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

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 122(14): 2346-57, 2013 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-23954891

RESUMO

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.


Assuntos
Ligante de CD40/biossíntese , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Ovariectomia , Linfócitos T/metabolismo , Proteínas Wnt/biossíntese , Animais , Transplante de Medula Óssea/imunologia , Feminino , Citometria de Fluxo , Células-Tronco Hematopoéticas/imunologia , Camundongos , Camundongos Knockout , Linfócitos T/imunologia , Proteínas Wnt/imunologia
2.
Blood ; 122(1): 143-53, 2013 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-23690447

RESUMO

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.


Assuntos
Células da Medula Óssea/fisiologia , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Cisplatino/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Paclitaxel/farmacologia , Receptor Notch1/fisiologia , Animais , Antígenos CD/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Células da Medula Óssea/citologia , Caderinas/metabolismo , Carcinoma Pulmonar de Lewis/genética , Neoplasias Colorretais/genética , Resistencia a Medicamentos Antineoplásicos/fisiologia , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Receptor Notch1/genética , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Stem Cells ; 32(6): 1503-14, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24677652

RESUMO

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.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/enzimologia , Eritropoese , Eritropoetina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Ciclo Celular , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Humanos , Megacariócitos/citologia , Megacariócitos/metabolismo , Multimerização Proteica , Estrutura Terciária de Proteína , Receptores de Trombopoetina/química , Receptores de Trombopoetina/metabolismo
4.
Development ; 138(4): 767-76, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21266412

RESUMO

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.


Assuntos
Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais , Animais , Moléculas de Adesão Celular Neuronais/metabolismo , Proliferação de Células , Células Cultivadas , Proteínas da Matriz Extracelular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/embriologia , Camundongos , Camundongos Transgênicos , Mutação , Proteínas do Tecido Nervoso/genética , Proteína Reelina , Serina Endopeptidases/metabolismo
5.
Blood ; 120(22): 4352-62, 2012 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-22955916

RESUMO

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.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Linfócitos T/fisiologia , Animais , Transplante de Medula Óssea , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Células Cultivadas , Feminino , Genes Codificadores da Cadeia beta de Receptores de Linfócitos T/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T/efeitos dos fármacos , Fatores de Tempo , Proteínas Wnt/genética
6.
EMBO J ; 27(3): 535-45, 2008 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-18219273

RESUMO

Estrogen deficiency in menopause is a major cause of osteoporosis in women. Estrogen acts to maintain the appropriate ratio between bone-forming osteoblasts and bone-resorbing osteoclasts in part through the induction of osteoclast apoptosis. Recent studies have suggested a role for Fas ligand (FasL) in estrogen-induced osteoclast apoptosis by an autocrine mechanism involving osteoclasts alone. In contrast, we describe a paracrine mechanism in which estrogen affects osteoclast survival through the upregulation of FasL in osteoblasts (and not osteoclasts) leading to the apoptosis of pre-osteoclasts. We have characterized a cell-type-specific hormone-inducible enhancer located 86 kb downstream of the FasL gene as the target of estrogen receptor-alpha induction of FasL expression in osteoblasts. In addition, tamoxifen and raloxifene, two selective estrogen receptor modulators that have protective effects in bone, induce apoptosis in pre-osteoclasts by the same osteoblast-dependent mechanism. These results demonstrate that estrogen protects bone by inducing a paracrine signal originating in osteoblasts leading to the death of pre-osteoclasts and offer an important new target for the prevention and treatment of osteoporosis.


Assuntos
Osso e Ossos/metabolismo , Estrogênios/fisiologia , Proteína Ligante Fas/biossíntese , Proteína Ligante Fas/fisiologia , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Células 3T3 , Animais , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/fisiologia , Técnicas de Cocultura , Proteína Ligante Fas/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/citologia , Osteoclastos/citologia , Receptor fas/fisiologia
7.
J Exp Med ; 199(1): 81-90, 2004 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-14699082

RESUMO

Toll-like receptor (TLR) signaling and phagocytosis are hallmarks of macrophage-mediated innate immune responses to bacterial infection. However, the relationship between these two processes is not well established. Our data indicate that TLR ligands specifically promote bacterial phagocytosis, in both murine and human cells, through induction of a phagocytic gene program. Importantly, TLR-induced phagocytosis of bacteria was found to be reliant on myeloid differentiation factor 88-dependent signaling through interleukin-1 receptor-associated kinase-4 and p38 leading to the up-regulation of scavenger receptors. Interestingly, individual TLRs promote phagocytosis to varying degrees with TLR9 being the strongest and TLR3 being the weakest inducer of this process. We also demonstrate that TLR ligands not only amplify the percentage of phagocytes uptaking Escherichia coli, but also increase the number of bacteria phagocytosed by individual macrophages. Taken together, our data describe an evolutionarily conserved mechanism by which TLRs can specifically promote phagocytic clearance of bacteria during infection.


Assuntos
Glicoproteínas de Membrana/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fagocitose/genética , Receptores de Superfície Celular/genética , Animais , Infecções Bacterianas/imunologia , Sequência de Bases , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Primers do DNA , Escherichia coli/fisiologia , Humanos , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/fisiologia , Camundongos , Receptor 3 Toll-Like , Receptor Toll-Like 9 , Receptores Toll-Like , Proteínas Quinases p38 Ativadas por Mitógeno
8.
J Exp Med ; 200(4): 437-45, 2004 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-15302901

RESUMO

Numerous bacterial products such as lipopolysaccharide potently induce type I interferons (IFNs); however, the contribution of this innate response to host defense against bacterial infection remains unclear. Although mice deficient in either IFN regulatory factor (IRF)3 or the type I IFN receptor (IFNAR)1 are highly susceptible to viral infection, we show that these mice exhibit a profound resistance to infection caused by the Gram-positive intracellular bacterium Listeria monocytogenes compared with wild-type controls. Furthermore, this enhanced bacterial clearance is accompanied by a block in L. monocytogenes-induced splenic apoptosis in IRF3- and IFNAR1-deficient mice. Thus, our results highlight the disparate roles of type I IFNs during bacterial versus viral infections and stress the importance of proper IFN modulation in host defense.


Assuntos
Apoptose/imunologia , Proteínas de Ligação a DNA/deficiência , Interferon Tipo I/imunologia , Listeriose/imunologia , Receptores de Interferon/deficiência , Fatores de Transcrição/deficiência , Animais , Primers do DNA , Suscetibilidade a Doenças , Ensaio de Imunoadsorção Enzimática , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Fator Regulador 3 de Interferon , Fígado/patologia , Macrófagos/imunologia , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase/métodos , Receptor de Interferon alfa e beta , Baço/imunologia
9.
J Gene Med ; 11(8): 655-63, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19455593

RESUMO

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.


Assuntos
Biotina/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Lentivirus/genética , Peptídeos/metabolismo , Avidina/metabolismo , Biotinilação , Western Blotting , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Humanos , Magnetismo , Mutagênese Insercional , Receptores da Transferrina/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Sindbis virus/genética , Transdução Genética , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo
10.
J Neurooncol ; 94(1): 1-19, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19468690

RESUMO

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.


Assuntos
Antígenos CD/genética , Neoplasias Encefálicas/patologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Glioblastoma/patologia , Glicoproteínas/genética , Células-Tronco Neoplásicas/fisiologia , Peptídeos/genética , Antígeno AC133 , Animais , Biomarcadores Tumorais/genética , Diferenciação Celular , Movimento Celular , Proliferação de Células , Feminino , Citometria de Fluxo/métodos , Perfilação da Expressão Gênica/métodos , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , Camundongos SCID , Transplante de Neoplasias , Células-Tronco Neoplásicas/citologia , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Esferoides Celulares/patologia , Fatores de Tempo , Células Tumorais Cultivadas
12.
Mol Biol Cell ; 16(4): 2039-48, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15673614

RESUMO

Adult myoblasts retain plasticity in developmental potential and can be induced to undergo myogenic, adipogenic, or osteoblastogenic differentiation in vitro. In this report, we show that the balance between myogenic and adipogenic potential in myoblasts is controlled by Wnt signaling. Furthermore, this balance is altered during aging such that aspects of both differentiation programs are coexpressed in myoblasts due to decreased Wnt10b abundance. Mimicking Wnt signaling in aged myoblasts through inhibition of glycogen synthase kinase or through overexpression of Wnt10b resulted in inhibition of adipogenic gene expression and sustained or enhanced myogenic differentiation. On the other hand, myoblasts isolated from Wnt10b null mice showed increased adipogenic potential, likely contributing to excessive lipid accumulation in actively regenerating myofibers in vivo in Wnt10b-/- mice. Whereas Wnt10b deficiency contributed to increased adipogenic potential in myoblasts, the augmented myogenic differentiation potential observed is likely the result of a compensatory increase in Wnt7b during differentiation of Wnt10b-/- myoblasts. No such compensation was apparent in aged myoblasts and in fact, both Wnt5b and Wnt10b were down-regulated. Thus, alteration in Wnt signaling in myoblasts with age may contribute to impaired muscle regenerative capacity and to increased muscle adiposity, both characteristic of aged muscle.


Assuntos
Adipócitos/citologia , Adipócitos/metabolismo , Diferenciação Celular , Mioblastos/citologia , Mioblastos/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Animais , Células Cultivadas , Regulação da Expressão Gênica , Metabolismo dos Lipídeos , Camundongos , Camundongos Knockout , Desenvolvimento Muscular , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Wnt
13.
Ann N Y Acad Sci ; 1415(1): 21-33, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29500936

RESUMO

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.


Assuntos
Abatacepte/farmacologia , Anabolizantes/farmacologia , Osso e Ossos/efeitos dos fármacos , Glicoproteínas/metabolismo , Linfócitos T/efeitos dos fármacos , Proteínas Wnt/metabolismo , Células 3T3 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antirreumáticos/farmacologia , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/metabolismo , Antígenos CD28/metabolismo , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , Proteínas Wnt/deficiência , Proteínas Wnt/genética , Microtomografia por Raio-X
14.
J Bone Miner Res ; 22(12): 1924-32, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17708715

RESUMO

UNLABELLED: Overexpression of Wnt10b from the osteocalcin promoter in transgenic mice increases postnatal bone mass. Increases in osteoblast perimeter, mineralizing surface, and bone formation rate without detectable changes in pre-osteoblast proliferation, osteoblast apoptosis, or osteoclast number and activity suggest that, in this animal model, Wnt10b primarily increases bone mass by stimulating osteoblastogenesis. INTRODUCTION: Wnt signaling regulates many aspects of development including postnatal accrual of bone. Potential mechanisms for how Wnt signaling increases bone mass include regulation of osteoblast and/or osteoclast number and activity. To help differentiate between these possibilities, we studied mice in which Wnt10b is expressed specifically in osteoblast lineage cells or in mice devoid of Wnt10b. MATERIALS AND METHODS: Transgenic mice, in which mouse Wnt10b is expressed from the human osteocalcin promoter (Oc-Wnt10b), were generated in C57BL/6 mice. Transgene expression was evaluated by RNase protection assay. Quantitative assessment of bone variables was done by radiography, muCT, and static and dynamic histomorphometry. Mechanisms of bone homeostasis were evaluated with assays for BrdU, TUNEL, and TRACP5b activity, as well as serum levels of C-terminal telopeptide of type I collagen (CTX). The endogenous role of Wnt10b in bone was assessed by dynamic histomorphometry in Wnt10b(-/-) mice. RESULTS: Oc-Wnt10b mice have increased mandibular bone and impaired eruption of incisors during postnatal development. Analyses of femoral distal metaphyses show significantly higher BMD, bone volume fraction, and trabecular number. Increased bone formation is caused by increases in number of osteoblasts per bone surface, rate of mineral apposition, and percent mineralizing surface. Although number of osteoclasts per bone surface is not altered, Oc-Wnt10b mice have increased total osteoclast activity because of higher bone mass. In Wnt10b(-/-) mice, changes in mineralizing variables and osteoblast perimeter in femoral distal metaphyses were not observed; however, bone formation rate is reduced because of decreased total bone volume and trabecular number. CONCLUSIONS: High bone mass in Oc-Wnt10b mice is primarily caused by increased osteoblastogenesis, with a minor contribution from elevated mineralizing activity of osteoblasts.


Assuntos
Diferenciação Celular , Osteoblastos/metabolismo , Osteocalcina , Osteogênese , Células-Tronco/metabolismo , Proteínas Wnt/biossíntese , Fosfatase Ácida/biossíntese , Fosfatase Ácida/genética , Animais , Animais Recém-Nascidos , Apoptose/genética , Densidade Óssea/genética , Diferenciação Celular/genética , Proliferação de Células , Homeostase/genética , Humanos , Incisivo/crescimento & desenvolvimento , Incisivo/metabolismo , Incisivo/patologia , Isoenzimas/biossíntese , Isoenzimas/genética , Mandíbula/crescimento & desenvolvimento , Mandíbula/metabolismo , Mandíbula/patologia , Camundongos , Camundongos Knockout , Modelos Biológicos , Tamanho do Órgão/genética , Osteoblastos/patologia , Osteoclastos/metabolismo , Osteoclastos/patologia , Células-Tronco/patologia , Fosfatase Ácida Resistente a Tartarato , Transgenes , Proteínas Wnt/genética
15.
Cancer Res ; 65(11): 4496-9, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15930264

RESUMO

Expression of cyclooxygenase 2 (COX-2) in breast cancer correlates with poor prognosis, and COX-2 enzyme inhibitors reduce breast cancer incidence in humans. We recently showed that COX-2 overexpression in the mammary gland of transgenic mice induced mammary cancer. Because prostaglandin E2 (PGE2) is the major eicosanoid and because the EP2 subtype of the PGE2 receptor is highly expressed in the mammary tumors, we tested if this G protein-coupled receptor is required for tumorigenesis. We crossed the MMTV-COX-2 transgenic mice with Ep2-/- mice and studied tumor development in bigenic mice. Lack of EP2 receptor strongly suppressed COX-2-induced effects such as precocious development of the mammary gland in virgins and the development of mammary hyperplasia in multiparous female mice. Interestingly, the expression of amphiregulin, a potent mammary epithelial cell growth factor was down regulated in mammary glands of Ep2-/- mice. Total cyclic AMP (cAMP) levels were reduced in Ep2-/- mammary glands suggesting that PGE2 signaling via the EP2 receptor activates the Gs/cAMP/protein kinase A pathway. In mammary tumor cell lines, expression of the EP2 receptor followed by treatment with CAY10399, an EP2-specific agonist, strongly induced amphiregulin mRNA levels in a protein kinase A-dependent manner. These data suggest that PGE2 signaling via the EP2 receptor in mammary epithelial cells regulate mammary gland hyperplasia by the cAMP-dependent induction of amphiregulin. Inhibition of the EP2 pathway in the mammary gland may be a novel approach in the prevention and/or treatment of mammary cancer.


Assuntos
Glândulas Mamárias Animais/patologia , Prostaglandina-Endoperóxido Sintases/fisiologia , Receptores de Prostaglandina E/fisiologia , Anfirregulina , Animais , Ciclo-Oxigenase 2 , Família de Proteínas EGF , Feminino , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/biossíntese , Hiperplasia , Endogamia , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Masculino , Glândulas Mamárias Animais/enzimologia , Glândulas Mamárias Animais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Prostaglandina-Endoperóxido Sintases/biossíntese , Prostaglandina-Endoperóxido Sintases/genética , Receptores de Prostaglandina E/deficiência , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E Subtipo EP2 , Transdução de Sinais
16.
Nat Commun ; 8(1): 1620, 2017 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-29158473

RESUMO

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.


Assuntos
Artérias/metabolismo , Mecanotransdução Celular , Receptor Notch1/metabolismo , Animais , Artérias/química , Cálcio/metabolismo , Células Endoteliais/química , Células Endoteliais/metabolismo , Endotélio Vascular/química , Endotélio Vascular/metabolismo , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor Notch1/genética , Estresse Mecânico
17.
Oncogene ; 22(38): 6032-44, 2003 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-12955082

RESUMO

Human BRCA1 has a genetically demonstrated role in DNA repair, and has been proposed to act as a transcriptional activator in a limited number of specialized settings. To gain insight into biologically conserved functional motifs, we isolated an ortholog of BRCA1 from cattle (Bos taurus). The predicted protein product shows 72.5% sequence identity with the human protein and conservation of amino acids involved in BRCA1 structure and function. Although the bovine C-terminus is truncated by seven amino acids as compared to human, bovine BRCA1 protein exhibited a similar cell cycle-regulated nuclear expression pattern. Expression was characteristically low and diffuse in the nucleus of G1/G0 cells, followed by increasing BRCA1-positive nuclear speckles in late S phase and G2/M phase cells. Bovine BRCA1 was phosphorylated and nuclear speckling was enhanced in response to DNA-damaging agents. Consistent with evidence from studies of human BRCA1, bovine BRCA1 was shown to interact with RNA polymerase II in vivo, an activity that was mapped to the C-terminal domain (CTD) (bBRCA(1364-1849)). Interestingly, when tested in the GAL4 transcriptional activation assay, full-length bovine and human BRCA1 lacked any ability to act as transcriptional activators and the CTD of bovine BRCA1 had five-fold lower activity when compared to the more acidic human C-terminus. These results provide evidence that phosphorylation and nuclear relocalization are highly conserved features of the BRCA1 response to genotoxic stress. In addition, bovine BRCA1 binds the RNA polymerase II holoenzyme, but this interaction lacks significant ability to correctly orient or recruit RNA polymerase II for transcription in the classic GAL4 transcriptional activation system.


Assuntos
Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Dano ao DNA , Ativação Transcricional/efeitos dos fármacos , Processamento Alternativo , Sequência de Aminoácidos , Animais , Afidicolina/toxicidade , Proteína BRCA1/imunologia , Bovinos , Células Cultivadas , Mapeamento Cromossômico , Clonagem Molecular , Sequência Conservada , DNA/efeitos dos fármacos , Dano ao DNA/fisiologia , Humanos , Hidroxiureia/toxicidade , Dados de Sequência Molecular , Testes de Mutagenicidade , Mutação , Fosforilação , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fase S/fisiologia , Homologia de Sequência de Aminoácidos
18.
Prostaglandins Other Lipid Mediat ; 76(1-4): 86-94, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15967164

RESUMO

Cyclooxygenase-2 (COX-2) overexpression is a widely recognized feature of human breast cancer and inhibitors of the enzyme have antitumor effects in a subset of tumor settings. Previously, we demonstrated that direct overexpression of COX-2 under control of the mammary-specific MMTV promoter/enhancer, was itself oncogenic and lead to the induction of mammary tumors in multiparous, outbred CD1 mice. In the present study, we provide evidence that COX-2 dependent tumor progression can also be studied in FVB/N, an inbred strain widely used for analysis of breast cancer progression. In these mice, the human COX-2 transgene was strongly induced during pregnancy/lactation and mammary tumors developed after multiple pregnancies. However, crossing the COX-2 FVB/N mice with the C57BL6 strain resulted in loss of the mammary tumorigenic phenotype despite the fact that the human COX-2 gene was induced. Treatment of the COX-2 transgenic mice in the FVB/N strain with celecoxib (1600 ppm), a COX-2 selective inhibitor, resulted significant reduction in tumor size and multiplicity when compared to transgenic mice fed with control chow. SC-560 (20 ppm), a COX-1 selective inhibitor did not have significant effect on tumorigenesis. These studies suggest that FVB/N is a susceptible mouse strain well suited to the study of COX-2 mediated tumor progression and may provide a tool for the identification of interacting genes and therapeutic treatments for this clinically important target.


Assuntos
Inibidores de Ciclo-Oxigenase/farmacologia , Neoplasias Mamárias Experimentais/patologia , Prostaglandina-Endoperóxido Sintases/efeitos dos fármacos , Animais , Sequência de Bases , Celecoxib , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Primers do DNA , Humanos , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Compostos Orgânicos/farmacologia , Prostaglandina-Endoperóxido Sintases/genética , Prostaglandina-Endoperóxido Sintases/fisiologia , Pirazóis/farmacologia , Sulfonamidas/farmacologia
19.
Cancer Res ; 75(22): 4960-72, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26400062

RESUMO

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.


Assuntos
Neoplasias da Mama/patologia , Invasividade Neoplásica/patologia , Transdução de Sinais/fisiologia , Proteínas ras/metabolismo , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Intervalo Livre de Doença , Feminino , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Camundongos , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Análise de Sequência com Séries de Oligonucleotídeos
20.
Cancer Biol Ther ; 3(6): 528-33, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15254397

RESUMO

The BRCA1 tumor suppressor gene is expressed in all mammalian cells. Within these cells, the BRCA1 protein product interacts with several seemingly distinct nuclear complexes. Proteins within these complexes are potential targets for the E3-ubiquitin ligase activity associated with BRCA1:BARD1 complexes. Recent breakthroughs have centered on elucidating critical DNA repair and chromatin-remodeling functions associated with BRCA1 activity. During both DNA replication and DNA repair, BRCA1 appears to serve both adaptor and enzymatic functions. Roles include transient physical recruitment of NBS1, gammaH2AX, FANCD2 and other proteins in specific repair associated complexes, and enzymatic activity as an E3-ubiquitin ligase against a subset of these proteins. BRCA1 has also been implicated as a regulator of transcription. It is in this second capacity that progress has been much more difficult to assess. In particular, unambiguous adaptor and enzymatic functions have yet to be demonstrated in transcriptional machinery. Addressing the critical gap in our understanding of enzymatic targets of BRCA1 will be required for significant future progress in this field. The following review puts forward a model for BRCA1 interactions with the transcriptional complex in undamaged cells, and a potential mechanism for substrate switching between transcription and DNA-repair complexes following exposure of cells to proliferative or genotoxic stress. This model incorporates recent evidence that BRCA1 interacts predominantly with hyper-phosphorylated, enzymatically active, RNA polymerase II (RNAPII) in undamaged cells. The model proposes that BRCA1 binds processive RNA polymerase as part of a genome surveillance function, upstream of critical roles in DNA repair.


Assuntos
Proteína BRCA1/genética , Reparo do DNA , RNA Polimerase II/genética , Transcrição Gênica , Dano ao DNA , Feminino , Humanos , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA