Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Signal ; 10(485)2017 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-28655862

RESUMO

Constitutive WNT activity drives the growth of various human tumors, including nearly all colorectal cancers (CRCs). Despite this prominence in cancer, no WNT inhibitor is currently approved for use in the clinic largely due to the small number of druggable signaling components in the WNT pathway and the substantial toxicity to normal gastrointestinal tissue. We have shown that pyrvinium, which activates casein kinase 1α (CK1α), is a potent inhibitor of WNT signaling. However, its poor bioavailability limited the ability to test this first-in-class WNT inhibitor in vivo. We characterized a novel small-molecule CK1α activator called SSTC3, which has better pharmacokinetic properties than pyrvinium, and found that it inhibited the growth of CRC xenografts in mice. SSTC3 also attenuated the growth of a patient-derived metastatic CRC xenograft, for which few therapies exist. SSTC3 exhibited minimal gastrointestinal toxicity compared to other classes of WNT inhibitors. Consistent with this observation, we showed that the abundance of the SSTC3 target, CK1α, was decreased in WNT-driven tumors relative to normal gastrointestinal tissue, and knocking down CK1α increased cellular sensitivity to SSTC3. Thus, we propose that distinct CK1α abundance provides an enhanced therapeutic index for pharmacological CK1α activators to target WNT-driven tumors.


Assuntos
Antineoplásicos/farmacologia , Benzoatos/farmacologia , Caseína Quinase Ialfa/metabolismo , Ativadores de Enzimas/farmacologia , Neoplasias/tratamento farmacológico , Proteínas Wnt/metabolismo , Animais , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Metástase Neoplásica , Técnicas de Cultura de Órgãos , Fosforilação , Compostos de Pirvínio/química , Transdução de Sinais , Ressonância de Plasmônio de Superfície , Via de Sinalização Wnt , Ensaios Antitumorais Modelo de Xenoenxerto , Xenopus laevis
2.
J Control Release ; 255: 12-26, 2017 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-28366646

RESUMO

A rationally-designed library of ternary siRNA polyplexes was developed and screened for gene silencing efficacy in vitro and in vivo with the goal of overcoming both cell-level and systemic delivery barriers. [2-(dimethylamino)ethyl methacrylate] (DMAEMA) was homopolymerized or copolymerized (50mol% each) with butyl methacrylate (BMA) from a reversible addition - fragmentation chain transfer (RAFT) chain transfer agent, with and without pre-conjugation to polyethylene glycol (PEG). Both single block polymers were tested as core-forming units, and both PEGylated, diblock polymers were screened as corona-forming units. Ternary siRNA polyplexes were assembled with varied amounts and ratios of core-forming polymers to PEGylated corona-forming polymers. The impact of polymer composition/ratio, hydrophobe (BMA) placement, and surface PEGylation density was correlated to important outcomes such as polyplex size, stability, pH-dependent membrane disruptive activity, biocompatibility, and gene silencing efficiency. The lead formulation, DB4-PDB12, was optimally PEGylated not only to ensure colloidal stability (no change in size by DLS between 0 and 24h) and neutral surface charge (0.139mV) but also to maintain higher cell uptake (>90% positive cells) than the most densely PEGylated particles. The DB4-PDB12 polyplexes also incorporated BMA in both the polyplex core- and corona-forming polymers, resulting in robust endosomolysis and in vitro siRNA silencing (~85% protein level knockdown) of the model gene luciferase across multiple cell types. Further, the DB4-PDB12 polyplexes exhibited greater stability, increased blood circulation time, reduced renal clearance, increased tumor biodistribution, and greater silencing of luciferase compared to our previously-optimized, binary parent formulation following intravenous (i.v.) delivery. This polyplex library approach enabled concomitant optimization of the composition and ratio of core- and corona-forming polymers (indirectly tuning PEGylation density) and identification of a ternary nanomedicine optimized to overcome important siRNA delivery barriers in vitro and in vivo.


Assuntos
Metacrilatos/química , Polietilenoglicóis/química , RNA Interferente Pequeno/administração & dosagem , Animais , Linhagem Celular , Linhagem Celular Tumoral , Feminino , Humanos , Interações Hidrofóbicas e Hidrofílicas , Luciferases/genética , Camundongos , Camundongos Nus , RNA Interferente Pequeno/química , RNA Interferente Pequeno/farmacocinética , Distribuição Tecidual
3.
J Cell Biol ; 214(2): 197-213, 2016 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-27402952

RESUMO

Exosomes are extracellular vesicles that influence cellular behavior and enhance cancer aggressiveness by carrying bioactive molecules. The mechanisms that regulate exosome secretion are poorly understood. Here, we show that the actin cytoskeletal regulatory protein cortactin promotes exosome secretion. Knockdown or overexpression of cortactin in cancer cells leads to a respective decrease or increase in exosome secretion, without altering exosome cargo content. Live-cell imaging revealed that cortactin controls both trafficking and plasma membrane docking of multivesicular late endosomes (MVEs). Regulation of exosome secretion by cortactin requires binding to the branched actin nucleating Arp2/3 complex and to actin filaments. Furthermore, cortactin, Rab27a, and coronin 1b coordinately control stability of cortical actin MVE docking sites and exosome secretion. Functionally, the addition of purified exosomes to cortactin-knockdown cells rescued defects of those cells in serum-independent growth and invasion. These data suggest a model in which cortactin promotes exosome secretion by stabilizing cortical actin-rich MVE docking sites.


Assuntos
Actinas/metabolismo , Cortactina/metabolismo , Exossomos/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Transporte Biológico , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Cortactina/ultraestrutura , Exossomos/ultraestrutura , Humanos , Proteínas dos Microfilamentos , Modelos Biológicos , Simulação de Acoplamento Molecular , Corpos Multivesiculares/metabolismo , Corpos Multivesiculares/ultraestrutura , Fenótipo , Ligação Proteica , Pseudópodes/metabolismo , RNA Interferente Pequeno/metabolismo , Tetraspanina 30/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
4.
Ann Biomed Eng ; 43(8): 1978-88, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25549772

RESUMO

Preterm premature rupture of membranes causes 40% of all preterm births, affecting 150000 women each year in the United States. Prenatal diagnostic procedures and surgical interventions increase incidence of adverse events, leading to iatrogenic membrane rupture after a fetoscopic procedure in 45% of cases. We propose an ultrathin, self-adherent, poly-L-lactic acid patch ("nanofilm") as a reparative wound closure after endoscopic/fetoscopic procedures. These nanofilms are compatible with application in wet conditions and with minimally invasive instrumentation. Ex vivo studies to evaluate the nanofilm were conducted using human chorion-amnion (CA) membranes. A custom-built inflation device was used for mechanical characterization of CA membranes and for assessment of nanofilm adhesion and sealing of membrane defects up to 3 mm in size. These ex vivo tests demonstrated the ability of the nanofilm to seal human CA defects ranging in size from 1 to 3 mm in diameter. In vivo survival studies were conducted in 25 mid-gestational rabbits, defects were created by perforating the uterus and the CA membranes and subsequently using the nanofilm to seal these wounds. These in vivo studies confirmed the successful sealing of defects smaller than 3 mm observed ex vivo. Histological analysis of whole harvested uteri 7 days after surgery showed intact uterine walls in 59% of the nanofilm repaired fetuses, along with increased uterine size and intrauterine development in 63% of the cases. In summary, we have developed an ultrathin, self-adhesive nanofilm for repair of uterine membrane defects.


Assuntos
Ruptura Prematura de Membranas Fetais/cirurgia , Ácido Láctico , Membranas Artificiais , Polímeros , Nascimento Prematuro/prevenção & controle , Adesivos Teciduais , Animais , Feminino , Humanos , Ácido Láctico/química , Ácido Láctico/farmacologia , Poliésteres , Polímeros/química , Polímeros/farmacologia , Gravidez , Coelhos , Adesivos Teciduais/química , Adesivos Teciduais/farmacologia
5.
Biomaterials ; 38: 97-107, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25453977

RESUMO

A series of endosomolytic mixed micelles was synthesized from two diblock polymers, poly[ethylene glycol-b-(dimethylaminoethyl methacrylate-co-propylacrylic acid-co-butyl methacrylate)] (PEG-b-pDPB) and poly[dimethylaminoethyl methacrylate-b-(dimethylaminoethyl methacrylate-co-propylacrylic acid-co-butyl methacrylate)] (pD-b-pDPB), and used to determine the impact of both surface PEG density and PEG molecular weight on overcoming both intracellular and systemic siRNA delivery barriers. As expected, the percent PEG composition and PEG molecular weight in the corona had an inverse relationship with mixed micelle zeta potential and rate of cellular internalization. Although mixed micelles were internalized more slowly, they generally produced similar gene silencing bioactivity (∼ 80% or greater) in MDA-MB-231 breast cancer cells as the micelles containing no PEG (100 D/no PEG). The mechanistic explanation for the potent bioactivity of the promising 50 mol% PEG-b-DPB/50 mol% pD-b-pDPB (50 D) mixed micelle formulation, despite its relatively low rate of cellular internalization, was further investigated as a function of PEG molecular weight (5 k, 10 k, or 20 k PEG). Results indicated that, although larger molecular weight PEG decreased cellular internalization, it improved cytoplasmic bioavailability due to increased intracellular unpackaging (quantitatively measured via FRET) and endosomal release. When delivered intravenously in vivo, 50 D mixed micelles with a larger molecular weight PEG in the corona also demonstrated significantly improved blood circulation half-life (17.8 min for 20 k PEG micelles vs. 4.6 min for 5 kDa PEG micelles) and a 4-fold decrease in lung accumulation. These studies provide new mechanistic insights into the functional effects of mixed micelle-based approaches to nanocarrier surface PEGylation. Furthermore, the ideal mixed micelle formulation identified (50 D/20 k PEG) demonstrated desirable intracellular and systemic pharmacokinetics and thus has strong potential for in vivo therapeutic use.


Assuntos
Composição de Medicamentos/métodos , Nanocápsulas/química , Nanocápsulas/ultraestrutura , Polietilenoglicóis/química , RNA Interferente Pequeno/química , RNA Interferente Pequeno/farmacocinética , Frações Subcelulares/metabolismo , Animais , Difusão , Humanos , Metacrilatos/química , Camundongos , Camundongos Endogâmicos BALB C , Micelas , Tamanho da Partícula , RNA Interferente Pequeno/sangue , Frações Subcelulares/química
6.
Biomacromolecules ; 16(1): 192-201, 2015 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-25414930

RESUMO

A dual-targeted siRNA nanocarrier has been synthesized and validated that is selectively activated in environments where there is colocalization of two breast cancer hallmarks, elevated matrix metalloproteinase (MMP) activity and folate receptor overexpression. This siRNA nanocarrier is self-assembled from two polymers containing the same pH-responsive, endosomolytic core-forming block but varying hydrophilic, corona-forming blocks. The corona block of one polymer consists of a 2 kDa PEG attached to a terminal folic acid (FA); the second polymer contains a larger (Y-shaped, 20 kDa) PEG attached to the core block by a proximity-activated targeting (PAT), MMP7-cleavable peptide. In mixed micelle smart polymer nanoparticles (SPNs) formed from the FA- and PAT-based polymers, the proteolytically removable PEG on the PAT polymers shields nonspecific SPN interactions with cells or proteins. When the PAT element is cleaved within an MMP-rich environment, the PEG shielding is removed, exposing the underlying FA and making it accessible for folate receptor-mediated SPN uptake. Characterization of mixed micelles prepared from these two polymers revealed that uptake and siRNA knockdown bioactivity of a 50% FA/50% PAT formulation was dependent on both proteolytic activation and FA receptor engagement. MMP activation and delivery of this formulation to breast cancer cells expressing the FA receptor achieved greater than 50% protein-level knockdown of a model gene with undetectable cytotoxicity. This modular nanoparticle design represents a new paradigm in cell-selective siRNA delivery and allows for stoichiometric tuning of dual-targeting components to achieve superior targeting specificity.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Receptores de Folato com Âncoras de GPI , Técnicas de Transferência de Genes , Metaloproteinase 7 da Matriz , Nanopartículas/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Receptores de Folato com Âncoras de GPI/metabolismo , Humanos , Células MCF-7 , Metaloproteinase 7 da Matriz/metabolismo , Nanopartículas/metabolismo , RNA Interferente Pequeno/metabolismo
7.
Cell Rep ; 5(5): 1159-68, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24290760

RESUMO

Unconventional secretion of exosome vesicles from multivesicular endosomes (MVEs) occurs across a broad set of systems and is reported to be upregulated in cancer, where it promotes aggressive behavior. However, regulatory control of exosome secretion is poorly understood. Using cancer cells, we identified specialized invasive actin structures called invadopodia as specific and critical docking and secretion sites for CD63- and Rab27a-positive MVEs. Thus, inhibition of invadopodia formation greatly reduced exosome secretion into conditioned media. Functionally, addition of purified exosomes or inhibition of exosome biogenesis or secretion greatly affected multiple invadopodia life cycle steps, including invadopodia formation, stabilization, and exocytosis of proteinases, indicating a key role for exosome cargoes in promoting invasive activity and providing in situ signaling feedback. Exosome secretion also controlled cellular invasion through three-dimensional matrix. These data identify a synergistic interaction between invadopodia biogenesis and exosome secretion and reveal a fundamental role for exosomes in promoting cancer cell invasiveness.


Assuntos
Movimento Celular , Exocitose , Exossomos/metabolismo , Pseudópodes/metabolismo , Actinas/metabolismo , Linhagem Celular Tumoral , Humanos , Via Secretória , Tetraspanina 30/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Proteínas rab27 de Ligação ao GTP
8.
Cytoskeleton (Hoboken) ; 69(9): 625-43, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22991200

RESUMO

Cortactin is a branched actin regulator and tumor-overexpressed protein that promotes vesicular trafficking at a variety of cellular sites, including endosomes and the trans-Golgi network. To better understand its role in secretory trafficking, we investigated its function in Golgi homeostasis. Here, we report that knockdown (KD) of cortactin leads to a dramatic change in Golgi morphology by light microscopy, dependent on binding the Arp2/3 actin-nucleating complex. Surprisingly, there was little effect of cortactin-KD on anterograde trafficking of the constitutive cargo vesicular stomatitis virus glycoprotein (VSVG), Golgi assembly from endoplasmic reticulum membranes upon Brefeldin A washout, or Golgi ultrastructure. Instead, electron microscopy studies revealed that cortactin-KD cells contained a large number of immature-appearing late endosomal/lysosomal (LE/Lys) hybrid organelles, similar to those found in lysosomal storage diseases. Consistent with a defect in LE/Lys trafficking, cortactin-KD cells also exhibited accumulation of free cholesterol and retention of the retrograde Golgi cargo mannose-6-phosphate receptor in LE. Inhibition of LE maturation by treatment of control cells with Rab7 siRNA or chloroquine led to a compact Golgi morphology similar to that observed in cortactin-KD cells. Furthermore, the Golgi morphology defects of cortactin-KD cells could be rescued by removal of cholesterol-containing lipids from the media, suggesting that buildup of cholesterol-rich membranes in immature LE/Lys induced disturbances in retrograde trafficking. Taken together, these data reveal that LE/Lys maturation and trafficking are highly sensitive to cortactin-regulated branched actin assembly and suggests that cytoskeletal-induced Golgi morphology changes can be a consequence of altered trafficking at late endosomes.


Assuntos
Actinas/metabolismo , Cortactina/metabolismo , Endossomos/metabolismo , Complexo de Golgi/metabolismo , Lisossomos/metabolismo , Linhagem Celular Tumoral , Cortactina/genética , Humanos , Microscopia Confocal , Microscopia Eletrônica de Transmissão
9.
Neoplasia ; 13(8): 758-70, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21847367

RESUMO

The type III TGF-ß receptor (TßRIII or betagylcan) is a TGF-ß superfamily coreceptor with emerging roles in regulating TGF-ß superfamily signaling and cancer progression. Alterations in TGF-ß superfamily signaling are common in colon cancer; however, the role of TßRIII has not been examined. Although TßRIII expression is frequently lost at the message and protein level in human cancers and suppresses cancer progression in these contexts, here we demonstrate that, in colon cancer, TßRIII messenger RNA expression is not significantly altered and TßRIII expression is more frequently increased at the protein level, suggesting a distinct role for TßRIII in colon cancer. Increasing TßRIII expression in colon cancer model systems enhanced ligand-mediated phosphorylation of p38 and the Smad proteins, while switching TGF-ß and BMP-2 from inhibitors to stimulators of colon cancer cell proliferation, inhibiting ligand-induced p21 and p27 expression. In addition, increasing TßRIII expression increased ligand-stimulated anchorage-independent growth, a resistance to ligand- and chemotherapy-induced apoptosis, cell migration and modestly increased tumorigenicity in vivo. In a reciprocal manner, silencing endogenous TßRIII expression decreased colon cancer cell migration. These data support a model whereby TßRIII mediates TGF-ß superfamily ligand-induced colon cancer progression and support a context-dependent role for TßRIII in regulating cancer progression.


Assuntos
Movimento Celular , Neoplasias do Colo/metabolismo , Proteoglicanas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Transformação Celular Neoplásica/genética , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células HT29 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteoglicanas/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cell Adh Migr ; 5(2): 187-98, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21258212

RESUMO

Branched actin assembly is critical for a variety of cellular processes that underlie cell motility and invasion, including cellular protrusion formation and membrane trafficking. Activation of branched actin assembly occurs at various subcellular locations via site-specific activation of distinct WASp family proteins and the Arp2/3 complex. A key branched actin regulator that promotes cell motility and links signaling, cytoskeletal and membrane trafficking proteins is the Src kinase substrate and Arp2/3 binding protein cortactin. Due to its frequent overexpression in advanced, invasive cancers and its general role in regulating branched actin assembly at multiple cellular locations, cortactin has been the subject of intense study. Recent studies suggest that cortactin has a complex role in cellular migration and invasion, promoting both on-site actin polymerization and modulation of autocrine secretion. Diverse cellular activities may derive from the interaction of cortactin with site-specific binding partners.


Assuntos
Proteína 2 Relacionada a Actina/metabolismo , Proteína 3 Relacionada a Actina/metabolismo , Actinas/metabolismo , Movimento Celular , Cortactina , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo , Citoesqueleto de Actina/fisiologia , Animais , Sítios de Ligação , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Comunicação Celular , Cortactina/genética , Cortactina/metabolismo , Citoesqueleto/metabolismo , Feminino , Expressão Gênica , Humanos , Camundongos , Neoplasias/genética , Neoplasias/metabolismo , Polimerização , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais
11.
Mol Biol Cell ; 20(20): 4362-70, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19726563

RESUMO

Bone morphogenetic proteins (BMPs) signal through the BMP type I and type II receptors to regulate cellular processes, including embryonic development. The type I BMP receptors activin-like kinase (ALK)3 and ALK6 share a high degree of homology, yet possess distinct signaling roles. Here, we report that although the transforming growth factor (TGF)-beta type III receptor (TbetaRIII) enhanced both ALK3 and ALK6 signaling, TbetaRIII more potently enhanced ALK6-mediated stimulation of the BMP-responsive promoters XVent2 and 3GC2, and up-regulation of the early response gene Smad6. In contrast, TbetaRIII specifically enhanced ALK3-mediated up-regulation of the early response gene ID-1. TbetaRIII associated with ALK3 primarily through their extracellular domains, whereas its interaction with ALK6 required both the extracellular and cytoplasmic domains. TbetaRIII, along with its interacting scaffolding protein beta-arrestin2, induced the internalization of ALK6. In contrast, TbetaRIII colocalized with and resulted in the cell surface retention of ALK3, independently of beta-arrestin2. Although complex formation between TbetaRIII, ALK6, and beta-arrestin2 and TbetaRIII/ALK6 internalization resulted in maximal BMP signaling, the TbetaRIII mutant unable to interact with beta-arrestin2, TbetaRIII-T841A, was unable to do so. These studies support a novel role for TbetaRIII in mediating differential ALK3 and ALK6 subcellular trafficking resulting in distinct signaling downstream of ALK3 and ALK6.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo I/fisiologia , Regulação da Expressão Gênica/fisiologia , Transporte Proteico/fisiologia , Proteoglicanas/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Transdução de Sinais/fisiologia , Animais , Arrestinas/fisiologia , Proteínas Morfogenéticas Ósseas/fisiologia , Células COS , Linhagem Celular/metabolismo , Linhagem Celular Tumoral/metabolismo , Chlorocebus aethiops , Humanos , Rim , Camundongos , Complexos Multiproteicos/fisiologia , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Proteoglicanas/química , Proteoglicanas/genética , Receptores de Fatores de Crescimento Transformadores beta/química , Receptores de Fatores de Crescimento Transformadores beta/genética , Relação Estrutura-Atividade , Frações Subcelulares/metabolismo , Teratocarcinoma/patologia , beta-Arrestinas
12.
Laryngoscope ; 119(12): 2315-23, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19693929

RESUMO

OBJECTIVES/HYPOTHESIS: To develop a reliable modeling system for head and neck squamous cell carcinoma (HNSCC). STUDY DESIGN: Laboratory-based translational study. METHODS: HNSCC tissue was obtained from patients at biopsy/resection, cultured, and implanted into mice. In vivo, tumor growth, and survival was monitored by bioluminescence imaging. Histology and immunohistochemistry (IHC) were used to confirm HNSCC and human origin. RESULTS: Short-term culture techniques were optimized allowing survival of primary HNSCC cells more than 7 days in 76% of tumors. The size of the tumor biopsy collected did not correlate with the success of short-term culture or xenograft establishment. Xenograft modeling was attempted in primary HNSCCs from 12 patients with a success rate of 92%. Immunostaining confirmed human origin of epithelial tumor cells within the modeled tumor. Bioluminescence and Ki67 IHC suggested tumor proliferation within the model. Luciferase expression was maintained for as long as 100 days in modeled tumors. CONCLUSIONS: The techniques developed for short-term primary tumor culture followed by xenograft modeling provide a low-cost and tractable model for evaluation of HNSCC response to standard and novel therapies. The high success rate of human-in-mouse tumor formation from primary HNSCC suggests that selection pressures for tumor growth in this model may be less than those observed for establishment of cell lines. Bioluminescent imaging provides a useful tool for evaluating tumor growth and could be expanded to measure response of the modeled tumor to therapy. This model could be adapted for xenograft modeled growth of other primary tumor types.


Assuntos
Carcinoma de Células Escamosas/patologia , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias Experimentais/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Biópsia , Proliferação de Células , Feminino , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Transplante de Neoplasias , Ratos , Traqueia/citologia , Traqueia/transplante , Transplante Heterólogo , Células Tumorais Cultivadas
13.
Carcinogenesis ; 30(2): 238-48, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19056927

RESUMO

Bone morphogenetic proteins (BMPs) have an emerging role in human cancers. Here we demonstrate that the BMP-signaling pathway is intact and functional in human pancreatic cancer cells, with several BMP signaling components and transcriptional targets upregulated in human pancreatic cancer specimens compared with normal pancreatic tissue. Functionally, multiple BMP family members, including BMP-2, BMP-4 and BMP-7, induce an epithelial to mesenchymal transition (EMT) in the human pancreatic cancer cell line Panc-1, as demonstrated by morphological alterations and loss of E-cadherin expression. BMP-mediated EMT results in an increase in invasiveness of Panc-1 cells, in part through increased expression and activity of matrix metalloproteinase (MMP)-2, a known mediator of pancreatic cancer cell invasiveness. Accompanying EMT, BMP reduces expression of the transforming growth factor (TGF)-beta superfamily receptor, transforming growth factor-beta type III receptor (TbetaRIII), for which we have previously demonstrated loss of expression during pancreatic cancer progression. Maintaining TbetaRIII expression inhibits BMP-mediated invasion and suppresses Smad1 activation. Further, Smad1 is required for BMP-induced invasiveness and partially responsible for BMP-mediated increases in MMP-2 activity. These data suggest that BMP signaling, through Smad1 induction and upregulation of MMP-2, is an important mediator of pancreatic cancer invasiveness and a potential therapeutic target for treating this deadly disease.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Metaloproteinase 2 da Matriz/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteína Smad1/metabolismo , Proteínas Morfogenéticas Ósseas/farmacologia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Humanos , Mesoderma/citologia , Mesoderma/fisiologia , Invasividade Neoplásica , Neoplasias Pancreáticas/patologia , Proteoglicanas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/fisiologia , Proteína Smad1/genética , Regulação para Cima
14.
J Biol Chem ; 283(12): 7628-37, 2008 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-18184661

RESUMO

The bone morphogenetic protein (BMP) family, the largest subfamily of the structurally conserved transforming growth factor-beta (TGF-beta) superfamily of growth factors, are multifunctional regulators of development, proliferation, and differentiation. The TGF-beta type III receptor (TbetaRIII or betaglycan) is an abundant cell surface proteoglycan that has been well characterized as a TGF-beta and inhibin receptor. Here we demonstrate that TbetaRIII functions as a BMP cell surface receptor. TbetaRIII directly and specifically binds to multiple members of the BMP subfamily, including BMP-2, BMP-4, BMP-7, and GDF-5, with similar kinetics and ligand binding domains as previously identified for TGF-beta. TbetaRIII also enhances ligand binding to the BMP type I receptors, whereas short hairpin RNA-mediated silencing of endogenous TbetaRIII attenuates BMP-mediated Smad1 phosphorylation. Using a biologically relevant model for TbetaRIII function, we demonstrate that BMP-2 specifically stimulates TbetaRIII-mediated epithelial to mesenchymal cell transformation. The ability of TbetaRIII to serve as a cell surface receptor and mediate BMP, inhibin, and TGF-beta signaling suggests a broader role for TbetaRIII in orchestrating TGF-beta superfamily signaling.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Inibinas/metabolismo , Proteoglicanas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Animais , Células COS , Chlorocebus aethiops , Humanos , Cinética , Fosforilação , Ligação Proteica , Proteína Smad1/metabolismo
15.
J Clin Invest ; 117(1): 206-17, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17160136

RESUMO

The TGF-beta signaling pathway has a complex role in regulating mammary carcinogenesis. Here we demonstrate that the type III TGF-beta receptor (TbetaRIII, or betaglycan), a ubiquitously expressed TGF-beta coreceptor, regulated breast cancer progression and metastasis. Most human breast cancers lost TbetaRIII expression, with loss of heterozygosity of the TGFBR3 gene locus correlating with decreased TbetaRIII expression. TbetaRIII expression decreased during breast cancer progression, and low TbetaRIII levels predicted decreased recurrence-free survival in breast cancer patients. Restoring TbetaRIII expression in breast cancer cells dramatically inhibited tumor invasiveness in vitro and tumor invasion, angiogenesis, and metastasis in vivo. TbetaRIII appeared to inhibit tumor invasion by undergoing ectodomain shedding and producing soluble TbetaRIII, which binds and sequesters TGF-beta to decrease TGF-beta signaling and reduce breast cancer cell invasion and tumor-induced angiogenesis. Our results indicate that loss of TbetaRIII through allelic imbalance is a frequent genetic event during human breast cancer development that increases metastatic potential.


Assuntos
Neoplasias Mamárias Animais/patologia , Neoplasias Mamárias Animais/prevenção & controle , Proteoglicanas/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Animais , Modelos Animais de Doenças , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Mamárias Animais/genética , Camundongos , Invasividade Neoplásica , Metástase Neoplásica , Proteoglicanas/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Transdução de Sinais
16.
Trends Biochem Sci ; 30(11): 611-21, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16185874

RESUMO

Extracellular signals are transmitted to cells through two classes of cell-surface receptors: signaling receptors that directly transduce signals and signaling co-receptors that bind ligand but that, traditionally, have not been thought to signal directly. Signaling co-receptors modulate the ligand binding and signaling of their respective signaling receptors. In recent years, roles for co-receptors have expanded to include essential functions in morphogen gradient formation, localizing signaling, signaling independently, regulating cell adhesion and orchestrating the signaling of several pathways. The importance of signaling co-receptors is demonstrated by their ubiquitous expression, their conservation during evolution, their prominent role in signaling cascades, their indispensable role during development and their frequent mutation or altered expression in human disease.


Assuntos
Doenças Genéticas Inatas/genética , Receptores de Superfície Celular/fisiologia , Transdução de Sinais , Animais , Adesão Celular , Desenvolvimento Embrionário , Humanos , Ligantes , Camundongos , Modelos Biológicos , Mutação , Receptores de Superfície Celular/metabolismo
17.
Science ; 301(5638): 1394-7, 2003 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-12958365

RESUMO

beta-Arrestins bind to activated seven transmembrane-spanning (7TMS) receptors (G protein-coupled receptors) after the receptors are phosphorylated by G protein-coupled receptor kinases (GRKs), thereby regulating their signaling and internalization. Here, we demonstrate an unexpected and analogous role of beta-arrestin 2 (betaarr2) for the single transmembrane-spanning type III transforming growth factor-beta (TGF-beta) receptor (TbetaRIII, also referred to as betaglycan). Binding of betaarr2 to TbetaRIII was also triggered by phosphorylation of the receptor on its cytoplasmic domain (likely at threonine 841). However, such phosphorylation was mediated by the type II TGF-beta receptor (TbetaRII), which is itself a kinase, rather than by a GRK. Association with betaarr2 led to internalization of both receptors and down-regulation of TGF-beta signaling. Thus, the regulatory actions of beta-arrestins are broader than previously appreciated, extending to the TGF-beta receptor family as well.


Assuntos
Arrestinas/metabolismo , Endocitose , Proteoglicanas/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Arrestinas/genética , Linhagem Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Regulação para Baixo , Humanos , Queratinócitos/metabolismo , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Mutagênese , Fosforilação , Proteínas Serina-Treonina Quinases , Estrutura Terciária de Proteína , Proteoglicanas/química , Proteoglicanas/genética , RNA Interferente Pequeno , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/química , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Fator de Crescimento Transformador beta , Fator de Crescimento Transformador beta1 , beta-Arrestina 2 , beta-Arrestinas
18.
Expert Opin Biol Ther ; 3(2): 251-61, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12662140

RESUMO

Cancers have developed numerous mechanisms for escaping the immune response, either by successfully evading a fully functional immune system or by actively suppressing the immune system so that they are no longer recognised or effectively eliminated. Current evidence supports active cancer cell-mediated immunosuppression via the secretion of the potent immunosuppressive cytokine, transforming growth factor-beta (TGF-beta), as the most general and potent mechanism for human cancer cells to escape the immune system. Efforts to bypass TGF-beta-mediated immunosuppression thereby represent an attractive therapeutic strategy for the chemoprevention and treatment of human cancers, both by directly increasing the efficacy of immunosurveillance and by increasing the efficacy of current immunotherapy strategies. Current approaches are limited by their nonspecific effects on the TGF-beta signalling pathway, as TGF-beta pathways which specifically mediate immunosuppression have not yet been defined. Future efforts should be directed towards elucidating specific TGF-beta pathways so that these can be targeted for the chemoprevention and treatment of human cancers.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Imunoterapia , Neoplasias/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/fisiologia , Animais , Humanos , Neoplasias/fisiopatologia , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...