Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Gastroenterology ; 144(1): 134-144.e6, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23041331

RESUMEN

BACKGROUND & AIMS: A fraction of gastrointestinal stromal tumor (GIST) cells overexpress the platelet-derived growth factor receptor (PDGFR)A, although most overexpress KIT. It is not known if this is because these receptor tyrosine kinases have complementary oncogenic potential, or because of heterogeneity in the cellular origin of GIST. Little also is known about why Hedgehog (HH) signaling is activated in some GIST. HH binds to and inactivates the receptor protein patched homolog (PTCH). METHODS: Ptch was conditionally inactivated in mice (to achieve constitutive HH signaling) using a Cre recombinase regulated by the lysozyme M promoter. Cre-expressing cells were traced using R26R-LacZ reporter mice. Tumors were characterized by in situ hybridization, immunohistochemistry, immunoblot, and quantitative reverse-transcriptase polymerase chain reaction analyses. Cell transformation was assessed by soft agar assay. RESULTS: Loss of Ptch from lysozyme M-expressing cells resulted in the development of tumors of GIST-like localization and histology; these were reduced when mice were given imatinib, a drug that targets KIT and PDGFRA. The Hh signaling pathway was activated in the tumor cells, and Pdgfrα, but not Kit, was overexpressed and activated. Lineage tracing revealed that Cre-expressing intestinal cells were Kit-negative. These cells sometimes expressed Pdgfrα and were located near Kit-positive interstitial cells of Cajal. In contrast to KIT, activation of PDGFRA increased anchorage-independent proliferation and was required for tumor formation in mice by cells with activated HH signaling. CONCLUSIONS: Inactivation of Ptch in mice leads to formation of GIST-like tumors that express Pdgfrα, but not Kit. Activation of Pdgfrα signaling appears to facilitate tumorigenesis.


Asunto(s)
Neoplasias Gastrointestinales/genética , Tumores del Estroma Gastrointestinal/metabolismo , Proteínas Hedgehog/genética , Leiomiosarcoma/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Superficie Celular/genética , Animales , Benzamidas , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/metabolismo , Neoplasias Gastrointestinales/patología , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/patología , Expresión Génica , Genotipo , Proteínas Hedgehog/metabolismo , Humanos , Mesilato de Imatinib , Integrasas/genética , Integrasas/metabolismo , Mucosa Intestinal/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Leiomiosarcoma/metabolismo , Ratones , Muramidasa/genética , Muramidasa/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Patched , Receptor Patched-1 , Piperazinas/uso terapéutico , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Receptores de Superficie Celular/metabolismo , Transducción de Señal/genética , Proteína con Dedos de Zinc GLI1 , Proteína Gli2 con Dedos de Zinc , Proteína Gli3 con Dedos de Zinc
2.
Oncogene ; 40(31): 4955-4966, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34172934

RESUMEN

A prototypic pediatric cancer that frequently shows activation of RAS signaling is embryonal rhabdomyosarcoma (ERMS). ERMS also show aberrant Hedgehog (HH)/GLI signaling activity and can be driven by germline mutations in this pathway. We show, that in ERMS cell lines derived from sporadic tumors i.e. from tumors not caused by an inherited genetic variant, HH/GLI signaling plays a subordinate role, because oncogenic mutations in HRAS, KRAS, or NRAS (collectively named oncRAS) inhibit the main HH target GLI1 via the MEK/ERK-axis, but simultaneously increase proliferation and tumorigenicity. oncRAS also modulate expression of stem cell markers in an isoform- and context-dependent manner. In Hh-driven murine ERMS that are caused by a Patched mutation, oncHRAS and mainly oncKRAS accelerate tumor development, whereas oncNRAS induces a more differentiated phenotype. These features occur when the oncRAS mutations are induced at the ERMS precursor stage, but not when induced in already established tumors. Moreover, in contrast to what is seen in human cell lines, oncRAS mutations do not alter Hh signaling activity and marginally affect expression of stem cell markers. Together, all three oncRAS mutations seem to be advantageous for ERMS cell lines despite inhibition of HH signaling and isoform-specific modulation of stem cell markers. In contrast, oncRAS mutations do not inhibit Hh-signaling in Hh-driven ERMS. In this model, oncRAS mutations seem to be advantageous for specific ERMS populations that occur within a specific time window during ERMS development. In addition, this window may be different for individual oncRAS isoforms, at least in the mouse.


Asunto(s)
Susceptibilidad a Enfermedades , Genes ras , Neoplasias/etiología , Neoplasias/metabolismo , Isoformas de Proteínas/genética , Factores de Edad , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Mutación , Neoplasias/patología , Células Madre Neoplásicas , Oncogenes , Receptor Patched-1/genética , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
3.
Carcinogenesis ; 30(6): 918-26, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19321799

RESUMEN

Mutations in Patched (PTCH) have been associated with tumors characteristic both for children [medulloblastoma (MB) and rhabdomyosarcoma (RMS)] and for elderly [basal cell carcinoma (BCC)]. The determinants of the variability in tumor onset and histology are unknown. We investigated the effects of the time-point and dosage of Ptch inactivation on tumor spectrum using conditional Ptch-knockout mice. Ptch heterozygosity induced prenatally resulted in the formation of RMS, which was accompanied by the silencing of the remaining wild-type Ptch allele. In contrast, RMS was observed neither after mono- nor biallelic postnatal deletion of Ptch. Postnatal biallelic deletion of Ptch led to BCC precancerous lesions of the gastrointestinal epithelium and mesenteric tumors. Hamartomatous gastrointestinal cystic tumors were induced by monoallelic, but not biallelic Ptch mutations, independently of the time-point of mutation induction. These data suggest that the expressivity of Ptch deficiency is largely determined by the time-point, the gene dose and mode of Ptch inactivation. Furthermore, they point to key differences in the tumorigenic mechanisms underlying adult and childhood tumors. The latter ones are unique among all tumors since their occurrence decreases rather than increases with age. A better understanding of mechanisms underlying this ontological restriction is of potential therapeutic value.


Asunto(s)
Envejecimiento/patología , Carcinoma Basocelular/genética , Dosificación de Gen , Silenciador del Gen , Receptores de Superficie Celular/fisiología , Rabdomiosarcoma/genética , Envejecimiento/genética , Animales , Carcinoma Basocelular/patología , Quistes/genética , Quistes/patología , Neoplasias Gastrointestinales/embriología , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/patología , Mutación de Línea Germinal , Ratones , Ratones Noqueados , Neoplasias de los Músculos/genética , Neoplasias de los Músculos/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutación , Receptores Patched , Receptor Patched-1 , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/patología , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Receptores de Superficie Celular/genética , Rabdomiosarcoma/embriología , Rabdomiosarcoma/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
4.
Front Genet ; 10: 1185, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31867038

RESUMEN

Cutaneous squamous cell carcinoma (cSCC) is the second most common skin tumor in humans. Although current therapies are sufficient to clear the tumor in many cases, the overall risk of cSCC metastasis is still 5%. Alternative treatment options could help to overcome this situation. Here we focused on the role of the Hedgehog (HH) signaling pathway and its interplay with epidermal growth factor receptor (EGFR) signaling in cSCC. The analyses revealed that, despite lack of Sonic HH (SHH) expression, a subset of human cSCC can express GLI1, a marker for active HH signaling, within distinct tumor areas. In contrast, all tumors strongly express EGFR and the hair follicle stem cell marker SOX9 at the highly proliferative tumor-stroma interface, whereas central tumor regions with a more differentiated stratum spinosum cell type lack both EGFR and SOX9 expression. In vitro experiments indicate that activation of EGFR signaling in the human cSCC cell lines SCL-1, MET-1, and MET-4 leads to GLI1 inhibition via the MEK/ERK axis without affecting cellular proliferation. Of note, EGFR activation also inhibits cellular migration of SCL-1 and MET-4 cells. Because proliferation and migration of the cells is also not altered by a GLI1 knockdown, GLI1 is apparently not involved in processes of aggressiveness in established cSCC tumors. In contrast, our data rather suggest a negative correlation between Gli1 expression level and cSCC formation because skin of Ptch +/- mice with slightly elevated Gli1 expression levels is significantly less susceptible to chemically-induced cSCC formation compared to murine wildtype skin. Although not yet formally validated, these data open the possibility that GLI1 (and thus HH signaling) may antagonize cSCC initiation and is not involved in cSCC aggressiveness, at least in a subset of cSCC.

5.
Oncotarget ; 6(11): 9113-24, 2015 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-25823816

RESUMEN

Mice with heterozygous loss of the tumor suppressor Patched1 (Ptch) develop rhabdomyosarcoma (RMS)-like tumors. However, Ptch transcripts are consistently overexpressed in these tumors. We have recently shown that the upregulated transcripts are derived from the mutated Ptch allele thus leading to the hypothesis that the wild-type allele is repressed during RMS development. Here we describe epigenetic changes taking place at the Ptch locus during RMS development. We showed a lower degree of DNA-methylation in methylation-sensitive CpG regions of the Ptch promoter in RMS compared to normal muscle from heterozygous Ptch animals. In agreement with these results, treatment of heterozygous Ptch mice with the DNA demethylating agent 5-aza-2-deoxycytidine (5-aza-dC) between embryonic days E9.5-E11.5 significantly accelerated RMS formation. Since Ptch promoter methylation occurs after/around E13.5, the window for RMS initiation during embryogenesis, these results provide additional evidence that Ptch promoter hypomethylation may contribute to RMS formation. We have also demonstrated increased trimethylation of histone H3 lysine 4 (H3K4me3) and preferential binding of Gli1, a known Ptch activator, to the mutant locus in RMS. Together, these findings support an alternative model for RMS formation in heterozygous Ptch mice including loss of methylation and concomitant occupancy by activating histone marks of mutant Ptch.


Asunto(s)
Metilación de ADN , Histonas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas de Neoplasias/fisiología , Regiones Promotoras Genéticas/genética , Procesamiento Proteico-Postraduccional , Receptores de Superficie Celular/fisiología , Rabdomiosarcoma/genética , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Islas de CpG , Metilación de ADN/efectos de los fármacos , Decitabina , Desarrollo Embrionario/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Genes Supresores de Tumor , Edad Gestacional , Heterocigoto , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Genéticos , Mutación , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Receptores Patched , Receptor Patched-1 , Polimorfismo de Nucleótido Simple , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética , Rabdomiosarcoma/metabolismo , Transducción de Señal , Proteína con Dedos de Zinc GLI1
6.
J Invest Dermatol ; 134(10): 2620-2629, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24662765

RESUMEN

The development of basal cell carcinoma (BCC), the most frequently diagnosed tumor among persons with European ancestry, is closely linked to mutations in the Hedgehog (Hh) receptor and tumor suppressor Patched1 (Ptch). Using Ptch(flox/flox)CD4Cre(+/-) mice, in which Ptch was ablated in CD4Cre-expressing cells, we demonstrate that the targeted cells can give rise to BCC after treatment with DMBA (7,12-dimethylbenz(a)anthracene)/TPA (12-O-tetradecanoylphorbol-13-acetate), but not after wounding of the skin. In addition, in this model, BCC are not caused by malfunctioning of Ptch-deficient T cells, as BCC did not develop when bone marrow (BM) of Ptch(flox/flox)CD4Cre(+/-) mice was transplanted into Ptch wild-type mice. Instead, lineage-tracing experiments and flow cytometric analyses suggest that the tumors are initiated from rare Ptch-deficient stem cell-like cells of the epidermis that express CD4. As DMBA/TPA is a prerequisite for BCC development in this model, the initiated cells need a second stimulus for expansion and tumor formation. However, in contrast to papilloma, this stimulus seems to be unrelated to alterations in the Ras signaling cascade. Together, these data suggest that biallelic loss of Ptch in CD4(+) cells does not suffice for BCC formation and that BCC formation requires a second so far unknown event, at least in the Ptch(flox/flox)CD4Cre(+/-) BCC mouse model.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno/efectos adversos , Linfocitos T CD4-Positivos/patología , Carcinogénesis/inducido químicamente , Carcinoma Basocelular/fisiopatología , Epidermis/patología , Receptores de Superficie Celular/deficiencia , Neoplasias Cutáneas/fisiopatología , Acetato de Tetradecanoilforbol/efectos adversos , 9,10-Dimetil-1,2-benzantraceno/farmacología , Alelos , Animales , Linfocitos T CD4-Positivos/fisiología , Carcinogénesis/efectos de los fármacos , Carcinógenos/farmacología , Carcinoma Basocelular/inducido químicamente , Carcinoma Basocelular/patología , Modelos Animales de Enfermedad , Epidermis/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mutación/genética , Receptores Patched , Receptor Patched-1 , Proteínas Proto-Oncogénicas p21(ras)/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Transducción de Señal/fisiología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/patología , Células Madre/patología , Células Madre/fisiología , Acetato de Tetradecanoilforbol/farmacología
7.
PLoS One ; 9(4): e93555, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24691432

RESUMEN

Basal cell carcinoma (BCC) belongs to the group of non-melanoma skin tumors and is the most common tumor in the western world. BCC arises due to mutations in the tumor suppressor gene Patched1 (Ptch). Analysis of the conditional Ptch knockout mouse model for BCC reveals that macrophages and dendritic cells (DC) of the skin play an important role in BCC growth restraining processes. This is based on the observation that a clodronate-liposome mediated depletion of these cells in the tumor-bearing skin results in significant BCC enlargement. The depletion of these cells does not modulate Ki67 or K10 expression, but is accompanied by a decrease in collagen-producing cells in the tumor stroma. Together, the data suggest that cutaneous macrophages and DC in the tumor microenvironment exert an antitumor effect on BCC.


Asunto(s)
Carcinogénesis/genética , Carcinoma Basocelular/genética , Receptores de Superficie Celular/genética , Neoplasias Cutáneas/genética , Animales , Carcinoma Basocelular/tratamiento farmacológico , Carcinoma Basocelular/metabolismo , Carcinoma Basocelular/patología , Ácido Clodrónico/administración & dosificación , Células Dendríticas/efectos de los fármacos , Células Dendríticas/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Liposomas/administración & dosificación , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Ratones Noqueados , Mutación , Receptores Patched , Receptor Patched-1 , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
8.
J Skin Cancer ; 2012: 907543, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23024864

RESUMEN

Basal cell carcinoma (BCC) is the most common human tumor. Mutations in the hedgehog (HH) receptor Patched (PTCH) are the main cause of BCC. Due to their high and increasing incidence, BCC are becoming all the more important for the health care system. Adequate animal models are required for the improvement of current treatment strategies. A good model should reflect the situation in humans (i.e., BCC initiation due to Ptch mutations on an immunocompetent background) and should allow for (i) BCC induction at a defined time point, (ii) analysis of defined BCC stages, and (iii) induction of BCC in 100% of animals. In addition, it should be easy to handle. Here, we compare several currently existing conventional and conditional Ptch knockout mouse models for BCC and their potential use in preclinical research. In addition, we provide new data using conditional Ptch(flox/flox) mice and the K5-Cre-ER(T+/-) driver.

9.
PLoS One ; 7(12): e52898, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23300809

RESUMEN

We searched for a drug capable of sensitization of sarcoma cells to doxorubicin (DOX). We report that the dual PI3K/mTOR inhibitor PI103 enhances the efficacy of DOX in several sarcoma cell lines and interacts with DOX in the induction of apoptosis. PI103 decreased the expression of MDR1 and MRP1, which resulted in DOX accumulation. However, the enhancement of DOX-induced apoptosis was unrelated to DOX accumulation. Neither did it involve inhibition of mTOR. Instead, the combination treatment of DOX plus PI103 activated Bax, the mitochondrial apoptosis pathway, and caspase 3. Caspase 3 activation was also observed in xenografts of sarcoma cells in nude mice upon combination of DOX with the specific PI3K inhibitor GDC-0941. Although the increase in apoptosis did not further impact on tumor growth when compared to the efficient growth inhibition by GDC-0941 alone, these findings suggest that inhibition of PI3K may improve DOX-induced proapoptotic effects in sarcoma. Taken together with similar recent studies of neuroblastoma- and glioblastoma-derived cells, PI3K inhibition seems to be a more general option to sensitize tumor cells to anthracyclines.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Sarcoma/tratamiento farmacológico , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Caspasa 3/metabolismo , Línea Celular Tumoral , Citocromos c/metabolismo , Doxorrubicina/administración & dosificación , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Furanos/administración & dosificación , Expresión Génica/efectos de los fármacos , Humanos , Indazoles/administración & dosificación , Ratones , Ratones Desnudos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Piridinas/administración & dosificación , Pirimidinas/administración & dosificación , Sarcoma/enzimología , Sulfonamidas/administración & dosificación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína X Asociada a bcl-2/metabolismo
10.
Mol Cancer Ther ; 10(11): 2179-88, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21878656

RESUMEN

Activation of the Hedgehog (Hh)-signaling pathway due to deficiency in the Hh receptor Patched1 (Ptch) is the pivotal defect leading to formation of basal cell carcinoma (BCC). Recent reports provided evidence of Ptch-dependent secretion of vitamin D(3)-related compound, which functions as an endogenous inhibitor of Hh signaling by repressing the activity of the signal transduction partner of Ptch, Smoothened (Smo). This suggests that Ptch-deficient tumor cells are devoid of this substance, which in turn results in activation of Hh-signaling. Here, we show that the application of the physiologically active form of vitamin D(3), calcitriol, inhibits proliferation and growth of BCC of Ptch mutant mice in vitro and in vivo. This is accompanied by the activation of the vitamin D receptor (Vdr) and induction of BCC differentiation. In addition, calcitriol inhibits Hh signaling at the level of Smo in a Vdr-independent manner. The concomitant antiproliferative effects on BCC growth are stronger than those of the Hh-specific inhibitor cyclopamine, even though the latter more efficiently inhibits Hh signaling. Taken together, we show that exogenous supply of calcitriol controls the activity of 2 independent pathways, Hh and Vdr signaling, which are relevant to tumorigenesis and tumor treatment. These data suggest that calcitriol could be a therapeutic option in the treatment of BCC, the most common tumor in humans.


Asunto(s)
Antineoplásicos/farmacología , Calcitriol/farmacología , Carcinoma Basocelular/metabolismo , Proteínas Hedgehog/antagonistas & inhibidores , Receptores de Calcitriol/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Carcinoma Basocelular/genética , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteínas Hedgehog/metabolismo , Ratones , Ratones Noqueados , Mutación , Proteínas Oncogénicas/metabolismo , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/genética , Transactivadores/metabolismo , Proteína con Dedos de Zinc GLI1
11.
Cancer Res ; 70(7): 2739-48, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20233865

RESUMEN

Basal cell carcinoma (BCC) is the most common skin tumor in humans. Although BCCs rarely metastasize, they can cause significant morbidity due to local aggressiveness. Approximately 20% of BCCs show signs of spontaneous regression. The understanding of molecular events mediating spontaneous regression has the potential to reduce morbidity of BCC and, potentially, other tumors, if translated into tumor therapies. We show that BCCs induced in conditional Ptch(flox/flox)ERT2(+/-) knockout mice regress with time and show a more differentiated phenotype. Differentiation is accompanied by Wnt5a expression in the tumor stroma, which is first detectable at the fully developed tumor stage. Coculture experiments revealed that Wnt5a is upregulated in tumor-adjacent macrophages by soluble signals derived from BCC cells. In turn, Wnt5a induces the expression of the differentiation marker K10 in tumor cells, which is mediated by Wnt/Ca(2+) signaling in a CaMKII-dependent manner. These data support a role of stromal Wnt5a in BCC differentiation and regression, which may have important implications for development of new treatment strategies for this tumor. Taken together, our results establish BCC as an easily accessible model of tumor regression. The regression of BCC despite sustained Hedgehog signaling activity seems to be mediated by tumor-stromal interactions via Wnt5a signaling.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Carcinoma Basocelular/metabolismo , Neoplasias Cutáneas/metabolismo , Proteínas Wnt/biosíntesis , Animales , Carcinoma Basocelular/genética , Carcinoma Basocelular/patología , Diferenciación Celular/fisiología , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Noqueados , Células 3T3 NIH , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Células del Estroma/metabolismo , Células del Estroma/patología , Tamoxifeno/farmacología , Transfección , Proteínas Wnt/genética , Proteína Wnt-5a
12.
Cancer Res ; 69(3): 887-95, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19155313

RESUMEN

Patched (Ptch) heterozygous mice develop medulloblastoma (MB) and rhabdomyosarcoma (RMS) resembling the corresponding human tumors. We have previously shown that epigenetic silencing of the intact Ptch allele contributes to tumor formation in this model. Here, we investigated whether targeting of epigenetic silencing mechanisms could be useful in the treatment of Ptch-associated cancers. A reduction of endogenous DNA methyltransferase1 (Dnmt1) activity significantly reduced tumor incidence in heterozygous Ptch knockout mice. A combined treatment with the Dnmt inhibitor 5-aza-2'deoxycytidine (5-aza-dC) and the histone deacetlyase (HDAC) inhibitor valproic acid (VPA) efficiently prevented MB and RMS formation, whereas monotherapies with either drug were less effective. Wild-type Ptch expression was efficiently reactivated in tumors by 5-aza-dC/VPA combination therapy. This was associated with reduced methylation of the Ptch promoter and induction of histone hyperacetylation suggesting inhibition of HDACs in vivo. However, the treatment was not effective in clinically overt, advanced stage tumors. This is a first in vivo demonstration that targeting of Dnmt and HDAC activities is highly effective in preventing formation of Ptch-associated tumors. The results suggest a novel clinical strategy for consolidation therapy of corresponding tumors in humans after completion of conventional treatment. Our data also suggest that epigenetic therapy may be less effective in treating advanced stages of tumors, at least in this tumor model.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Meduloblastoma/tratamiento farmacológico , Receptores de Superficie Celular/genética , Rabdomiosarcoma/tratamiento farmacológico , Acetilación , Animales , Azacitidina/administración & dosificación , Azacitidina/análogos & derivados , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Decitabina , Expresión Génica/efectos de los fármacos , Silenciador del Gen , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Meduloblastoma/genética , Meduloblastoma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Receptores Patched , Receptor Patched-1 , Rabdomiosarcoma/genética , Rabdomiosarcoma/metabolismo , Ácido Valproico/administración & dosificación
13.
Blood ; 110(6): 1814-23, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17536012

RESUMEN

A first step in hematopoiesis is the specification of the lymphoid and myeloid lineages from multipotent progenitor cells in the bone marrow. Using a conditional ablation strategy in adult mice, we show that this differentiation step requires Patched (Ptch), the cell surface-bound receptor for Hedgehog (Hh). In the absence of Ptch, the development of T- and B-lymphoid lineages is blocked at the level of the common lymphoid progenitor in the bone marrow. Consequently, the generation of peripheral T and B cells is abrogated. Cells of the myeloid lineage develop normally in Ptch mutant mice. Finally, adoptive transfer experiments identified the stromal cell compartment as a critical Ptch-dependent inducer of lymphoid versus myeloid lineage commitment. Our data show that Ptch acts as a master switch for proper diversification of hematopoietic stem cells in the adult organism.


Asunto(s)
Linfocitos B/metabolismo , Linaje de la Célula , Células Madre Multipotentes/metabolismo , Receptores de Superficie Celular/fisiología , Linfocitos T/metabolismo , Traslado Adoptivo , Animales , Linfocitos B/patología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Femenino , Citometría de Flujo , Granulocitos/citología , Granulocitos/metabolismo , Hematopoyesis , Inmunofenotipificación , Integrasas/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Multipotentes/citología , Células Mieloides/citología , Células Mieloides/metabolismo , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/genética , Células Madre/citología , Células del Estroma/citología , Células del Estroma/metabolismo , Linfocitos T/patología , Timo/patología , Factores de Tiempo
14.
Eur J Immunol ; 34(11): 3257-66, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15376195

RESUMEN

The 155-kDa complement regulator factor H (FH) is the predominant soluble regulatory protein of the complement system. It acts as a cofactor for the factor I-mediated conversion of the component C3b to iC3b, competes with factor B for a binding site on C3b and C3(H2O) and promotes the dissociation of the C3bBb complex. The primary site of synthesis is the liver, i.e. FH-specific mRNA and protein were identified in both hepatocytes (HC) and Kupffer cells (KC). Previous studies in rat primary HC and KC had shown that the proinflammatory cytokine IFN-gamma influences the balance between activation and inhibition of the complement system through up-regulation of the inhibitory FH. In this study we show that C5a, as a product of complement activation, stimulates the expression of FH-specific mRNA and protein in KC and thus induces a negative feedback. Quantitative-competitive RT-PCR showed an approximate threefold C5a-induced up-regulation of FH. ELISA analyses revealed a corresponding increase in FH protein in the supernatants of KC. The up-regulation of FH was completely inhibited by the C5a-blocking monoclonal antibody 6-9F. Furthermore, an involvement of LPS and IFN-gamma was excluded, which strongly indicates a direct effect of C5a on the expression of FH in KC.


Asunto(s)
Activación de Complemento/inmunología , Complemento C5a/inmunología , Factor H de Complemento/inmunología , Animales , Anticuerpos Bloqueadores/inmunología , Northern Blotting , Factor H de Complemento/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/metabolismo , Hígado/inmunología , Masculino , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba/inmunología , alfa-N-Acetilgalactosaminidasa/inmunología
15.
Genomics ; 84(5): 853-8, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15475264

RESUMEN

Mutations in the Patched (Ptch1) gene are responsible for various familial and sporadic cancers. Ptch1(neo67/+) mice, in which exons 6 and 7 are deleted, show genetic background-dependent susceptibility to the development of muscle tumors resembling human rhabdomyosarcoma (RMS); BALB/c (BALB) is a susceptible strain whereas C57BL/6 (B6) shows resistance. A genome-wide linkage analysis was carried out using Ptch1(neo67/+)mice produced from B6 x (BALB x B6) backcrosses to identify loci involved in the control of RMS susceptibility. Quantitative trait locus mapping with the censored tumor latency time as the quantitative parameter was used to detect a significant RMS susceptibility modifier locus, Parms1 (Patched-Associated RMS 1), on chromosome 2 between D2Mit37 and D2Mit102 (LRS = 10). A Kaplan-Meier survival curve revealed that mice with the B6/BALB genotype develop tumors more frequently and much faster as compared to mice homozygous for the B6 allele (P = 0.02). Additional loci not reaching linkage significance were also detected for medulloblastoma resistance.


Asunto(s)
Cromosomas de los Mamíferos/genética , Predisposición Genética a la Enfermedad/genética , Proteínas/genética , Rabdomiosarcoma/genética , Animales , Mapeo Cromosómico , Cruzamientos Genéticos , Femenino , Ligamiento Genético , Marcadores Genéticos/genética , Péptidos y Proteínas de Señalización Intracelular , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores Patched , Receptor Patched-1 , Sitios de Carácter Cuantitativo/genética , Receptores de Superficie Celular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA