Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
JAMA Dermatol ; 152(4): 452-6, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26765315

RESUMEN

IMPORTANCE: Tumor resistance is an emerging problem for Smoothened (SMO) inhibitor-treated metastatic basal cell carcinoma (BCC). Arsenic trioxide and itraconazole antagonize the hedgehog (HH) pathway at sites distinct from those treated by SMO inhibitors. OBJECTIVE: To determine whether administration of intravenous arsenic trioxide and oral itraconazole in patients with metastatic BCC is associated with a reduction in GLI1 messenger RNA expression in tumor and/or normal skin biopsy samples. DESIGN, SETTING, AND PARTICIPANTS: Five men with metastatic BCC who experienced relapse after SMO inhibitor treatment underwent intravenous arsenic trioxide treatment for 5 days, every 28 days, and oral itraconazole treatment on days 6 to 28. Data were collected from April 10 to November 14, 2013. Follow-up was completed on October 3, 2015, and data were analyzed from June 5 to October 6, 2015. MAIN OUTCOMES AND MEASURES: The primary outcome was the change in messenger RNA levels of the GLI family zinc finger 1 (GLI1) gene (HH-pathway target gene) in biopsy specimens of normal skin or BCC before and after treatment. Secondary objectives were evaluation of tumor response and tolerability. RESULTS: Of the 5 patients (mean [SD] age, 52 [9] years; age range, 43-62 years), 3 completed 3 cycles of treatment and 2 discontinued treatment early owing to disease progression or adverse events. Adverse effects included grade 2 transaminitis and grade 4 leukopenia with a grade 3 infection. Overall, arsenic trioxide and itraconazole reduced GLI1 messenger RNA levels by 75% from baseline (P < .001). The best overall response after 3 treatment cycles was stable disease in 3 patients. CONCLUSIONS AND RELEVANCE: Targeting the HH pathway with sequential arsenic trioxide and itraconazole treatment is a feasible treatment for metastatic BCC. Although some patients experienced stable disease for 3 months, none had tumor shrinkage, which may be owing to transient GLI1 suppression with sequential dosing. Continuous dosing may be required to fully inhibit the HH pathway and achieve clinical response.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma Basocelular/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Factores de Transcripción/genética , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Trióxido de Arsénico , Arsenicales/administración & dosificación , Carcinoma Basocelular/patología , Resistencia a Antineoplásicos , Estudios de Seguimiento , Proteínas Hedgehog/antagonistas & inhibidores , Humanos , Itraconazol/administración & dosificación , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Óxidos/administración & dosificación , ARN Mensajero/metabolismo , Neoplasias Cutáneas/patología , Resultado del Tratamiento , Proteína con Dedos de Zinc GLI1
2.
Sci Signal ; 8(379): ra55, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-26038600

RESUMEN

Defects in the biogenesis of or transport through primary cilia affect Hedgehog protein signaling, and many Hedgehog pathway components traffic through or accumulate in cilia. The Hedgehog receptor Patched negatively regulates the activity and ciliary accumulation of Smoothened, a seven-transmembrane protein that is essential for transducing the Hedgehog signal. We found that this negative regulation of Smoothened required the ciliary localization of Patched, as specified either by its own cytoplasmic tail or by provision of heterologous ciliary localization signals. Surprisingly, given that Hedgehog binding promotes the exit of Patched from the cilium, we observed that an altered form of Patched that is retained in the cilium nevertheless responded to Hedgehog, resulting in Smoothened activation. Our results indicate that whereas ciliary localization of Patched is essential for suppression of Smoothened activation, the primary event enabling Smoothened activation is binding of Hedgehog to Patched, and Patched ciliary removal is secondary.


Asunto(s)
Receptores de Superficie Celular/metabolismo , Transducción de Señal/fisiología , Animales , Cilios/genética , Cilios/metabolismo , Células HEK293 , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Ratones Noqueados , Receptores Patched , Transporte de Proteínas/fisiología , Receptores de Superficie Celular/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened
3.
PLoS One ; 9(8): e104070, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25119726

RESUMEN

Ciliary accumulation of signaling proteins must result from a rate of ciliary entry that exceeds ciliary exit, but approaches for distinguishing ciliary entry vs. exit are lacking. Using a photoconvertible fluorescent protein tag, we establish an assay that allows a separate but simultaneous examination of ciliary entry and exit of the Hedgehog signaling protein Smoothened in individual cells. We show that KAAD-cyclopamine selectively blocks entry, whereas ciliobrevin interferes initially with exit and eventually with both entry and exit of ciliary Smoothened. Our study provides an approach to understanding regulation of ciliary entry vs. exit of Hedgehog signaling components as well as other ciliary proteins.


Asunto(s)
Cilios/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Proteínas Luminiscentes/análisis , Ratones , Microscopía Fluorescente/métodos , Células 3T3 NIH , Transporte de Proteínas , Receptores Acoplados a Proteínas G/análisis , Proteínas Recombinantes de Fusión/análisis , Transducción de Señal , Receptor Smoothened
4.
Cancer Cell ; 23(1): 23-34, 2013 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-23291299

RESUMEN

Recognition of the multiple roles of Hedgehog signaling in cancer has prompted intensive efforts to develop targeted pathway inhibitors. Leading inhibitors in clinical development act by binding to a common site within Smoothened, a critical pathway component. Acquired Smoothened mutations, including SMO(D477G), confer resistance to these inhibitors. Here, we report that itraconazole and arsenic trioxide, two agents in clinical use that inhibit Hedgehog signaling by mechanisms distinct from that of current Smoothened antagonists, retain inhibitory activity in vitro in the context of all reported resistance-conferring Smoothened mutants and GLI2 overexpression. Itraconazole and arsenic trioxide, alone or in combination, inhibit the growth of medulloblastoma and basal cell carcinoma in vivo, and prolong survival of mice with intracranial drug-resistant SMO(D477G) medulloblastoma.


Asunto(s)
Antineoplásicos/uso terapéutico , Arsenicales/uso terapéutico , Carcinoma Basocelular/tratamiento farmacológico , Proteínas Hedgehog/fisiología , Itraconazol/uso terapéutico , Meduloblastoma/tratamiento farmacológico , Óxidos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Anilidas/farmacología , Anilidas/uso terapéutico , Animales , Antineoplásicos/farmacología , Trióxido de Arsénico , Arsenicales/farmacología , Carcinoma Basocelular/metabolismo , Carcinoma Basocelular/patología , Resistencia a Antineoplásicos , Proteínas Hedgehog/metabolismo , Itraconazol/farmacología , Meduloblastoma/metabolismo , Meduloblastoma/patología , Ratones , Óxidos/farmacología , Piridinas/farmacología , Piridinas/uso terapéutico , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptor Smoothened
5.
Proc Natl Acad Sci U S A ; 107(30): 13432-7, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20624968

RESUMEN

Aberrant Hedgehog (Hh) pathway activation has been implicated in cancers of diverse tissues and organs, and the tumor growth-inhibiting effects of pathway antagonists in animal models have stimulated efforts to develop pathway antagonists for human therapeutic purposes. These efforts have focused largely on cyclopamine derivatives or other compounds that mimic cyclopamine action in binding to and antagonizing Smoothened, a membrane transductory component. We report here that arsenicals, in contrast, antagonize the Hh pathway by targeting Gli transcriptional effectors; in the short term, arsenic blocks Hh-induced ciliary accumulation of Gli2, the primary activator of Hh-dependent transcription, and with prolonged incubation arsenic reduces steady-state levels of Gli2. Arsenicals active in Hh pathway antagonism include arsenic trioxide (ATO), a curative agent in clinical use for acute promyelocytic leukemia (APL); in our studies, ATO inhibited growth of Hh pathway-driven medulloblastoma allografts derived from Ptch+/-p53-/- mice within a range of serum levels comparable to those achieved in treatment of human APL. Arsenic thus could be tested rapidly as a therapeutic agent in malignant diseases associated with Hh pathway activation and could be particularly useful in such diseases that are inherently resistant or have acquired resistance to cyclopamine mimics.


Asunto(s)
Arsenicales/farmacología , Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Trióxido de Arsénico , Arsenitos/farmacología , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/prevención & control , Relación Dosis-Respuesta a Droga , Proteínas Hedgehog/genética , Factores de Transcripción de Tipo Kruppel/genética , Luciferasas/genética , Luciferasas/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Meduloblastoma/genética , Meduloblastoma/patología , Meduloblastoma/prevención & control , Ratones , Ratones Noqueados , Ratones Desnudos , Células 3T3 NIH , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Óxidos/farmacología , Estabilidad Proteica/efectos de los fármacos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Compuestos de Sodio/farmacología , Transfección , Trasplante Homólogo , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteína Gli2 con Dedos de Zinc
6.
Cancer Cell ; 17(4): 388-99, 2010 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-20385363

RESUMEN

In a screen of drugs previously tested in humans we identified itraconazole, a systemic antifungal, as a potent antagonist of the Hedgehog (Hh) signaling pathway that acts by a mechanism distinct from its inhibitory effect on fungal sterol biosynthesis. Systemically administered itraconazole, like other Hh pathway antagonists, can suppress Hh pathway activity and the growth of medulloblastoma in a mouse allograft model and does so at serum levels comparable to those in patients undergoing antifungal therapy. Mechanistically, itraconazole appears to act on the essential Hh pathway component Smoothened (SMO) by a mechanism distinct from that of cyclopamine and other known SMO antagonists, and prevents the ciliary accumulation of SMO normally caused by Hh stimulation.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas Hedgehog/antagonistas & inhibidores , Itraconazol/uso terapéutico , Transducción de Señal/efectos de los fármacos , Animales , Antifúngicos/farmacología , Antifúngicos/uso terapéutico , Carcinoma Basocelular/tratamiento farmacológico , División Celular/efectos de los fármacos , Ciclodextrinas/farmacología , Proteínas Hedgehog/química , Proteínas Hedgehog/efectos de los fármacos , Proteínas Hedgehog/fisiología , Humanos , Itraconazol/farmacología , Cinética , Lipoproteínas LDL/fisiología , Ratones , Modelos Moleculares
7.
Proc Natl Acad Sci U S A ; 106(51): 21666-71, 2009 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-19996169

RESUMEN

Stimulation by the extracellular Hedgehog (Hh) protein signal has been shown to alter ciliary localization of the mammalian Hh receptor components Smoothened (Smo) and Patched (Ptc), and mutations that disrupt the structure and function of the cilium also disrupt Hh-induced changes in gene expression. But how ciliary events affect gene expression in the nucleus is not known, and to address this question we have characterized the cellular trafficking of Gli2, the principal mediator of Hh-dependent transcriptional activation. From a combination of pharmacological and genetic manipulations we find in resting cells that both Gli2 and Smo appear to shuttle in and out of the cilium, with Gli2 but not Smo requiring intact cytoplasmic microtubules for ciliary entry and both requiring the ciliary retrograde motor, cytoplasmic dynein 2, for ciliary exit. We also find that changes in ciliary and nuclear trafficking of Gli2 are triggered by the Hh-dependent accumulation of activated Smo in the cilium, resulting in a shift from primarily cytoplasmic localization to accumulation at the distal tip of the cilium and within the nucleus. Gli2 thus functions as a dynamic monitor of Smo activity in the cilium and thereby links Hh pathway activation in the cilium to transcriptional activation in the nucleus.


Asunto(s)
Núcleo Celular/metabolismo , Cilios/metabolismo , Proteínas Hedgehog/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Activación Transcripcional , Animales , Ratones , Células 3T3 NIH , Transporte de Proteínas , Receptor Smoothened , Proteína Gli2 con Dedos de Zinc
8.
Nature ; 458(7239): 776-9, 2009 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-19169242

RESUMEN

Although the role of Hedgehog (Hh) signalling in embryonic pattern formation is well established, its functions in adult tissue renewal and maintenance remain unclear, and the relationship of these functions to cancer development has not been determined. Here we show that the loss of Smoothened (Smo), an essential component of the Hh pathway, impairs haematopoietic stem cell renewal and decreases induction of chronic myelogenous leukaemia (CML) by the BCR-ABL1 oncoprotein. Loss of Smo causes depletion of CML stem cells--the cells that propagate the leukaemia--whereas constitutively active Smo augments CML stem cell number and accelerates disease. As a possible mechanism for Smo action, we show that the cell fate determinant Numb, which depletes CML stem cells, is increased in the absence of Smo activity. Furthermore, pharmacological inhibition of Hh signalling impairs not only the propagation of CML driven by wild-type BCR-ABL1, but also the growth of imatinib-resistant mouse and human CML. These data indicate that Hh pathway activity is required for maintenance of normal and neoplastic stem cells of the haematopoietic system and raise the possibility that the drug resistance and disease recurrence associated with imatinib treatment of CML might be avoided by targeting this essential stem cell maintenance pathway.


Asunto(s)
Proteínas Hedgehog/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/fisiopatología , Células Madre Neoplásicas/fisiología , Transducción de Señal , Animales , Antineoplásicos/farmacología , Células Cultivadas , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptor Smoothened , Tomatina/análogos & derivados , Tomatina/farmacología , Alcaloides de Veratrum/farmacología
9.
Proc Natl Acad Sci U S A ; 104(10): 4048-53, 2007 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-17360475

RESUMEN

The cancer stem cell hypothesis suggests that malignant growth depends on a subset of tumor cells with stem cell-like properties of self-renewal. Because hedgehog (Hh) signaling regulates progenitor cell fate in normal development and homeostasis, aberrant pathway activation might be involved in the maintenance of such a population in cancer. Indeed, mutational activation of the Hh pathway is associated with medulloblastoma and basal cell carcinoma; pathway activity is also critical for growth of other tumors lacking such mutations, although the mechanism of pathway activation is poorly understood. Here we study the role and mechanism of Hh pathway activation in multiple myeloma (MM), a malignancy with a well defined stem cell compartment. In this model, rare malignant progenitors capable of clonal expansion resemble B cells, whereas the much larger tumor cell population manifests a differentiated plasma cell phenotype that pathologically defines the disease. We show that the subset of MM cells that manifests Hh pathway activity is markedly concentrated within the tumor stem cell compartment. The Hh ligand promotes expansion of MM stem cells without differentiation, whereas the Hh pathway blockade, while having little or no effect on malignant plasma cell growth, markedly inhibits clonal expansion accompanied by terminal differentiation of purified MM stem cells. These data reveal that Hh pathway activation is heterogeneous across the spectrum of MM tumor stem cells and their more differentiated progeny. The potential existence of similar relationships in other adult cancers may have important biologic and clinical implications for the study of aberrant Hh signaling.


Asunto(s)
Proteínas Hedgehog/fisiología , Mieloma Múltiple/patología , Transducción de Señal , Células Madre/metabolismo , Animales , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Fenotipo , Células Plasmáticas/metabolismo , Sindecano-1/biosíntesis , Alcaloides de Veratrum/farmacología
10.
Blood Cells Mol Dis ; 32(2): 302-8, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15003822

RESUMEN

The c-fps/fes protooncogene encodes a 92-kDa protein tyrosine kinase that is involved in myeloid cell development and immune responses of granulocytes and macrophages. To help define its biological role and mechanism of action, we have developed a gain of function allele of Fes that has potent biological activity in myeloid cells. Introduction of constitutively active Fes into myeloid progenitors induced the appearance of fully differentiated macrophages or granulocytes depending on the lineage commitment of the transduced cells. We found that Fes-induced macrophage differentiation correlated with activation of the ets family transcription factor PU.1, which is essential for macrophage development. On the other hand, granulocyte differentiation by Fes was mediated through activation of CCAAT/enhancer-binding protein alpha (C/EBP-alpha) and STAT3, two transcription factors that are critical for granulocytic differentiation. We postulate that Fes transduces inductive signals for terminal macrophage and granulocyte differentiation, and that this biological activity is mediated through the activation of lineage-specific transcription factors.


Asunto(s)
Proteínas de Fusión gag-onc/fisiología , Células Mieloides/metabolismo , Proteínas Tirosina Quinasas/fisiología , Transducción de Señal , Activación Transcripcional , Activación Enzimática , Proteínas de Fusión gag-onc/genética , Regulación de la Expresión Génica , Granulocitos/citología , Humanos , Imitación Molecular , Monocitos/citología , Células Mieloides/enzimología , Mielopoyesis , Proteínas Tirosina Quinasas/genética , Factores de Transcripción/metabolismo , Transfección , Células U937
11.
J Virol ; 77(10): 5759-73, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12719569

RESUMEN

Human herpesvirus 8 (HHV-8), the etiologic agent of Kaposi's sarcoma (KS), encodes a chemokine receptor homologue, the viral G protein-coupled receptor (vGPCR), that has been implicated in KS pathogenesis. Expression of vGPCR constitutively activates several signaling pathways, including NF-kappa B, and induces the expression of proinflammatory and angiogenic factors, consistent with the inflammatory hyperproliferative nature of KS lesions. Here we show that vGPCR also constitutively activates the nuclear factor of activated T cells (NF-AT), another transcription factor important in regulation of the expression of inflammatory cytokines and related factors. NF-AT activation by vGPCR depended upon signaling through the phosphatidylinositol 3-kinase-Akt-glycogen synthetase kinase 3 (PI3-K/Akt/GSK-3) pathway and resulted in increased expression of NF-AT-dependent cell surface molecules (CD25, CD29, Fas ligand), proinflammatory cytokines (interleukin-2 [IL-2], IL-4), and proangiogenic factors (granulocyte-macrophage colony-stimulating factor GMCSF and TNF alpha). vGPCR expression also increased endothelial cell-T-cell adhesion. Although infection with HHV-8 is necessary to cause KS, coinfection with human immunodeficiency virus type 1 (HIV-1), in the absence of antiretroviral suppressive therapy, increases the risk of KS by many orders of magnitude. NF-AT and NF-kappa B activation by vGPCR was greatly increased by the HIV-1 Tat protein, although Tat alone had little effect on NF-AT. The enhancement of NF-AT by Tat appears to be mediated through collaborative stimulation of the PI3-K/Akt/GSK-3 pathway by vGPCR and Tat. Our data further support the idea that vGPCR contributes to the pathogenesis of KS by a paracrine mechanism and, in addition, provide the first evidence of collaboration between an HIV-1 protein and an HHV-8 protein.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Productos del Gen tat/metabolismo , Herpesvirus Humano 8/patogenicidad , Proteínas Nucleares , Proteínas Serina-Treonina Quinasas , Receptores de Quimiocina/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Adhesión Celular , Línea Celular , Células Cultivadas , Citocinas/metabolismo , Endotelio Vascular/citología , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Factores de Transcripción NFATC , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Piel/irrigación sanguínea , Linfocitos T/inmunología
12.
J Biol Chem ; 278(17): 14978-84, 2003 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-12584192

RESUMEN

The c-fps/fes proto-oncogene encodes a 92-kDa protein-tyrosine kinase that is involved in myeloid cell development and function. We have recently shown that expression of an activated allele of Fes (Fes(act)) in monocyte precursors resulted in their differentiation into functional macrophages through the activation of lineage-specific transcription factors. We now report that this kinase also plays a role in the survival and terminal differentiation of granulocyte progenitors. The expression of Fes(act) in factor-dependent 32D cells prevented their apoptotic death after interleukin-3 removal, but Fes(act)-expressing cells remained factor-dependent for proliferation. Removal of interleukin-3 from the Fes(act)-expressing cells was followed by granulocytic differentiation in the absence of granulocyte colony-stimulating factor within 4-8 days. The differentiated cells had distinctive granulocyte morphology and there was up-regulation of CD11b, Gr-1, and late differentiation markers such as lactoferrin, suggesting that this kinase induced terminal granulocytic differentiation. Concomitantly, Fes(act) down-regulated the macrophage marker F4/80, suggesting that the biological activity of Fes was coordinated in a lineage-specific manner. Further analysis showed that Fes(act) caused activation of CCAAT/enhancer-binding protein-alpha and STAT3, two transcription factors that are involved in granulocyte differentiation. Our results provide evidence that Fes may be a key component of the granulocyte differentiation machinery, and suggest a potential mechanism by which this kinase may regulate granulocyte-specific gene expression.


Asunto(s)
Granulocitos/citología , Células Progenitoras Mieloides/citología , Proteínas Tirosina Quinasas , Proteínas Proto-Oncogénicas/fisiología , Factores de Transcripción/metabolismo , Animales , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular , Línea Celular , Linaje de la Célula , Supervivencia Celular , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Interleucina-3/farmacología , Ratones , Proteínas Proto-Oncogénicas c-fes , Factor de Transcripción STAT3 , Transactivadores/metabolismo
13.
Mol Cell Biol ; 22(6): 1903-18, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11865067

RESUMEN

The c-fps/fes proto-oncogene encodes a 92-kDa protein tyrosine kinase that is preferentially expressed in myeloid and endothelial cells. Fes is believed to play a role in vascular development and myelopoiesis and in the inflammatory responses of granulocytes and macrophages. To help define the biological role of this kinase and identify its downstream targets, we have developed a gain-of-function allele of Fes that has potent biological activity in myeloid cell progenitors. Introduction of constitutively active Fes into bipotential U937 cells induced the appearance of fully differentiated macrophages within 6 to 12 days. The Fes-expressing differentiated cells became adherent, had distinctive macrophage morphology, and exhibited increased expression of myelomonocytic differentiation markers, including CD11b, CD11c, CD18, CD14, and the macrophage colony-stimulating factor receptor. These cells acquired phagocytic properties and exhibited NADPH oxidase and nonspecific esterase activities, confirming that they were functionally active macrophages. Concomitantly, there was downregulation of the granulocytic marker granulocyte colony-stimulating factor receptor, indicating that the biological activity of Fes was coordinated in a lineage-specific manner. A constitutively active Src did not induce macrophage morphology or upregulation of myelomonocytic markers in U937 cells, suggesting that the biological activity we observed was not a general consequence of expression of an activated nonreceptor tyrosine kinase. Analysis of possible downstream targets of Fes revealed that this kinase activated the ets family transcription factor PU.1, which is essential for macrophage development. Our results strongly implicate Fes as a key regulator of terminal macrophage differentiation and identify PU.1 as a transcription factor that may mediate some of its biological activities in myeloid cells.


Asunto(s)
Proteínas de Fusión gag-onc/metabolismo , Macrófagos , Células Progenitoras Mieloides/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transactivadores/metabolismo , Alelos , Antígenos CD/biosíntesis , Antígenos de Diferenciación/biosíntesis , Diferenciación Celular/efectos de los fármacos , Citoplasma/metabolismo , Activación Enzimática/fisiología , Proteínas de Fusión gag-onc/genética , Proteínas de Fusión gag-onc/farmacología , Humanos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/efectos de los fármacos , Fenotipo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/farmacología , Proto-Oncogenes Mas , Receptor de Factor Estimulante de Colonias de Macrófagos/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Transducción de Señal/fisiología , Tromboplastina/metabolismo , Transfección , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA