Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Ann N Y Acad Sci ; 1415(1): 21-33, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29500936

RESUMEN

Activated lymphocytes promote inflammation and bone destruction in rheumatoid arthritis (RA), making T cells and B cells therapeutic targets. Indeed, pharmacological blockade of CD28 costimulation using CTLA-4Ig (abatacept), approved for amelioration of RA, renders T cells dormant (anergic). CTLA-4Ig also promotes bone accretion in healthy mice; surprisingly, however, this effect is driven exclusively through upregulation of bone formation, rather than anti-inflammatory effects on resorption. In the study presented here, we utilized T cell receptor ß gene and Wnt-10b gene knockout mice to investigate the roles of T cells and Wnt-10b in CTLA-4Ig-induced bone anabolism. Ablation of either T cells or Wnt-10b not only abolished CTLA-4Ig-induced bone anabolism but also, paradoxically, suppressed bone formation leading to bone loss. Stalled bone formation was accompanied by bone marrow stromal cell expression of the Wnt pathway inhibitor sclerostin. Our data suggest that an immunoskeletal pivot may promote or suppress bone formation, depending on the net outcome of CTLA-4Ig action directed independently on T cells and osteoblast-linage cells that counter Wnt-10b-induced bone anabolism, by secretion of sclerostin. While CTLA-4Ig action is tipped in favor of bone formation under physiological conditions, pathological immunodeficiency may lead to suppressed bone formation and skeletal damage.


Asunto(s)
Abatacept/farmacología , Anabolizantes/farmacología , Huesos/efectos de los fármacos , Glicoproteínas/metabolismo , Linfocitos T/efectos de los fármacos , Proteínas Wnt/metabolismo , Células 3T3 , Proteínas Adaptadoras Transductoras de Señales , Animales , Antirreumáticos/farmacología , Densidad Ósea/efectos de los fármacos , Huesos/diagnóstico por imagen , Huesos/metabolismo , Antígenos CD28/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Proteínas Wnt/deficiencia , Proteínas Wnt/genética , Microtomografía por Rayos X
2.
Nat Commun ; 8(1): 1620, 2017 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-29158473

RESUMEN

Endothelial cells transduce mechanical forces from blood flow into intracellular signals required for vascular homeostasis. Here we show that endothelial NOTCH1 is responsive to shear stress, and is necessary for the maintenance of junctional integrity, cell elongation, and suppression of proliferation, phenotypes induced by laminar shear stress. NOTCH1 receptor localizes downstream of flow and canonical NOTCH signaling scales with the magnitude of fluid shear stress. Reduction of NOTCH1 destabilizes cellular junctions and triggers endothelial proliferation. NOTCH1 suppression results in changes in expression of genes involved in the regulation of intracellular calcium and proliferation, and preventing the increase of calcium signaling rescues the cell-cell junctional defects. Furthermore, loss of Notch1 in adult endothelium increases hypercholesterolemia-induced atherosclerosis in the descending aorta. We propose that NOTCH1 is atheroprotective and acts as a mechanosensor in adult arteries, where it integrates responses to laminar shear stress and regulates junctional integrity through modulation of calcium signaling.


Asunto(s)
Arterias/metabolismo , Mecanotransducción Celular , Receptor Notch1/metabolismo , Animales , Arterias/química , Calcio/metabolismo , Células Endoteliales/química , Células Endoteliales/metabolismo , Endotelio Vascular/química , Endotelio Vascular/metabolismo , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Notch1/genética , Estrés Mecánico
3.
Cancer Res ; 75(22): 4960-72, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26400062

RESUMEN

Breast cancer is associated with alterations in a number of growth factor and hormone-regulated signaling pathways. Mouse models of metastatic breast cancer typically feature mutated oncoproteins that activate PI3K, Stat3, and Ras signaling, but the individual and combined roles of these pathways in breast cancer progression are poorly understood. In this study, we examined the relationship between oncogenic pathway activation and breast cancer subtype by analyzing mouse mammary tumor formation in which each pathway was activated singly or pairwise. All three oncogenes showed cooperation during primary tumor formation, but efficient dissemination was only dependent on Ras. In addition, transcriptional profiling demonstrated that Ras induced adenocarcinomas with molecular characteristics related to human basal-like and HER2(+) tumors. In contrast, Ras combined with PIK3CA(H1047R), an oncogenic mutant linked to ERα(+)/luminal breast cancer in humans, induced metastatic luminal B-like tumors. Consistent with these data, elevated Ras signaling was associated with basal-like and HER2(+) subtype tumors in humans and showed a statistically significant negative association with estrogen receptor (ER) signaling across all breast cancer. Despite this, there are luminal tumors with elevated Ras signaling. Importantly, when considered as a continuous variable, Ras pathway activation was strongly linked to reduced survival of patients with ERα(+) disease independent of PI3K or Stat3 activation. Therefore, our studies suggest that Ras activation is a key determinant for dissemination and poor prognosis of ERα(+)/luminal breast cancer in humans, and hormone therapy supplemented with Ras-targeting agents may be beneficial for treating this aggressive subtype.


Asunto(s)
Neoplasias de la Mama/patología , Invasividad Neoplásica/patología , Transducción de Señal/fisiología , Proteínas ras/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Supervivencia sin Enfermedad , Femenino , Inmunohistoquímica , Estimación de Kaplan-Meier , Ratones , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Análisis de Secuencia por Matrices de Oligonucleótidos
4.
PLoS One ; 9(11): e111606, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25369058

RESUMEN

We recently reported that the PPM1l gene encodes an endoplasmic reticulum (ER) membrane targeted protein phosphatase (named PP2Ce) with highly specific activity towards Inositol-requiring protein-1 (IRE1) and regulates the functional outcome of ER stress. In the present report, we found that the PP2Ce protein is highly expressed in lactating epithelium of the mammary gland. Loss of PP2Ce in vivo impairs physiological unfolded protein response (UPR) and induces stress kinase activation, resulting in loss of milk production and induction of epithelial apoptosis in the lactating mammary gland. This study provides the first in vivo evidence that PP2Ce is an essential regulator of normal lactation, possibly involving IRE1 signaling and ER stress regulation in mammary epithelium.


Asunto(s)
Estrés del Retículo Endoplásmico , Lactancia , Glándulas Mamarias Animales/fisiología , Proteínas de la Membrana/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Apoptosis , Femenino , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/ultraestructura , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas Fosfatasas/análisis , Fosfoproteínas Fosfatasas/genética , Periodo Posparto , Proteína Fosfatasa 2C , Transducción de Señal , Respuesta de Proteína Desplegada
5.
PLoS One ; 9(2): e89464, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586797

RESUMEN

Epigenetic gene silencing by histone modifications and DNA methylation is essential for cancer development. The molecular mechanism that promotes selective epigenetic changes during tumorigenesis is not understood. We report here that the PIAS1 SUMO ligase is involved in the progression of breast tumorigenesis. Elevated PIAS1 expression was observed in breast tumor samples. PIAS1 knockdown in breast cancer cells reduced the subpopulation of tumor-initiating cells, and inhibited breast tumor growth in vivo. PIAS1 acts by delineating histone modifications and DNA methylation to silence the expression of a subset of clinically relevant genes, including breast cancer DNA methylation signature genes such as cyclin D2 and estrogen receptor, and breast tumor suppressor WNT5A. Our studies identify a novel epigenetic mechanism that regulates breast tumorigenesis through selective gene silencing.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Mama/patología , Carcinogénesis/genética , Epigénesis Genética/genética , Proteínas Inhibidoras de STAT Activados/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Animales , Línea Celular Tumoral , Ciclina D2/genética , Metilación de ADN/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Silenciador del Gen , Humanos , Ratones SCID , Proteínas Proto-Oncogénicas/genética , Receptores de Estrógenos/genética , Ubiquitina-Proteína Ligasas/genética , Proteínas Wnt/genética , Proteína Wnt-5a
6.
Stem Cells ; 32(6): 1503-14, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24677652

RESUMEN

Unlimited self renewal capacity and differentiation potential make human pluripotent stem cells (PSC) a promising source for the ex vivo manufacture of red blood cells (RBCs) for safe transfusion. Current methods to induce erythropoiesis from PSC suffer from low yields of RBCs, most of which are immature and contain embryonic and fetal rather than adult hemoglobins. We have previously shown that homodimerization of the intracellular component of MPL (ic-MPL) induces erythropoiesis from human cord blood progenitors. The goal of this study was to investigate the potential of ic-MPL dimerization to induce erythropoiesis from human embryonic stem cells (hESCs) and to identify the signaling pathways activated by this strategy. We present here the evidence that ic-MPL dimerization induces erythropoietin (EPO)-independent erythroid differentiation from hESC by inducing the generation of erythroid progenitors and by promoting more efficient erythroid maturation with increased RBC enucleation as well as increased gamma:epsilon globin ratio and production of beta-globin protein. ic-MPL dimerization is significantly more potent than EPO in inducing erythropoiesis, and its effect is additive to EPO. Signaling studies show that dimerization of ic-MPL, unlike stimulation of the wild type MPL receptor, activates AKT in the absence of JAK2/STAT5 signaling. AKT activation upregulates GATA-1 and FOXO3 transcriptional pathways with resulting inhibition of apoptosis, modulation of cell cycle, and enhanced maturation of erythroid cells. These findings open up potential new targets for the generation of therapeutically relevant RBC products from hPSC.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/enzimología , Eritropoyesis , Eritropoyetina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Ciclo Celular , Diferenciación Celular , Línea Celular , Proliferación Celular , Supervivencia Celular , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Humanos , Megacariocitos/citología , Megacariocitos/metabolismo , Multimerización de Proteína , Estructura Terciaria de Proteína , Receptores de Trombopoyetina/química , Receptores de Trombopoyetina/metabolismo
7.
Mol Metab ; 2(4): 405-16, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24327956

RESUMEN

The protein phosphatase 1-like gene (PPM1l) was identified as causal gene for obesity and metabolic abnormalities in mice. However, the underlying mechanisms were unknown. In this report, we find PPM1l encodes an endoplasmic reticulum (ER) membrane targeted protein phosphatase (PP2Ce) and has specific activity to basal and ER stress induced auto-phosphorylation of Inositol-REquiring protein-1 (IRE1). PP2Ce inactivation resulted in elevated IRE1 phosphorylation and higher expression of XBP-1, CHOP, and BiP at basal. However, ER stress stimulated XBP-1 and BiP induction was blunted while CHOP induction was further enhanced in PP2Ce null cells. PP2Ce protein levels are significantly induced during adipogenesis in vitro and are necessary for normal adipocyte maturation. Finally, we provide evidence that common genetic variation of PPM11 gene is significantly associated with human lipid profile. Therefore, PPM1l mediated IRE1 regulation and downstream ER stress signaling is a plausible molecular basis for its role in metabolic regulation and disorder.

8.
Blood ; 122(14): 2346-57, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-23954891

RESUMEN

Estrogen deficiency expands hemopoietic stem and progenitor cells (HSPCs) and mature blood lineages, but the involved mechanism and the affected HSPC populations are mostly unknown. Here we show that ovariectomy (ovx) expands short-term HSPCs (ST-HSPCs) and improves blood cell engraftment and host survival after bone marrow (BM) transplantation through a dual role of the T-cell costimulatory molecule CD40 ligand (CD40L). This surface receptor is required for ovx to stimulate T-cell production of Wnt10b, a Wnt ligand that activates Wnt signaling in HSPCs and stromal cells (SCs). Moreover, CD40L is required for ovx to increase SC production of the hemopoietic cytokines interleukin (IL)-6, IL-7, and granulocyte macrophage-colony-stimulating factor. Attesting to the relevance of CD40L and Wnt10b, ovx fails to expand ST-HSPCs in CD40L-null mice and in animals lacking global or T-cell expression of Wnt10b. In summary, T cells expressed CD40L, and the resulting increased production of Wnt10b and hemopoietic cytokines by T cells and SCs, respectively, plays a pivotal role in the mechanism by which ovx regulates hemopoiesis. The data suggest that antiestrogens may represent pharmacological targets to improve ST-HSPC function through activation of the microenvironment.


Asunto(s)
Ligando de CD40/biosíntesis , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Ovariectomía , Linfocitos T/metabolismo , Proteínas Wnt/biosíntesis , Animales , Trasplante de Médula Ósea/inmunología , Femenino , Citometría de Flujo , Células Madre Hematopoyéticas/inmunología , Ratones , Ratones Noqueados , Linfocitos T/inmunología , Proteínas Wnt/inmunología
9.
Blood ; 122(1): 143-53, 2013 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-23690447

RESUMEN

Host responses to chemotherapy can induce resistance mechanisms that facilitate tumor regrowth. To determine the contribution of bone marrow-derived cells (BMDCs), we exposed tumor-bearing mice to chemotherapeutic agents and evaluated the influx and contribution of a genetically traceable subpopulation of BMDCs (vascular endothelial-cadherin-Cre-enhanced yellow fluorescent protein [VE-Cad-Cre-EYFP]). Treatment of tumor-bearing mice with different chemotherapeutics resulted in a three- to 10-fold increase in the influx of VE-Cad-Cre-EYFP. This enhanced influx was accompanied by a significant increase in angiogenesis. Expression profile analysis revealed a progressive change in the EYFP population with loss of endothelial markers and an increase in mononuclear markers. In the tumor, 2 specific populations of VE-Cad-Cre-EYFP BMDCs were identified: Gr1⁺/CD11b⁺ and Tie2high/platelet endothelial cell adhesion moleculelow cells, both located in perivascular areas. A common signature of the EYFP population that exits the bone marrow is an increase in Notch. Inducible inactivation of Notch in the EYFP⁺ BMDCs impaired homing of these BMDCs to the tumor. Importantly, Notch deletion reduced therapy-enhanced angiogenesis, and was associated with an increased antitumor effect of the chemotherapy. These findings revealed the functional significance of a specific population of supportive BMDCs in response to chemotherapeutics and uncovered a new potential strategy to enhance anticancer therapy.


Asunto(s)
Células de la Médula Ósea/fisiología , Carcinoma Pulmonar de Lewis/tratamiento farmacológico , Cisplatino/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Paclitaxel/farmacología , Receptor Notch1/fisiología , Animales , Antígenos CD/metabolismo , Antineoplásicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Células de la Médula Ósea/citología , Cadherinas/metabolismo , Carcinoma Pulmonar de Lewis/genética , Neoplasias Colorrectales/genética , Resistencia a Antineoplásicos/fisiología , Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Receptor Notch1/genética , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Blood ; 120(22): 4352-62, 2012 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-22955916

RESUMEN

Intermittent parathyroid hormone (iPTH) treatment expands hemopoietic stem and progenitor cells (HSPCs), but the involved mechanisms and the affected HSPC populations are mostly unknown. Here we show that T cells are required for iPTH to expand short-term HSPCs (ST-HSPCs) and improve blood cell engraftment and host survival after BM transplantation. Silencing of PTH/PTH-related protein receptor (PPR) in T cells abrogates the effects of iPTH, thus demonstrating a requirement for direct PPR signaling in T cells. Mechanistically, iPTH expands ST-HSPCs by activating Wnt signaling in HSPCs and stromal cells (SCs) through T-cell production of the Wnt ligand Wnt10b. Attesting to the relevance of Wnt10b, iPTH fails to expand ST-HSPCs in mice with Wnt10b(-/-) T cells. Moreover, iPTH fails to promote engraftment and survival after BM transplantation in Wnt10b null mice. In summary, direct PPR signaling in T cells and the resulting production of Wnt10b play a pivotal role in the mechanism by which iPTH expands ST-HSPCs. The data suggest that T cells may provide pharmacologic targets for HSPC expansion.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Hormona Paratiroidea/farmacología , Linfocitos T/fisiología , Animales , Trasplante de Médula Ósea , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas , Femenino , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/efectos de los fármacos , Factores de Tiempo , Proteínas Wnt/genética
11.
Cancer Res ; 71(7): 2706-17, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21324922

RESUMEN

PIK3CA, which codes for the p110α catalytic subunit of phosphatidylinositol 3-kinase, is one of the most frequently mutated genes in human breast cancer. Here, we describe a mouse model for PIK3CA-induced breast cancer by using the ROSA26 (R26) knock-in system, in which targeted Pik3ca alleles can be activated through transgenic expression of Cre recombinase. We mated Pik3ca(H1047R) and Pik3ca(wt) knock-in lines with MMTV-Cre transgenics, which express Cre in mammary epithelium. Starting at approximately 5 months of age, female R26-Pik3ca(H1047R);MMTV-Cre mice, but not control R26-Pik3ca(wt);MMTV-Cre mice, developed mammary tumors, as well as lymphoid and skin malignancies. R26-Pik3ca(H1047R);MMTV-Cre mammary tumors were typically either adenosquamous carcinoma or adenomyoepithelioma. As p53 is the most commonly mutated gene in breast cancer, we tested for genetic interaction between Pik3ca(H1047R) and p53 loss-of-function mutations in R26-Pik3ca(H1047R);p53(loxP/+);MMTV-Cre mice. This led to decreased survival of double-mutant animals, which developed lymphoma and mammary tumors with rapid kinetics. Mammary tumors that formed in p53(loxP/+);MMTV-Cre conditional mutants were either poorly differentiated adenocarcinoma or spindle cell/EMT, whereas R26-Pik3ca(H1047R);p53(loxP/+);MMTV-Cre mammary tumors were mostly adenosquamous carcinoma or spindle cell/EMT indicating that double-mutant mice develop a distinct spectrum of mammary tumors. Thus, an oncogenic variant of PIK3CA implicated in multiple human breast cancer subtypes can induce a very diverse spectrum of mammary tumors in mice. Furthermore, Pik3ca(H1047R) shows cooperation with p53, which altered the specific tumors that formed. Thus, the two most frequently mutated genes in human breast cancer show cooperation in mammary tumor formation.


Asunto(s)
Neoplasias Mamarias Experimentales/genética , Mutación , Fosfatidilinositol 3-Quinasas/genética , Proteína p53 Supresora de Tumor/genética , Alelos , Animales , Fosfatidilinositol 3-Quinasa Clase I , Femenino , Técnicas de Sustitución del Gen , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/biosíntesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/deficiencia
12.
Development ; 138(4): 767-76, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21266412

RESUMEN

Reelin signaling is required for appropriate cell migration and ductal patterning during mammary gland morphogenesis. Dab1, an intracellular adaptor protein activated in response to reelin signaling, is expressed in the developing mammary bud and in luminal epithelial cells in the adult gland. Reelin protein is expressed in a complementary pattern, first in the epithelium overlying the mammary bud during embryogenesis and then in the myoepithelium and periductal stroma in the adult. Deletion in mouse of either reelin or Dab1 induced alterations in the development of the ductal network, including significant retardation in ductal elongation, decreased terminal branching, and thickening and disorganization of the luminal wall. At later stages, some mutant glands overcame these early delays, but went on to exhibit enlarged and chaotic ductal morphologies and decreased terminal branching: these phenotypes are suggestive of a role for reelin in spatial patterning or structural organization of the mammary epithelium. Isolated mammary epithelial cells exhibited decreased migration in response to exogenous reelin in vitro, a response that required Dab1. These observations highlight a role for reelin signaling in the directed migration of mammary epithelial cells driving ductal elongation into the mammary fat pad and provide the first evidence that reelin signaling may be crucial for regulating the migration and organization of non-neural tissues.


Asunto(s)
Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Proliferación Celular , Células Cultivadas , Proteínas de la Matriz Extracelular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/embriología , Ratones , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/genética , Proteína Reelina , Serina Endopeptidasas/metabolismo
13.
J Nucleic Acids ; 20102010 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-20936109

RESUMEN

Following genotoxic stress, the histone H2AX becomes phosphorylated at serine 139 by the ATM/ATR family of kinases. The tumor suppressor BRCA1, also phosphorylated by ATM/ATR kinases, is one of several proteins that colocalize with phospho-H2AX (γ-H2AX) at sites of active DNA repair. Both the precise mechanism and the purpose of BRCA1 recruitment to sites of DNA damage are unknown. Here we show that BRCA1 and γ-H2AX form an acid-stable biochemical complex on chromatin after DNA damage. Maximal association of BRCA1 with γ-H2AX correlates with reduced global γ-H2AX levels on chromatin late in the repair process. Since BRCA1 is known to have E3 ubiquitin ligase activity in vitro, we examined H2AX for evidence of ubiquitination. We found that H2AX is ubiquitinated at lysines 119 and 119 in vivo and that blockage of 26S proteasome function stabilizes γ-H2AX levels within cells. When BRCA1 levels were reduced, ubiquitination of H2AX was also reduced, and the cells retained higher levels of phosphorylated H2AX. These results indicate that BRCA1 is recruited into stable complexes with γ-H2AX and that the complex is involved in attenuation of the γ-H2AX repair signal after DNA damage.

14.
J Clin Invest ; 120(9): 3296-309, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20679727

RESUMEN

Breast cancer is a highly heterogeneous disease, with several different subtypes being characterized by distinct histology, gene expression patterns, and genetic alterations. The tumor suppressor gene retinoblastoma 1 (RB1) is frequently lost in both luminal-B and triple-negative tumor (TNT; i.e., estrogen receptor-, progesterone receptor-, and human epidermal growth factor receptor 2-negative) breast cancer subtypes. However, a causal role for RB1 loss in different subtypes remains undefined. Here we report that deletion of Rb alone or together with its relative p107 in mouse mammary stem/bipotent progenitor cells induced focal acinar hyperplasia with squamous metaplasia. These lesions progressed into histologically diverse, transplantable mammary tumors with features of either luminal-B or TNT subtypes. The TNTs included basal-like tumors as well as tumors that exhibited epithelial-to-mesenchymal transition (EMT). The EMT-type tumors and a subset of the basal-like tumors, but not luminal-B-like tumors, expressed mutant forms of the tumor suppressor p53. Accordingly, targeted deletion of both Rb and p53 in stem/bipotent progenitors led to histologically uniform, aggressive, EMT-type tumors. Reintroduction of Rb into these tumor cells suppressed growth in vitro and tumor formation in vivo. These results establish a causal role for Rb loss in breast cancer in mice and demonstrate that cooperating oncogenic events, such as mutations in p53, dictate tumor subtype after Rb inactivation.


Asunto(s)
Eliminación de Gen , Genes de Retinoblastoma , Genes p53 , Neoplasias Mamarias Experimentales/genética , Células Madre/metabolismo , Proteína p53 Supresora de Tumor/genética , Animales , Línea Celular , Femenino , Humanos , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Noqueados
15.
J Bone Miner Res ; 25(10): 2138-47, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20499361

RESUMEN

Wnt10b is a canonical Wnt ligand expressed in developing bone and has been linked to mesenchymal progenitor functions in mice and humans. Because Wnt signaling has been shown to play an important role in progenitor maintenance in a variety of adult tissues, we examined bone deposition and growth rates throughout postnatal development in Wnt10b-null mice. Using bone histomorphometry and micro-computed tomographic (µCT) studies, we demonstrate that trabecular bone deposition is slightly enhanced in Wnt10b-null mice at 1 month of age, followed by progressive loss with age. Importantly, we find that Wnt10b is required for maintenance of adult bone density in multiple backgrounds of inbred mice and that both copies of the Wnt10b gene are required to maintain normal bone density in 6-month-old animals. We go on to show that the loss in trabecular bone in Wnt10b-null mice is associated with a reduction in the number of bone marrow-derived mesenchymal progenitors (MPCs) using in vitro colony-forming unit assays and marker analysis. Analysis of osteogenic gene expression in primary bone marrow stromal cells demonstrated reductions in expression of several osteoblast differentiation markers. Taken together, our results indicate that Wnt10b is uniquely required for maintenance of mesenchymal progenitor activity in adult bone. The results show the significance of studying individual Wnt ligands and their potentially unique contribution in the context of aging and disease.


Asunto(s)
Células Madre Mesenquimatosas/citología , Osteoporosis/genética , Proteínas Wnt/fisiología , Factores de Edad , Animales , Diferenciación Celular , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tomografía Computarizada por Rayos X , Proteínas Wnt/genética
16.
Dev Cell ; 18(1): 39-51, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20152176

RESUMEN

Maintenance of single-layered endothelium, squamous endothelial cell shape, and formation of a patent vascular lumen all require defined endothelial cell polarity. Loss of beta1 integrin (Itgb1) in nascent endothelium leads to disruption of arterial endothelial cell polarity and lumen formation. The loss of polarity is manifested as cuboidal-shaped endothelial cells with dysregulated levels and mislocalization of normally polarized cell-cell adhesion molecules, as well as decreased expression of the polarity gene Par3 (pard3). beta1 integrin and Par3 are both localized to the endothelial layer, with preferential expression of Par3 in arterial endothelium. Luminal occlusion is also exclusively noted in arteries, and is partially rescued by replacement of Par3 protein in beta1-deficient vessels. Combined, our findings demonstrate that beta1 integrin functions upstream of Par3 as part of a molecular cascade required for endothelial cell polarity and lumen formation.


Asunto(s)
Arteriolas/embriología , Arteriolas/metabolismo , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/metabolismo , Integrina beta1/metabolismo , Neovascularización Fisiológica/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Arteriolas/citología , Adhesión Celular/fisiología , Moléculas de Adhesión Celular/genética , Proteínas de Ciclo Celular , Diferenciación Celular/fisiología , Polaridad Celular/fisiología , Forma de la Célula/fisiología , Modelos Animales de Enfermedad , Células Endoteliales/citología , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones , Ratones Noqueados , Transducción de Señal/fisiología
18.
Cell Metab ; 10(3): 229-40, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19723499

RESUMEN

Intermittent administration of parathyroid hormone (iPTH) is used to treat osteoporosis because it improves bone architecture and strength, but the underlying cellular and molecular mechanisms are unclear. Here, we show that iPTH increases the production of Wnt10b by bone marrow CD8+ T cells and induces these lymphocytes to activate canonical Wnt signaling in preosteoblasts. Accordingly, in responses to iPTH, T cell null mice display diminished Wnt signaling in preosteoblasts and blunted osteoblastic commitment, proliferation, differentiation, and life span, which result in decreased trabecular bone anabolism and no increase in strength. Demonstrating the specific role of lymphocytic Wnt10b, iPTH has no anabolic activity in mice lacking T-cell-produced Wnt10b. Therefore, T-cell-mediated activation of Wnt signaling in osteoblastic cells plays a key permissive role in the mechanism by which iPTH increases bone strength, suggesting that T cell osteoblast crosstalk pathways may provide pharmacological targets for bone anabolism.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Hormona Paratiroidea/farmacología , Proteínas Wnt/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Proliferación Celular , Ratones , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/deficiencia , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Transducción de Señal
19.
J Neurooncol ; 94(1): 1-19, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19468690

RESUMEN

Glioblastoma multiforme (GBM) remains refractory to conventional therapy. CD133+ GBM cells have been recently isolated and characterized as chemo-/radio-resistant tumor-initiating cells and are hypothesized to be responsible for post-treatment recurrence. In order to explore the molecular properties of tumorigenic CD133+ GBM cells that resist treatment, we isolated CD133+ GBM cells from tumors that are recurrent and have previously received chemo-/radio-therapy. We found that the purified CD133+ GBM cells sorted from the CD133+ GBM spheres express SOX2 and CD44 and are capable of clonal self-renewal and dividing to produce fast-growing CD133- progeny, which form the major cell population within GBM spheres. Intracranial injection of purified CD133+, not CD133- GBM daughter cells, can lead to the development of YKL-40+ infiltrating tumors that display hypervascularity and pseudopalisading necrosis-like features in mouse brain. The molecular profile of purified CD133+ GBM cells revealed characteristics of neuroectoderm-like cells, expressing both radial glial and neural crest cell developmental genes, and portraying a slow-growing, non-differentiated, polarized/migratory, astrogliogenic, and chondrogenic phenotype. These data suggest that at least a subset of treated and recurrent GBM tumors may be seeded by CD133+ GBM cells with neural and mesenchymal properties. The data also imply that CD133+ GBM cells may be clinically indolent/quiescent prior to undergoing proliferative cell division (PCD) to produce CD133- GBM effector progeny. Identifying intrinsic and extrinsic cues, which promote CD133+ GBM cell self-renewal and PCD to support ongoing tumor regeneration may highlight novel therapeutic strategies to greatly diminish the recurrence rate of GBM.


Asunto(s)
Antígenos CD/genética , Neoplasias Encefálicas/patología , Regulación Neoplásica de la Expresión Génica/fisiología , Glioblastoma/patología , Glicoproteínas/genética , Células Madre Neoplásicas/fisiología , Péptidos/genética , Antígeno AC133 , Animales , Biomarcadores de Tumor/genética , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Femenino , Citometría de Flujo/métodos , Perfilación de la Expresión Génica/métodos , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones SCID , Trasplante de Neoplasias , Células Madre Neoplásicas/citología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Esferoides Celulares/patología , Factores de Tiempo , Células Tumorales Cultivadas
20.
J Gene Med ; 11(8): 655-63, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19455593

RESUMEN

BACKGROUND: Targeted gene transduction in vivo is the ultimate preferred method for gene delivery. We previously developed targeting lentiviral vectors that specifically recognize cell surface molecules with conjugated antibodies and mediate targeted gene transduction both in vitro and in vivo. Although effective in some experimental settings, the conjugation of virus with antibodies is mediated by the interaction between protein A and the Fc region of antibodies, which is not as stable as covalent conjugation. We have now developed a more stable conjugation strategy utilizing the interaction between avidin and biotin. METHODS: We inserted the biotin-adaptor-peptide, which was biotinylated by secretory biotin ligase at specific sites, into our targeting envelope proteins, enabling conjugation of the pseudotyped virus with avidin, streptavidin or neutravidin. RESULTS: When conjugated with avidin-antibody fusion proteins or the complex of avidin and biotinylated targeting molecules, the vectors could mediate specific transduction to targeted cells recognized by the targeting molecules. When conjugated with streptavidin-coated magnetic beads, transduction by the vectors was targeted to the locations of magnets. CONCLUSIONS: This targeting vector system can be used for broad applications of targeted gene transduction using biotinylated targeting molecules or targeting molecules fused with avidin.


Asunto(s)
Biotina/metabolismo , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Lentivirus/genética , Péptidos/metabolismo , Avidina/metabolismo , Biotinilación , Western Blotting , Línea Celular , Electroforesis en Gel de Poliacrilamida , Humanos , Magnetismo , Mutagénesis Insercional , Receptores de Transferrina/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Virus Sindbis/genética , Transducción Genética , Proteínas del Envoltorio Viral/metabolismo , Virión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...