Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Br J Haematol ; 162(2): 210-20, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23647456

RESUMEN

Multiple myeloma (MM) is an incurable neoplasm caused by proliferation of malignant plasma cells in the bone marrow (BM). MM is characterized frequently by a complete or partial deletion of chromosome 13q14, seen in more than 50% of patients at diagnosis. Within this deleted region the tripartite motif containing 13 (TRIM13, also termed RFP2) gene product has been proposed to be a tumour suppressor gene (TSG). Here, we show that low expression levels of TRIM13 in MM are associated with chromosome 13q deletion and poor clinical outcome. We present a functional analysis of TRIM13 using a loss-of-function approach, and demonstrate that TRIM13 downregulation decreases tumour cell survival as well as cell cycle progression and proliferation of MM cells. In addition, we provide evidence for the involvement of TRIM13 downregulation in inhibiting the NF kappa B pathway and the activity of the 20S proteasome. Although this data does not support a role of TRIM13 as a TSG, it substantiates important roles of TRIM13 in MM tumour survival and proliferation, underscoring its potential role as a novel target for therapeutic intervention.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Mieloma Múltiple/genética , Mieloma Múltiple/patología , FN-kappa B/antagonistas & inhibidores , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Apoptosis/genética , Ciclo Celular/genética , División Celular/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Deleción Cromosómica , Cromosomas Humanos Par 13 , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Mieloma Múltiple/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
2.
PLoS One ; 8(1): e53015, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23308131

RESUMEN

Colon cancer is a deadly disease affecting millions of people worldwide. Current treatment challenges include management of disease burden as well as improvements in detection and targeting of tumor cells. To identify disease state-specific surface antigen signatures, we combined fluorescent cell barcoding with high-throughput flow cytometric profiling of primary and metastatic colon cancer lines (SW480, SW620, and HCT116). Our multiplexed technique offers improvements over conventional methods by permitting the simultaneous and rapid screening of cancer cells with reduced effort and cost. The method uses a protein-level analysis with commercially available antibodies on live cells with intact epitopes to detect potential tumor-specific targets that can be further investigated for their clinical utility. Multiplexed antibody arrays can easily be applied to other tumor types or pathologies for discovery-based approaches to target identification.


Asunto(s)
Antígenos/análisis , Biomarcadores de Tumor/análisis , Colon/patología , Neoplasias del Colon/patología , Citometría de Flujo/métodos , Metástasis de la Neoplasia/patología , Análisis por Matrices de Proteínas/métodos , Línea Celular Tumoral , Biología Computacional/métodos , Biología Computacional/organización & administración , Técnica del Anticuerpo Fluorescente/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Inmunohistoquímica/métodos , Células Tumorales Cultivadas
3.
Blood ; 120(9): 1877-87, 2012 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-22689860

RESUMEN

Bruton tyrosine kinase (Btk) has a well-defined role in B-cell development, whereas its expression in osteoclasts (OCs) further suggests a role in osteoclastogenesis. Here we investigated effects of PCI-32765, an oral and selective Btk inhibitor, on osteoclastogenesis as well as on multiple myeloma (MM) growth within the BM microenvironment. PCI-32765 blocked RANKL/M-CSF-induced phosphorylation of Btk and downstream PLC-γ2 in OCs, resulting in diminished TRAP5b (ED50 = 17 nM) and bone resorption activity. PCI-32765 also inhibited secretion of multiple cytokines and chemokines from OC and BM stromal cell cultures from both normal donors (ED50 = 0.5 nM) and MM patients. It decreased SDF-1-induced migration of MM cells, and down-regulated MIP1-α/CCL3 in MM cells. It also blocked MM cell growth and survival triggered by IL-6 or coculture with BM stromal cells or OCs in vitro. Importantly, PCI-32765 treatment significantly inhibits in vivo MM cell growth (P < .03) and MM cell-induced osteolysis of implanted human bone chips in SCID mice. Moreover, PCI-32765 prevents in vitro colony formation by stem-like cells from MM patients. Together, these results delineate functional sequelae of Btk activation mediating osteolysis and growth of MM cells, supporting evaluation of PCI-32765 as a novel therapeutic in MM.


Asunto(s)
Médula Ósea/efectos de los fármacos , Mieloma Múltiple/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Pirimidinas/farmacología , Adenina/análogos & derivados , Agammaglobulinemia Tirosina Quinasa , Animales , Médula Ósea/metabolismo , Médula Ósea/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimiocinas/metabolismo , Técnicas de Cocultivo , Citocinas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Humanos , Immunoblotting , Ratones , Ratones SCID , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteólisis/genética , Osteólisis/metabolismo , Osteólisis/prevención & control , Piperidinas , Proteínas Tirosina Quinasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
4.
Blood ; 2010 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-20962322

RESUMEN

Multiple myeloma is characterized by frequent chromosomal alterations. Deletion of chr 13, especially band 13q14, is commonly observed in early stages of MM, suggesting the presence of tumor suppressor genes within this region. Here, we functionally validate the role of the microRNAs-15a/16-1 cluster, centered at the deleted region, as TSGs and delineate their downstream target genes in MM. Using "sponge" lentiviral vectors to competitive stably inhibit mature microRNAs in vitro and in vivo, we have documented enhanced proliferative and invasive capacity of cells with stably inhibition of miR-16. Importantly, miR-16 inhibition decreased animal survival in a xenograft model of MM by increasing tumor load and host angiogenesis. Expression profiling analysis of miR-16-deficient cells identified a large number of downstream target genes including FGFR1, PI3KCa, MDM4, VEGFa, as well as secondary affected genes such as JUN and Jag1. We validated designated genes showing binding sites within the conserved 3'-UTR and also within the mRNA coding region as direct miR-16 targets, thus indicating that the miRNAs may have many more targets than anticipated by conventional prediction methods. This loss-of-function system, which mimics the 13q chromosomal deletion, provides a valuable tool to investigate their function in MM pathogenesis and their potential use as therapeutic targets.

5.
Blood ; 115(18): 3772-5, 2010 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-20228272

RESUMEN

Constitutive B-cell lymphoma 6 (Bcl-6) expression was undetectable in multiple myeloma (MM) cell lines, except U266 cells. However, it was up-regulated by coculture with bone marrow (BM) stromal cell-culture supernatant (SCCS). Bcl-6 expression in patient MM cells in the BM was positive. Anti-interleukin-6 (IL-6)-neutralizing antibody significantly blocked SCCS-induced Bcl-6 in MM cells. Indeed, IL-6 strongly triggered Bcl-6 expression in MM cells, whereas Janus kinase inhibitor and STAT3 siRNA down-regulated Bcl-6. Tumor necrosis factor-alpha (TNF-alpha) also triggered Bcl-6, but independently of STAT3, whereas IkappaB kinasebeta inhibitor down-regulated TNF-alpha-induced Bcl-6, indicating that the canonical nuclear factor-kappaB pathway mediates TNF-alpha-induced Bcl-6 expression. Importantly, down-regulation of Bcl-6 by shRNA significantly inhibited MM cell growth in the presence of SCCS. Our results therefore suggest that Bcl-6 expression in MM cells is modulated, at least in part, via Janus kinase/STAT3 and canonical nuclear factor-kappaB pathways and that targeting Bcl-6, either directly or via these cascades, inhibits MM cell growth in the BM milieu.


Asunto(s)
Células de la Médula Ósea/metabolismo , Médula Ósea/metabolismo , Proteínas de Unión al ADN/metabolismo , Mieloma Múltiple/metabolismo , Anticuerpos Neutralizantes/farmacología , Western Blotting , Proliferación Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Humanos , Interleucina-6/farmacología , Quinasas Janus/genética , Quinasas Janus/metabolismo , Mieloma Múltiple/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-bcl-6 , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Regulación hacia Arriba
6.
Cancer Res ; 69(19): 7577-86, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19738061

RESUMEN

Several components of the Wnt signaling cascade have been shown to function either as tumor suppressor proteins or as oncogenes in multiple human cancers, underscoring the relevance of this pathway in oncogenesis and the need for further investigation of Wnt signaling components as potential targets for cancer therapy. Here, using expression profiling analysis as well as in vitro and in vivo functional studies, we show that the Wnt pathway component BCL9 is a novel oncogene that is aberrantly expressed in human multiple myeloma as well as colon carcinoma. We show that BCL9 enhances beta-catenin-mediated transcriptional activity regardless of the mutational status of the Wnt signaling components and increases cell proliferation, migration, invasion, and the metastatic potential of tumor cells by promoting loss of epithelial and gain of mesenchymal-like phenotype. Most importantly, BCL9 knockdown significantly increased the survival of xenograft mouse models of cancer by reducing tumor load, metastasis, and host angiogenesis through down-regulation of c-Myc, cyclin D1, CD44, and vascular endothelial growth factor expression by tumor cells. Together, these findings suggest that deregulation of BCL9 is an important contributing factor to tumor progression. The pleiotropic roles of BCL9 reported in this study underscore its value as a drug target for therapeutic intervention in several malignancies associated with aberrant Wnt signaling.


Asunto(s)
Neoplasias del Colon/metabolismo , Mieloma Múltiple/metabolismo , Proteínas de Neoplasias/biosíntesis , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Progresión de la Enfermedad , Humanos , Receptores de Hialuranos/biosíntesis , Receptores de Hialuranos/genética , Mieloma Múltiple/irrigación sanguínea , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética , Proteínas Wnt/metabolismo
7.
Blood ; 114(13): 2699-708, 2009 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-19652203

RESUMEN

Multiple myeloma (MM) is a cancer of plasma cells with complex molecular characteristics that evolves from monoclonal gammopathy of undetermined significance, a highly prevalent premalignant condition. MM is the second most frequent hematologic cancer in the United States, and it remains incurable, thereby highlighting the need for new therapeutic approaches, particularly those targeting common molecular pathways involved in disease progression and maintenance, shared across different MM subtypes. Here we report that Wnt/beta-catenin is one such pathway. We document the involvement of beta-catenin in cell-cycle regulation, proliferation, and invasion contributing to enhanced proliferative and metastatic properties of MM. The pleiotropic effects of beta-catenin in MM correlate with its transcriptional function, and we demonstrate regulation of a novel target gene, Aurora kinase A, implicating beta-catenin in G2/M regulation. beta-catenin and Aurora kinase A are present in most MM but not in normal plasma cells and are expressed in a pattern that parallels progression from monoclonal gammopathy of undetermined significance to MM. Our data provide evidence for a novel functional link between beta-catenin and Aurora kinase A, underscoring a critical role of these pathways in MM disease progression.


Asunto(s)
Mieloma Múltiple/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Wnt/fisiología , beta Catenina/fisiología , Animales , Aurora Quinasa A , Aurora Quinasas , Ciclo Celular/genética , Proliferación Celular , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Mieloma Múltiple/patología , Proteínas Serina-Treonina Quinasas/fisiología , Trasplante Heterólogo , Células Tumorales Cultivadas , Proteínas Wnt/genética , beta Catenina/genética
8.
Cancer Res ; 68(21): 8889-98, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18974133

RESUMEN

The pathogenesis of malignant melanoma involves the interplay of tumor cells with normal host elements, but the underlying mechanisms are incompletely understood. Here, we show that milk fat globule EGF-8 (MFG-E8), a secreted protein expressed at high levels in the vertical growth phase of melanoma, promotes disease progression through coordinated alpha(v)beta(3) integrin signaling in the tumor microenvironment. In a murine model of melanoma, MFG-E8 enhanced tumorigenicity and metastatic capacity through Akt-dependent and Twist-dependent pathways. MFG-E8 augmented melanoma cell resistance to apoptosis, triggered an epithelial-to-mesenchymal transition (EMT), and stimulated invasion and immune suppression. In human melanoma cells, MFG-E8 knockdown attenuated Akt and Twist signaling and thereby compromised tumor cell survival, EMT, and invasive ability. MFG-E8-deficient human melanoma cells also showed increased sensitivity to small molecule inhibitors of insulin-like growth factor I receptor and c-Met. Together, these findings delineate pleiotropic roles for MFG-E8 in the tumor microenvironment and raise the possibility that systemic MFG-E8 blockade might prove therapeutic for melanoma patients.


Asunto(s)
Antígenos de Superficie/fisiología , Melanoma/patología , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología , Proteína 1 Relacionada con Twist/metabolismo , Animales , Apoptosis , Secuencia de Bases , Cartilla de ADN , Progresión de la Enfermedad , Humanos , Melanoma/enzimología , Melanoma/metabolismo , Ratones , Microscopía Fluorescente , Proteínas de la Leche , Invasividad Neoplásica
9.
Blood ; 111(10): 5068-77, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18334673

RESUMEN

The nuclear factor-kappaB (NF-kappaB) path-way has been implicated in tumor B-cell survival, growth, and resistance to therapy. Because tumor cells overcome single-agent antitumor activity, we hypothesized that combination of agents that target differentially NF-kappaB pathway will induce significant cytotoxicity. Therapeutic agents that target proteasome and Akt pathways should induce significant activity in B-cell malignancies as both pathways impact NF-kappaB activity. We demonstrated that perifosine and bortezomib both targeted NF-kappaB through its recruitment to the promoter of its target gene IkappaB using chromatin immunoprecipitation assay. This combination led to synergistic cytotoxicity in Waldenstrom macroglobulinemia (WM) cells that was mediated through a combined reduction of the PI3K/Akt and ERK signaling pathways, found to be critical for survival of WM cells. Moreover, a combination of these drugs with the CD20 monoclonal antibody rituximab further increased their cytotoxic activity. Thus, effective WM therapy may require combination regimens targeting the NF-kappaB pathway.


Asunto(s)
FN-kappa B/antagonistas & inhibidores , Macroglobulinemia de Waldenström/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Ácidos Borónicos/farmacología , Bortezomib , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , FN-kappa B/metabolismo , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacología , Pirazinas/farmacología , Transducción de Señal/efectos de los fármacos , Macroglobulinemia de Waldenström/patología
10.
Hematol Oncol Clin North Am ; 21(6): 1051-69, viii, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17996588

RESUMEN

Multiple myeloma (MM) remains incurable despite high-dose chemotherapy with stem cell support. There is need, therefore, for continuous efforts directed toward the development of novel rational-based therapeutics for MM, which requires a detailed knowledge of the mutations driving this malignancy. In improving the success rate of effective drug development, it is equally imperative that biologic systems be developed to better validate these target genes. Here we review the recent developments in the generation of mouse models of MM and their impact as preclinical models for designing and assessing target-based therapeutic approaches.


Asunto(s)
Quimera/fisiología , Modelos Animales de Enfermedad , Mieloma Múltiple/fisiopatología , Animales , Trasplante Óseo , Quimera/inmunología , Humanos , Ratones , Ratones SCID , Ratones Transgénicos , Trasplante Heterólogo/inmunología , Trasplante Heterólogo/métodos
11.
J Clin Invest ; 117(7): 1902-13, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17557120

RESUMEN

Granulocyte-macrophage colony-stimulating factor (GM-CSF) enhances protection against tumors and infections, but GM-CSF-deficient mice develop inflammatory disease. Here we show that GM-CSF is required for the expression of milk fat globule EGF 8 (MFG-E8) in antigen-presenting cells, and that MFG-E8-mediated uptake of apoptotic cells is a key determinant of GM-CSF-triggered tolerance and immunity. Upon exposure to apoptotic cells, GM-CSF-deficient antigen-presenting cells (APCs) produce an altered cytokine profile that results in decreased Tregs and increased Th1 cells, whereas concurrent ablation of IFN-gamma promotes Th17 cells. In wild-type mice, MFG-E8 attenuates the vaccination activity of GM-CSF-secreting tumor cells through Treg induction, whereas a dominant-negative MFG-E8 mutant potentiates GM-CSF-stimulated tumor destruction through Treg inhibition. These findings clarify the immunoregulatory effects of apoptotic cells and suggest new therapeutic strategies to modulate CD4(+) T cell subsets in cancer and autoimmunity.


Asunto(s)
Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/metabolismo , Antígenos de Superficie/metabolismo , Apoptosis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Proteínas de la Leche/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Vacunas contra el Cáncer , Diferenciación Celular , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Homeostasis , Inmunoterapia , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Fagocitosis
12.
Cancer Res ; 67(10): 4732-41, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17510401

RESUMEN

Cell cycle progression from G(1) to S phase depends on phosphorylation of pRb by complexes containing a cyclin (D type or E type) and cyclin-dependent kinase (e.g., cdk2, cdk4, or cdk6). Ink4 proteins function to oppose the action of cdk4/6-cyclin D complexes by inhibiting cdk4/6. We employed genetic and pharmacologic approaches to study the interplay among Ink4 proteins and cdk4/6 activity in vivo. Mouse embryo fibroblasts (MEF) lacking p16(Ink4a) and p18(Ink4c) showed similar growth kinetics as wild-type MEFs despite increased cdk4 activity. In vivo, germline deficiency of p16(Ink4a) and p18(Ink4c) resulted in increased proliferation in the intermediate pituitary and pancreatic islets of adult mice, and survival of p16(Ink4a-/-);p18(Ink4c-/-) mice was significantly reduced due to aggressive pituitary tumors. Compensation among the Ink4 proteins was observed both in vivo in p18(Ink4c-/-) mice and in MEFs from p16(Ink4a-/-), p18(Ink4c-/-), or p16(Ink4a-/-);p18(Ink4c-/-) mice. Treatment with PD 0332991, a specific cdk4/6 kinase inhibitor, abrogated proliferation in those compartments where Ink4 deficiency was associated with enhanced proliferation (i.e., islets, pituitary, and B lymphocytes) but had no effect on proliferation in other tissues such as the small bowel. These data suggest that p16(Ink4a) and p18(Ink4c) coordinately regulate the in vivo catalytic activity of cdk4/6 in specific compartments of adult mice.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/deficiencia , Neoplasias Hipofisarias/enzimología , Animales , Procesos de Crecimiento Celular/fisiología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Embrión de Mamíferos , Fibroblastos/citología , Fibroblastos/enzimología , Fibroblastos/fisiología , Ratones , Ratones Transgénicos , Piperazinas/farmacología , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Piridinas/farmacología
13.
Proc Natl Acad Sci U S A ; 104(18): 7516-21, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17452641

RESUMEN

Multiple myeloma (MM) is an invariably fatal form of cancer characterized by clonal proliferation of malignant plasma cells in the bone marrow. The canonical Wnt signaling pathway is activated in MM cells through constitutively active beta-catenin, a messenger molecule relevant to growth, survival, and migration of MM cells. The identification of a number of small molecular compounds, such as PKF115-584, which disrupt the interaction of the transcriptionally active beta-catenin/TCF protein complex, provides valuable new therapeutic tools to target an alternative pathway in MM independent of the proteasome. Here we evaluated the transcriptional, proteomic, signaling changes, and biological sequelae associated with the inhibition of Wnt signaling in MM by PKF115-584. The compound blocks expression of Wnt target genes and induces cytotoxicity in both patient MM cells and MM cell lines without a significant effect in normal plasma cells. In xenograft models of human MM, PKF115-584 inhibits tumor growth and prolongs survival. Taken together, these data demonstrate the efficacy of disrupting the beta-catenin/TCF transcriptional complex to exploit tumor dependence on Wnt signaling as a therapeutic approach in the treatment of MM.


Asunto(s)
Mieloma Múltiple/metabolismo , Factores de Transcripción TCF/metabolismo , Transcripción Genética/genética , beta Catenina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-6/farmacología , Ratones , Ratones SCID , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Perileno/análogos & derivados , Perileno/farmacología , Unión Proteica , Transducción de Señal , Tasa de Supervivencia , Transcripción Genética/efectos de los fármacos , Trasplante Heterólogo , Proteínas Wnt/metabolismo , beta Catenina/genética
14.
Cancer Cell ; 11(4): 349-60, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17418411

RESUMEN

Multiple myeloma (MM) evolves from a highly prevalent premalignant condition termed MGUS. The factors underlying the malignant transformation of MGUS are unknown. We report a MGUS/MM phenotype in transgenic mice with Emu-directed expression of the XBP-1 spliced isoform (XBP-1s), a factor governing unfolded protein/ER stress response and plasma-cell development. Emu-XBP-1s elicited elevated serum Ig and skin alterations. With age, Emu-xbp-1s transgenics develop features diagnostic of human MM, including bone lytic lesions and subendothelial Ig deposition. Furthermore, transcriptional profiles of Emu-xbp-1s lymphoid and MM cells show aberrant expression of known human MM dysregulated genes. The similarities of this model with the human disease, coupled with documented frequent XBP-1s overexpression in human MM, serve to implicate XBP-1s dysregulation in MM pathogenesis.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Retículo Endoplásmico/patología , Mieloma Múltiple/patología , Proteínas Nucleares/metabolismo , Células Plasmáticas/citología , Envejecimiento/patología , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Enfermedades Óseas/patología , Células Cultivadas , Proteínas de Unión al ADN/genética , Dromaiidae/genética , Ensayo de Cambio de Movilidad Electroforética , Retículo Endoplásmico/metabolismo , Femenino , Humanos , Hipergammaglobulinemia/patología , Enfermedades Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mieloma Múltiple/metabolismo , Proteínas Nucleares/genética , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Empalme del ARN , Factores de Transcripción del Factor Regulador X , Enfermedades de la Piel/patología , Factores de Transcripción , Transcripción Genética , Proteína 1 de Unión a la X-Box
15.
Proc Natl Acad Sci U S A ; 104(10): 3931-6, 2007 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-17360455

RESUMEN

The Rb/p16(Ink4a) and p53/p19Arf tumor suppressor pathways have been linked to diverse cancer-relevant processes, including those governing the cellular responses to telomere dysfunction. In this study, we sought to provide direct genetic evidence of a role for the Ink4a/Arf tumor suppressor gene, encoding both p16(Ink4a) and p19(Arf), in modulating the cellular and tissue phenotypes associated with telomere dysfunction by using the mTerc Ink4a/Arf mouse model. In contrast to the rescue associated with p53 deficiency, Ink4a/Arf deficiency did not attenuate the degenerative phenotypes elicited by telomere dysfunction in the late-generation mTerc-/- mice. Furthermore, in contrast to accelerated cancer onset and increased epithelial cancers of late-generation mTerc-/- p53 mutant mice, late-generation mTerc-/- Ink4a/Arf mutant mice experienced a delayed tumor onset and maintained the lymphoma and sarcoma spectrum. Consistent with the negligible role of Ink4a/Arf in the telomere checkpoint response in vivo, late-generation mTerc-/- Ink4a/Arf-/- tissues show activated p53, and derivative tumor cell lines sustain frequent loss of p53 function, whereas all early generation mTerc Ink4a/Arf-/- tumor cell lines remain intact for p53. In addition, the late-generation mTerc-/- Ink4a/Arf-/- tumors showed activation of the alternative lengthening of telomere mechanism, underscoring the need for adaptation to the presence of telomere dysfunction in the absence of p16(Ink4a) and p19(Arf). These observations highlight the importance of genetic context in dictating whether telomere dysfunction promotes or suppresses age-related degenerative conditions as well as the rate of initiation and type of spontaneous cancers.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Genes Supresores de Tumor , Telomerasa/genética , Telomerasa/fisiología , Células 3T3 , Animales , Genes p53 , Hibridación Fluorescente in Situ , Ratones , Ratones Transgénicos , Neoplasias/metabolismo , Neoplasias Glandulares y Epiteliales/genética , Fenotipo , Telomerasa/deficiencia , Telómero/metabolismo , Telómero/ultraestructura , Factores de Tiempo , Proteína p53 Supresora de Tumor/metabolismo
16.
Cell ; 128(2): 309-23, 2007 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-17254969

RESUMEN

Activated phosphoinositide 3-kinase (PI3K)-AKT signaling appears to be an obligate event in the development of cancer. The highly related members of the mammalian FoxO transcription factor family, FoxO1, FoxO3, and FoxO4, represent one of several effector arms of PI3K-AKT signaling, prompting genetic analysis of the role of FoxOs in the neoplastic phenotypes linked to PI3K-AKT activation. While germline or somatic deletion of up to five FoxO alleles produced remarkably modest neoplastic phenotypes, broad somatic deletion of all FoxOs engendered a progressive cancer-prone condition characterized by thymic lymphomas and hemangiomas, demonstrating that the mammalian FoxOs are indeed bona fide tumor suppressors. Transcriptome and promoter analyses of differentially affected endothelium identified direct FoxO targets and revealed that FoxO regulation of these targets in vivo is highly context-specific, even in the same cell type. Functional studies validated Sprouty2 and PBX1, among others, as FoxO-regulated mediators of endothelial cell morphogenesis and vascular homeostasis.


Asunto(s)
Linaje de la Célula/genética , Transformación Celular Neoplásica/genética , Células Endoteliales/metabolismo , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica/genética , Proteínas Supresoras de Tumor/genética , Animales , Proteínas de Ciclo Celular , Diferenciación Celular/genética , Transformación Celular Neoplásica/metabolismo , Proteínas de Drosophila , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Hemangioma/genética , Hemangioma/metabolismo , Proteínas de Homeodominio/genética , Homeostasis/genética , Linfoma/genética , Linfoma/metabolismo , Ratones , Ratones Noqueados , Neovascularización Fisiológica/genética , Proteínas del Tejido Nervioso/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/metabolismo
17.
Cancer Cell ; 9(5): 379-90, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16697958

RESUMEN

Histiocytic sarcoma (HS) is a rare malignant proliferation of histiocytes of uncertain molecular pathogenesis. Here, genetic analysis of coincident loss of Pten and Ink4a/Arf tumor suppressors in the mouse revealed a neoplastic phenotype dominated by a premalignant expansion of biphenotypic myelolymphoid cells followed by the development of HS. Pten protein loss occurred only in the histiocytic portion of tumors, suggesting a stepwise genetic inactivation in the generation of HS. Similarly, human HS showed genetic or epigenetic inactivation of PTEN, p16(INK4A), and p14(ARF), supporting the relevance of this genetically engineered mouse model of HS. These genetic and translational observations establish a cooperative role of Pten and Ink4a/Arf in the development of HS and provide mechanistic insights into the pathogenesis of human HS.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Trastornos Histiocíticos Malignos/patología , Linfocitos/inmunología , Células Mieloides/inmunología , Fosfohidrolasa PTEN/metabolismo , Sarcoma/patología , Proteína p14ARF Supresora de Tumor/metabolismo , Animales , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Trastornos Histiocíticos Malignos/inmunología , Homeostasis , Humanos , Inmunofenotipificación , Metilación , Ratones , Mutación/genética , Fosfohidrolasa PTEN/deficiencia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sarcoma/inmunología , Proteína p14ARF Supresora de Tumor/deficiencia
18.
Cancer Cell ; 9(4): 313-25, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16616336

RESUMEN

To identify genetic events underlying the genesis and progression of multiple myeloma (MM), we conducted a high-resolution analysis of recurrent copy number alterations (CNAs) and expression profiles in a collection of MM cell lines and outcome-annotated clinical specimens. Attesting to the molecular heterogeneity of MM, unsupervised classification using nonnegative matrix factorization (NMF) designed for array comparative genomic hybridization (aCGH) analysis uncovered distinct genomic subtypes. Additionally, we defined 87 discrete minimal common regions (MCRs) within recurrent and highly focal CNAs. Further integration with expression data generated a refined list of MM gene candidates residing within these MCRs, thereby providing a genomic framework for dissection of disease pathogenesis, improved clinical management, and initiation of targeted drug discovery for specific MM patients.


Asunto(s)
Genoma Humano/genética , Genómica , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Cromosomas Humanos/clasificación , Cromosomas Humanos/genética , Diploidia , Supervivencia sin Enfermedad , Dosificación de Gen , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Mieloma Múltiple/clasificación , Mieloma Múltiple/diagnóstico , Pronóstico
19.
Blood ; 106(2): 713-6, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15817674

RESUMEN

We developed a novel in vivo multiple myeloma (MM) model by engrafting the interleukin 6 (IL-6)-dependent human MM cell line INA-6 into severe combined immunodeficiency (SCID) mice previously given implants of a human fetal bone chip (SCID-hu mice). INA-6 cells require either exogenous human IL-6 (huIL-6) or bone marrow stromal cells (BMSCs) to proliferate in vitro. In this model, we monitored the in vivo growth of INA-6 cells stably transduced with a green fluorescent protein (GFP) gene (INA-6GFP+ cells). INA-6 MM cells engrafted in SCID-hu mice but not in SCID mice that had not been given implants of human fetal bone. The level of soluble human IL-6 receptor (shuIL-6R) in murine serum and fluorescence imaging of host animals were sensitive indicators of tumor growth. Dexamethasone as well as experimental drugs, such as Atiprimod and B-B4-DM1, were used to confirm the utility of the model for evaluation of anti-MM agents. We report that this model is highly reproducible and allows for evaluation of investigational drugs targeting IL-6-dependent MM cells in the human bone marrow (huBM) milieu.


Asunto(s)
Mieloma Múltiple/patología , Animales , Trasplante Óseo , Línea Celular Tumoral , Dexametasona/farmacología , Modelos Animales de Enfermedad , Trasplante de Tejido Fetal , Proteínas Fluorescentes Verdes/genética , Humanos , Inmunotoxinas/farmacología , Ratones , Ratones SCID , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/inmunología , Trasplante de Neoplasias , Receptores de Interleucina-6/sangre , Proteínas Recombinantes/genética , Compuestos de Espiro/farmacología , Transducción Genética , Trasplante Heterólogo
20.
Blood ; 104(12): 3688-96, 2004 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-15292058

RESUMEN

We tested the in vitro and in vivo antitumor activity of the maytansinoid DM1 (N(2')-deacetyl-N(2')-(3-mercapto-1-oxopropyl)-maytansine), a potent antimicrotubule agent, covalently linked to the murine monoclonal antibody (mAb) B-B4 targeting syndecan-1 (CD138). We evaluated the in vitro activity of B-B4-DM1 against a panel of CD138(+) and CD138(-) cell lines, as well as CD138(+) patient multiple myeloma (MM) cells. Treatment with B-B4-DM1 selectively decreased growth and survival of MM cell lines, patient MM cells, and MM cells adherent to bone marrow stromal cells. We further examined the activity of B-B4-DM1 in 3 human MM models in mice: (1) severe combined immunodeficient (SCID) mice bearing subcutaneous xenografts; (2) SCID mice bearing green fluorescent protein-positive (GFP(+)) xenografts; and (3) SCID mice implanted with human fetal bone (SCID-hu) and subsequently injected with patient MM cells. Tumor regression and inhibition of tumor growth, improvement in overall survival, and reduction in levels of circulating human paraprotein were observed in mice treated with B-B4-DM1. Although immunohistochemical analysis demonstrates restricted CD138 expression in human tissues, the lack of B-B4 reactivity with mouse tissues precludes evaluation of its toxicity in these models. In conclusion, B-B4-DM1 is a potent anti-MM agent that kills cells in an antigen-dependent manner in vitro and mediates in vivo antitumor activity at doses that are well tolerated, providing the rationale for clinical trials of this immunoconjugate in MM.


Asunto(s)
Inmunoconjugados/farmacología , Maitansina/farmacología , Glicoproteínas de Membrana , Mieloma Múltiple/tratamiento farmacológico , Proteoglicanos , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Células de la Médula Ósea/patología , Estudios de Casos y Controles , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Inmunoconjugados/uso terapéutico , Masculino , Maitansina/uso terapéutico , Ratones , Ratones SCID , Mieloma Múltiple/patología , Trasplante de Neoplasias , Tasa de Supervivencia , Sindecano-1 , Sindecanos , Trasplante Heterólogo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...