales , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/inmunología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Glioma/tratamiento farmacológico , Glioma/inmunología , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Bibliotecas de Moléculas Pequeñas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Bioorg Med Chem ; 26(3): 590-602, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29289448

RESUMEN

Casein kinase 1δ/ε have been identified as promising therapeutic target for oncology application, including breast and brain cancer. Here, we described our continued efforts in optimization of a lead series of purine scaffold inhibitors that led to identification of two new CK1δ/ε inhibitors 17 and 28 displaying low nanomolar values in antiproliferative assays against the human MDA-MB-231 triple negative breast cancer cell line and have physical, in vitro and in vivo pharmacokinetic properties suitable for use in proof of principle animal xenograft studies against human cancers.


Asunto(s)
Caseína Cinasa 1 épsilon/antagonistas & inhibidores , Quinasa Idelta de la Caseína/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Animales , Sitios de Unión , Caseína Cinasa 1 épsilon/metabolismo , Quinasa Idelta de la Caseína/metabolismo , Dominio Catalítico , Línea Celular Tumoral , Femenino , Semivida , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Simulación del Acoplamiento Molecular , Permeabilidad/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/uso terapéutico , Ratas , Relación Estructura-Actividad , Trasplante Heterólogo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
9.
Bioorg Med Chem Lett ; 28(3): 400-404, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29277458

RESUMEN

The development of a new series of apoptosis signal-regulating kinase 1 (ASK1) inhibitors is described. Starting from purine, pyrimidine and quinazoline scaffolds identified by high throughput screening, we used tools of structure-based drug design to develop a series of potent kinase inhibitors, including 2-arylquinazoline derivatives 12 and 23, with submicromolar inhibitory activities against ASK1. Kinetic analysis demonstrated that the 2-arylquinazoline scaffold ASK1 inhibitors described herein are ATP competitive.


Asunto(s)
Descubrimiento de Drogas , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , MAP Quinasa Quinasa Quinasa 5/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , MAP Quinasa Quinasa 6/antagonistas & inhibidores , MAP Quinasa Quinasa 6/metabolismo , Modelos Moleculares , Estructura Molecular , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Quinazolinas/síntesis química , Quinazolinas/química , Ratas , Relación Estructura-Actividad , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/farmacología
10.
Sci Transl Med ; 7(318): 318ra202, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26676609

RESUMEN

Identification of specific drivers of human cancer is required to instruct the development of targeted therapeutics. We demonstrate that CSNK1D is amplified and/or overexpressed in human breast tumors and that casein kinase 1δ (CK1δ) is a vulnerability of human breast cancer subtypes overexpressing this kinase. Specifically, selective knockdown of CK1δ, or treatment with a highly selective and potent CK1δ inhibitor, triggers apoptosis of CK1δ-expressing breast tumor cells ex vivo, tumor regression in orthotopic models of triple-negative breast cancer, including patient-derived xenografts, and tumor growth inhibition in human epidermal growth factor receptor 2-positive (HER2(+)) breast cancer models. We also show that Wnt/ß-catenin signaling is a hallmark of human tumors overexpressing CK1δ, that disabling CK1δ blocks nuclear accumulation of ß-catenin and T cell factor transcriptional activity, and that constitutively active ß-catenin overrides the effects of inhibition or silencing of CK1δ. Thus, CK1δ inhibition represents a promising strategy for targeted treatment in human breast cancer with Wnt/ß-catenin involvement.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Quinasa Idelta de la Caseína/antagonistas & inhibidores , Terapia Molecular Dirigida , Inhibidores de Proteínas Quinasas/farmacología , Purinas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Quinasa Idelta de la Caseína/genética , Quinasa Idelta de la Caseína/metabolismo , Proliferación Celular/efectos de los fármacos , Biología Computacional , Bases de Datos Genéticas , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Ratones Desnudos , Ratones SCID , Interferencia de ARN , Factores de Transcripción TCF/metabolismo , Factores de Tiempo , Transfección , Neoplasias de la Mama Triple Negativas/enzimología , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Carga Tumoral/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/metabolismo
11.
Mol Pharmacol ; 88(6): 962-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26452771

RESUMEN

Glioblastoma multiforme (GBM) is the most aggressive and common form of adult brain cancer. Current therapeutic strategies include surgical resection, followed by radiotherapy and chemotherapy. Despite such aggressive multimodal therapy, prognosis remains poor, with a median patient survival of 14 months. A proper understanding of the molecular drivers responsible for GBM progression are therefore necessary to instruct the development of novel targeted agents and enable the design of effective treatment strategies. Activation of the c-Jun N-terminal kinase isoform 2 (JNK2) is reported in primary brain cancers, where it associates with the histologic grade and amplification of the epidermal growth factor receptor (EGFR). In this manuscript, we demonstrate an important role for JNK2 in the tumor promoting an invasive capacity of EGFR variant III, a constitutively active mutant form of the receptor commonly found in GBM. Expression of EGFR variant III induces transactivation of JNK2 in GBM cells, which is required for a tumorigenic phenotype in vivo. Furthermore, JNK2 expression and activity is required to promote increased cellular invasion through stimulation of a hepatocyte growth factor-c-Met signaling circuit, whereby secretion of this extracellular ligand activates the receptor tyrosine kinase in both a cell autonomous and nonautonomous manner. Collectively, these findings demonstrate the cooperative and parallel activation of multiple RTKs in GBM and suggest that the development of selective JNK2 inhibitors could be therapeutically beneficial either as single agents or in combination with inhibitors of EGFR and/or c-Met.


Asunto(s)
Receptores ErbB/biosíntesis , Glioblastoma/metabolismo , Factor de Crecimiento de Hepatocito/biosíntesis , Proteína Quinasa 9 Activada por Mitógenos/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Transducción de Señal/fisiología , Animales , Línea Celular Tumoral , Glioblastoma/patología , Humanos , Uniones Intercelulares/metabolismo , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
12.
Cell ; 162(5): 1127-39, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26279190

RESUMEN

The peripheral nervous system has remarkable regenerative capacities in that it can repair a fully cut nerve. This requires Schwann cells to migrate collectively to guide regrowing axons across a 'bridge' of new tissue, which forms to reconnect a severed nerve. Here we show that blood vessels direct the migrating cords of Schwann cells. This multicellular process is initiated by hypoxia, selectively sensed by macrophages within the bridge, which via VEGF-A secretion induce a polarized vasculature that relieves the hypoxia. Schwann cells then use the blood vessels as "tracks" to cross the bridge taking regrowing axons with them. Importantly, disrupting the organization of the newly formed blood vessels in vivo, either by inhibiting the angiogenic signal or by re-orienting them, compromises Schwann cell directionality resulting in defective nerve repair. This study provides important insights into how the choreography of multiple cell-types is required for the regeneration of an adult tissue.


Asunto(s)
Vasos Sanguíneos/metabolismo , Macrófagos/metabolismo , Nervios Periféricos/fisiología , Células de Schwann/metabolismo , Animales , Axones/metabolismo , Hipoxia de la Célula , Células Endoteliales/metabolismo , Inflamación/metabolismo , Masculino , Ratones , Neovascularización Fisiológica , Ratas , Ratas Sprague-Dawley , Regeneración , Factor A de Crecimiento Endotelial Vascular/genética
13.
Blood ; 124(15): 2380-90, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25157181

RESUMEN

Cdk4 and Cdk6 are related protein kinases that bind d-type cyclins and regulate cell-cycle progression. Cdk4/6 inhibitors are currently being used in advanced clinical trials and show great promise against many types of tumors. Cdk4 and Cdk6 are inhibited by INK4 proteins, which exert tumor-suppressing functions. To test the significance of this inhibitory mechanism, we generated knock-in mice that express a Cdk6 mutant (Cdk6 R31C) insensitive to INK4-mediated inhibition. Cdk6(R/R) mice display altered development of the hematopoietic system without enhanced tumor susceptibility, either in the presence or absence of p53. Unexpectedly, Cdk6 R31C impairs the potential of hematopoietic progenitors to repopulate upon adoptive transfer or after 5-fluorouracil-induced damage. The defects are overcome by eliminating sensitivity of cells to INK4 inhibitors by introducing the INK4-insensitive Cdk4 R24C allele, and INK4-resistant mice are more susceptible to hematopoietic and endocrine tumors. In BCR-ABL-transformed hematopoietic cells, Cdk6 R31C causes increased binding of p16(INK4a) to wild-type Cdk4, whereas cells harboring Cdk4 R24C and Cdk6 R31C are fully insensitive to INK4 inhibitors, resulting in accelerated disease onset. Our observations reveal that Cdk4 and Cdk6 cooperate in hematopoietic tumor development and suggest a role for Cdk6 in sequestering INK4 proteins away from Cdk4.


Asunto(s)
Carcinogénesis/metabolismo , Carcinogénesis/patología , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/metabolismo , Alelos , Animales , Muerte Celular , Línea Celular Transformada , Proliferación Celular , Quinasa 6 Dependiente de la Ciclina/genética , Proteínas de Fusión bcr-abl/metabolismo , Ontología de Genes , Hematopoyesis , Células Madre Hematopoyéticas/metabolismo , Ratones , Proteínas Mutantes/metabolismo
14.
PLoS One ; 4(3): e4815, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19283075

RESUMEN

BACKGROUND: The adult endocrine pituitary is known to host several hormone-producing cells regulating major physiological processes during life. Some candidates to progenitor/stem cells have been proposed. However, not much is known about pituitary cell renewal throughout life and its homeostatic regulation during specific physiological changes, such as puberty or pregnancy, or in pathological conditions such as tumor development. PRINCIPAL FINDINGS: We have identified in rodents and humans a niche of non-endocrine cells characterized by the expression of GFRa2, a Ret co-receptor for Neurturin. These cells also express b-Catenin and E-cadherin in an oriented manner suggesting a planar polarity organization for the niche. In addition, cells in the niche uniquely express the pituitary-specific transcription factor Prop1, as well as known progenitor/stem markers such as Sox2, Sox9 and Oct4. Half of these GPS (GFRa2/Prop1/Stem) cells express S-100 whereas surrounding elongated cells in contact with GPS cells express Vimentin. GFRa2+-cells form non-endocrine spheroids in culture. These spheroids can be differentiated to hormone-producing cells or neurons outlining the neuroectoderm potential of these progenitors. In vivo, GPSs cells display slow proliferation after birth, retain BrdU label and show long telomeres in its nuclei, indicating progenitor/stem cell properties in vivo. SIGNIFICANCE: Our results suggest the presence in the adult pituitary of a specific niche of cells characterized by the expression of GFRa2, the pituitary-specific protein Prop1 and stem cell markers. These GPS cells are able to produce different hormone-producing and neuron-like cells and they may therefore contribute to postnatal pituitary homeostasis. Indeed, the relative abundance of GPS numbers is altered in Cdk4-deficient mice, a model of hypopituitarism induced by the lack of this cyclin-dependent kinase. Thus, GPS cells may display functional relevance in the physiological expansion of the pituitary gland throughout life as well as protection from pituitary disease.


Asunto(s)
Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Hipófisis/citología , Nicho de Células Madre/metabolismo , Células Madre/citología , Animales , Bromodesoxiuridina/farmacología , Proliferación Celular , Expresión Génica , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Hipopituitarismo/metabolismo , Ratones , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Hipófisis/metabolismo , Hormonas Hipofisarias/metabolismo , Ratas , Antígenos Embrionarios Específico de Estadio/metabolismo , Células Madre/metabolismo , Telómero/ultraestructura , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
J Mol Endocrinol ; 42(2): 75-86, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18987159

RESUMEN

The pituitary gland regulates diverse physiological functions, including growth, metabolism, reproduction, stress response, and ageing. Early genetic models in the mouse taught us that the pituitary is highly sensitive to genetic alteration of specific cell cycle regulators such as the retinoblastoma protein (pRB) or the cell cycle inhibitor p27(Kip1). The molecular analysis of human pituitary neoplasias has now corroborated that cell cycle deregulation is significantly implicated in pituitary tumorigenesis. In particular, proteins involved in cyclin-dependent kinase regulation or the pRB pathway are altered in nearly all human pituitary tumors. Additional cell cycle regulators such as PTTG1/securin may have critical roles in promoting genomic instability in pituitary neoplasias. Recent experimental data suggest that these cell cycle regulators may have significant implications in the biology of putative progenitor cells and pituitary homeostasis. Understanding how cell cycle regulation controls pituitary biology may provide us with new therapeutic approaches against pituitary diseases.


Asunto(s)
Ciclo Celular , Enfermedades de la Hipófisis/patología , Hipófisis/citología , Hipófisis/embriología , Animales , Quinasas Ciclina-Dependientes/metabolismo , Humanos , Enfermedades de la Hipófisis/enzimología , Hipófisis/enzimología , Neoplasias Hipofisarias/enzimología , Neoplasias Hipofisarias/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...