Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 32(6): 803-14, 2008 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-19111660

RESUMEN

p73 has been identified as a structural and functional homolog of the tumor suppressor p53. The transcriptional coactivator Yes-associated protein (YAP) has been demonstrated to interact with and to enhance p73-dependent apoptosis in response to DNA damage. Here, we show the existence of a proapoptotic autoregulatory feedback loop between p73, YAP, and the promyelocytic leukemia (PML) tumor suppressor gene. We demonstrate that PML is a direct transcriptional target of p73/YAP, and we show that PML transcriptional activation by p73/YAP is under the negative control of the proto-oncogenic Akt/PKB kinase. Importantly, we find that PML and YAP physically interact through their PVPVY and WW domains, respectively, causing PML-mediated sumoylation and stabilization of YAP. Hence, we determine a mechanistic pathway in response to DNA damage that could have relevant implications for the treatment of human cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis , Proteínas de Unión al ADN/metabolismo , Retroalimentación Fisiológica , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular , Línea Celular , Cisplatino/farmacología , Retroalimentación Fisiológica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Modelos Biológicos , Proteínas Nucleares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína de la Leucemia Promielocítica , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Secuencias Reguladoras de Ácidos Nucleicos/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Proteínas Supresoras de Tumor/genética , Ubiquitina/metabolismo , Proteínas Señalizadoras YAP
2.
Biochem Biophys Res Commun ; 433(2): 157-62, 2013 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-23438436

RESUMEN

Pancreas cancer, is the fourth leading cause of cancer death but its cell of origin is controversial. We compared the localization of stem cells in normal and cancerous pancreas using antibodies to the stem cell markers Nanog and LGR5. Here we show, for the first time, that LGR5 is expressed in normal pancreas, exclusively in the islets of Langerhans and it is co-localized, surprisingly, with Nanog and insulin in clusters of beta cells. In cancerous pancreas Nanog and LGR5 are expressed in the remaining islets and in all ductal cancer cells. We observed insulin staining among the ductal cancer cells, but not in metastases. This indicates that the islet's beta cells, expressing LGR5 and Nanog markers are the initiating cells of pancreas cancer, which migrated from the islets to form the ductal cancerous tissue, probably after mutation and de-differentiation. This discovery may facilitate treatment of this devastating cancer.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Receptores Acoplados a Proteínas G/metabolismo , Anciano , Biomarcadores/análisis , Biomarcadores/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/patología , Islotes Pancreáticos/metabolismo , Persona de Mediana Edad , Proteína Homeótica Nanog , Células Madre Neoplásicas/patología , Conductos Pancreáticos/metabolismo , Valores de Referencia
3.
Int J Cancer ; 131(4): E562-8, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21932419

RESUMEN

Activated p53 is necessary for tumor suppression. Homeodomain-interacting protein kinase-2 (HIPK2) is a positive regulator of functional p53. HIPK2 modulates wild-type p53 activity toward proapoptotic transcription and tumor suppression by the phosphorylation of serine 46. Knock-down of HIPK2 interferes with tumor suppression and sensitivity to chemotherapy. Combined administration of adriamycin and zinc restores activity of misfolded p53 and enables the induction of its proapoptotic and tumor suppressor functions in vitro and in vivo. We therefore looked for a cancer model where HIPK2 expression is low. MMTV-neu transgenic mice overexpressing HER2/neu, develop mammary tumors at puberty with a long latency, showing very low expression of HIPK2. Here we show that whereas these tumors are resistant to adriamycin treatment, a combination of adriamycin and zinc suppresses tumor growth in vivo in these mice, an effect evidenced by the histological features of the mammary tumors. The combined treatment of adriamycin and zinc also restores wild-type p53 conformation and induces proapoptotic transcription activity. These findings may open up new possibilities for the treatment of human cancers via the combination of zinc with chemotherapeutic agents, for a selected group of patients expressing low levels of HIPK2, with an intact p53. In addition, HIPK2 may serve as a new biomarker for tumor aggressiveness.


Asunto(s)
Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Proteína p53 Supresora de Tumor/fisiología , Zinc/administración & dosificación , Animales , Apoptosis , Secuencia de Bases , Cartilla de ADN , Genes erbB-2 , Neoplasias Mamarias Experimentales/virología , Virus del Tumor Mamario del Ratón/aislamiento & purificación , Ratones , Ratones Transgénicos , Pliegue de Proteína , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína p53 Supresora de Tumor/metabolismo
4.
Hum Mol Genet ; 18(2): 227-40, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18923003

RESUMEN

Activating mutations in FGFR3 cause achondroplasia and thanatophoric dysplasia, the most common human skeletal dysplasias. In these disorders, spinal canal and foramen magnum stenosis can cause serious neurologic complications. Here, we provide evidence that FGFR3 and MAPK signaling in chondrocytes promote synchondrosis closure and fusion of ossification centers. We observed premature synchondrosis closure in the spine and cranial base in human cases of homozygous achondroplasia and thanatophoric dysplasia as well as in mouse models of achondroplasia. In both species, premature synchondrosis closure was associated with increased bone formation. Chondrocyte-specific activation of Fgfr3 in mice induced premature synchondrosis closure and enhanced osteoblast differentiation around synchondroses. FGF signaling in chondrocytes increases Bmp ligand mRNA expression and decreases Bmp antagonist mRNA expression in a MAPK-dependent manner, suggesting a role for Bmp signaling in the increased bone formation. The enhanced bone formation would accelerate the fusion of ossification centers and limit the endochondral bone growth. Spinal canal and foramen magnum stenosis in heterozygous achondroplasia patients, therefore, may occur through premature synchondrosis closure. If this is the case, then any growth-promoting treatment for these complications of achondroplasia must precede the timing of the synchondrosis closure.


Asunto(s)
Acondroplasia/metabolismo , Desarrollo Óseo , Sistema de Señalización de MAP Quinasas , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Displasia Tanatofórica/metabolismo , Acondroplasia/genética , Acondroplasia/fisiopatología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Células Cultivadas , Condrocitos/metabolismo , Condrogénesis , Humanos , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteoblastos/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Displasia Tanatofórica/genética , Displasia Tanatofórica/fisiopatología
5.
Biochem Biophys Res Commun ; 394(1): 189-93, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20188704

RESUMEN

Reprogramming of differentiated cells into induced pluripotent cells (iPS) was accomplished in 2006 by expressing four, or less, embryonic stem cell (ESC)-specific transcription factors. Due to the possible danger of DNA damage and the potential tumorigenicity associated with such DNA damage, attempts were made to minimize DNA integration by the vectors involved in this process without complete success. Here we present a method of using RNA transfection as a tool for reprogramming human fibroblasts to iPS. We used RNA synthesized in vitro from cDNA of the same reprogramming four transcription factors. After transfection of the RNA, we show intracellular expression and nuclear localization of the respective proteins in at least 70% of the cells. We used five consecutive transfections to support continuous protein expression resulting in the formation of iPS colonies that express alkaline phosphatase and several ESC markers and that can be expanded. This method completely avoids DNA integration and may be developed to replace the use of DNA vectors in the formation of iPS.


Asunto(s)
Técnicas de Cultivo de Célula , Transdiferenciación Celular , Reprogramación Celular , Fibroblastos/citología , Células Madre Pluripotentes/citología , ARN Mensajero/genética , Humanos , Factores de Transcripción/genética , Transfección
6.
Exp Cell Res ; 315(1): 67-75, 2009 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18996371

RESUMEN

The maintenance of p53 transactivation activity is important for p53 apoptotic function. We have shown that stable knockdown of HIPK2 induces p53 misfolding with inhibition of p53 target gene transcription. In this study we established a lentiviral-based system for doxycyclin (Dox)-induced conditional interference of HIPK2 expression to evaluate the molecular mechanisms involved in p53 deregulation. We found that HIPK2 knockdown induced metallothionein 2A (MT2A) upregulation as assessed by RT-PCR analysis, increased promoter acetylation, and increased promoter luciferase activity. The MT2A upregulation correlated with resistance to Adriamycin (ADR)-driven apoptosis and with p53 inhibition. Thus, acute knockdown of HIPK2 (HIPK2i) induced misfolded p53 protein in MCF7 breast cancer cells and inhibited p53 DNA-binding and transcription activities in response to ADR treatment. Previous works show that MT may modulate p53 activity through zinc exchange. Here, we found that inhibition of MT2A expression by siRNA in the HIPK2i cells restored p53 transcription activity. Similarly zinc supplementation to HIPK2i cells restored p53 transcription activity and drug-induced apoptosis. These data support the notion that MT2A is involved in p53 deregulation and strengthen the possibility that combination of chemotherapy and zinc might be useful to treat tumors with inactive wtp53.


Asunto(s)
Metalotioneína/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteína p53 Supresora de Tumor/metabolismo , Zinc/metabolismo , Apoptosis/efectos de los fármacos , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , ADN de Neoplasias/metabolismo , Doxorrubicina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lentivirus , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Unión Proteica/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Transcripción Genética/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba/efectos de los fármacos , Zinc/farmacología
7.
Mol Cancer ; 8: 85, 2009 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-19828042

RESUMEN

BACKGROUND: Activation of p53-mediated gene transcription is a critical cellular response to DNA damage and involves a phosphorylation-acetylation cascade of p53. The discovery of differences in the response to different agents raises the question whether some of the p53 oncosuppressor functions might be exerted by different posttranslational modifications. Stress-induced homeodomain-interacting protein kinase-2 (HIPK2) phosphorylates p53 at serine-46 (Ser46) for p53 apoptotic activity; p53 acetylation at different C-terminus lysines including p300-mediated lysine-382 (Lys382) is also required for full activation of p53 transcriptional activity. The purpose of the current study was to evaluate the interplay among HIPK2, p300, and p53 in p53 acetylation and apoptotic transcriptional activity in response to drug by using siRNA interference, p300 overexpression or deacetylase inhibitors, in cancer cells. RESULTS: Knockdown of HIPK2 inhibited both adriamycin-induced Ser46 phosphorylation and Lys382 acetylation in p53 protein; however, while combination of ADR and zinc restored Ser46 phosphorylation it did not recover Lys382 acetylation. Chromatin immunoprecipitation studies showed that HIPK2 was required in vivo for efficient p300/p53 co-recruitment onto apoptotic promoters and that both p53 modifications at Ser46 and Lys382 were necessary for p53 apoptotic transcription. Thus, p53Lys382 acetylation in HIPK2 knockdown as well as p53 apoptotic activity in response to drug could be rescued by p300 overexpression. Similar effect was obtained with the Sirt1-inhibitor nicotinamide. Interestingly trichostatin A (TSA), the inhibitor of histone deacetylase complexes (HDAC) did not have effect, suggesting that Sirt1 was the deacetylase involved in p53 deacetylation in HIPK2 knockdown. CONCLUSION: These results reveal a novel role for HIPK2 in activating p53 apoptotic transcription. Our results indicate that HIPK2 may regulate the balance between p53 acetylation and deacetylation, by stimulating on one hand co-recruitment of p300 and p53Lys382 on apoptotic promoters and on the other hand by inhibiting Sirt1 deacetylase activity. We attempted to reactivate p53 apoptotic transcriptional activity by rescuing both Ser46 and Lys382 modification in response to drug. Our data propose combination strategies for the treatment of tumors with dysfunctional p53 and/or HIPK2 that include classical chemotherapy with pharmacological or natural agents such as Sirt1-deacetylase inhibitors or zinc, respectively.


Asunto(s)
Apoptosis/genética , Proteínas Portadoras/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Acetilación/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Doxorrubicina/farmacología , Proteína p300 Asociada a E1A/metabolismo , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Humanos , Ácidos Hidroxámicos/farmacología , Lisina/metabolismo , Niacinamida/farmacología , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , Sirtuina 1/metabolismo , Transcripción Genética/efectos de los fármacos , Zinc/farmacología , Proteína X Asociada a bcl-2/metabolismo
8.
Biochem Biophys Res Commun ; 365(4): 846-50, 2008 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-18039466

RESUMEN

The homeodomain transcription factor Nanog has been implicated in inhibiting differentiation and controlling pluripotency of embryonic stem (ES) cells. We used ectopic expression of Nanog in the myogenic committed C2 cells to dissect these properties. Expression of Nanog in C2 cells does not alter terminal muscle differentiation but has a profound effect on their switch to differentiate along the osteogenic lineage upon BMP treatment. Gene expression profiling revealed that ERK 1/2 phosphorylation, alkaline-phosphatase activity and osteocalcin expression were induced to much lower extent and remained suppressed even after 96h. in Nanog expressing C2 cells, compared to control C2 cells. Hence, Nanog does not inhibit terminal differentiation of committed cells but it is an inhibitor of trans-differentiation that is dependent on de-novo activation of gene transcription.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Osteogénesis/fisiología , Diferenciación Celular , Línea Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Proteína Homeótica Nanog
9.
Biochem Biophys Res Commun ; 376(1): 86-90, 2008 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-18765229

RESUMEN

We examined the microRNA profiles of Glioblastoma stem (CD133+) and non-stem (CD133-) cell populations and found up-regulation of several miRs in the CD133- cells, including miR-451, miR-486, and miR-425, some of which may be involved in regulation of brain differentiation. Transfection of GBM cells with the above miRs inhibited neurosphere formation and transfection with the mature miR-451 dispersed neurospheres, and inhibited GBM cell growth. Furthermore, transfection of miR-451 combined with Imatinib mesylate treatment had a cooperative effect in dispersal of GBM neurospheres. In addition, we identified a target site for SMAD in the promoter region of miR-451 and showed that SMAD3 and 4 activate such a promoter-luciferase construct. Transfection of SMAD in GBM cells inhibited their growth, suggesting that SMAD may drive GBM stem cells to differentiate to CD133- cells through up-regulation of miR-451 and reduces their tumorigenicity. Identification of additional miRs and target genes that regulate GBM stem cells may provide new potential drugs for therapy.


Asunto(s)
Antineoplásicos/farmacología , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , MicroARNs/genética , Células Madre Neoplásicas/efectos de los fármacos , Piperazinas/farmacología , Pirimidinas/farmacología , Antígeno AC133 , Antígenos CD/análisis , Benzamidas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/genética , Glicoproteínas/análisis , Humanos , Mesilato de Imatinib , Células Madre Neoplásicas/metabolismo , Péptidos/análisis , Regiones Promotoras Genéticas , Proteína smad3/metabolismo , Proteína Smad4/metabolismo
10.
FASEB J ; 19(1): 147-9, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15498892

RESUMEN

Human embryonic stem cells (ESC) are undifferentiated and are endowed with the capacities of self-renewal and pluripotential differentiation. Adult stem cells renew their own tissue, but whether they can transdifferentiate to other tissues is still controversial. To understand the genetic program that underlies the pluripotency of stem cells, we compared the transcription profile of ESC with that of progenitor/stem cells of human hematopoietic and keratinocytic origins, along with their mature cells to be viewed as snapshots along tissue differentiation. ESC gene profiles show higher complexity with significantly more highly expressed genes than adult cells. We hypothesize that ESC use a strategy of expressing genes that represent various differentiation pathways and selection of only a few for continuous expression upon differentiation to a particular target. Such a strategy may be necessary for the pluripotency of ESC. The progenitors of either hematopoietic or keratinocytic cells also follow the same design principle. Using advanced clustering, we show that many of the ESC expressed genes are turned off in the progenitors/stem cells followed by a further down-regulation in adult tissues. Concomitantly, genes specific to the target tissue are up-regulated toward mature cells of skin or blood.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Proyectos de Investigación/normas , Células Madre/química , Células Madre/metabolismo , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula/genética , Análisis por Conglomerados , Biología Computacional/métodos , Regulación hacia Abajo/genética , Embrión de Mamíferos/citología , Perfilación de la Expresión Génica/métodos , Perfilación de la Expresión Génica/estadística & datos numéricos , Regulación de la Expresión Génica/genética , Marcadores Genéticos/genética , Células Madre Hematopoyéticas/química , Células Madre Hematopoyéticas/metabolismo , Humanos , Queratinocitos/química , Queratinocitos/metabolismo , Queratinocitos/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Especificidad de Órganos/genética , Células Madre Pluripotentes/química , Células Madre Pluripotentes/metabolismo
11.
Oncogene ; 21(10): 1469-76, 2002 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-11896574

RESUMEN

p53 exerts its tumor suppressor effects by activating genes involved in cell growth arrest and programmed cell death. The p53 target genes inducing growth arrest are well defined whereas those inducing apoptosis are not fully characterized. Proapoptotic activity of p53 was shown to involve several genes like Bax, Noxa and Puma, which may function in the release of cytochrome c from the mitochondria. Cytochrome c associates with Apaf-1 and caspase 9 to form the apoptosome. Genetic and cellular data indicate that Apaf-1 deficiency abrogates the apoptotic effect of p53 and substitutes for p53 loss in promoting tumor formation. Here we show that Apaf-1, the mammalian homologue of C. elegans CED4, is a direct target of p53 as demonstrated by gel shift analysis of the target site sequence in the presence of p53 and by Apaf-1 promoter-luciferase assays. We also show that the p53 activation of the Apaf-1 luciferase construct can be enhanced by the putative tumor suppressor gene product, Zac-1, a transcription factor that has previously been shown to inhibit cell proliferation. Furthermore, we demonstrate that Zac-1 is a possible direct target of p53 since the sequence upstream to the first coding exon of Zac-1 contains a p53 recognition site and the luciferase construct containing this region is activated by p53. These results suggests the existence of a tightly controlled self amplifying mechanism of transcriptional activation leading to apoptosis by p53.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Genes Supresores de Tumor , Proteínas/genética , Transactivadores/metabolismo , Factores de Transcripción , Activación Transcripcional , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba , Animales , Factor Apoptótico 1 Activador de Proteasas , Secuencia de Bases , Sitios de Unión , Línea Celular , Ensayo de Cambio de Movilidad Electroforética , Genes Reporteros , Humanos , Luciferasas/análisis , Luciferasas/genética , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Biosíntesis de Proteínas , ARN Mensajero/biosíntesis , Proteínas Supresoras de Tumor
12.
Oncogene ; 22(19): 2993-3006, 2003 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-12771951

RESUMEN

To gain insight into the transformation of epidermal cells into squamous carcinoma cells (SCC), we compared the response to ultraviolet B radiation (UVB) of normal human epidermal keratinocytes (NHEK) versus their transformed counterpart, SCC, using biological and molecular profiling. DNA microarray analyses (Affymetrix), approximately 12000 genes) indicated that the major group of upregulated genes in keratinocytes fall into three categories: (i). antiapoptotic and cell survival factors, including chemokines of the CXC/CC subfamilies (e.g. IL-8, GRO-1, -2, -3, SCYA20), growth factors (e.g. HB-EGF, CTGF, INSL-4), and proinflammatory mediators (e.g. COX-2, S100A9), (ii). DNA repair-related genes (e.g. GADD45, ERCC, BTG-1, Histones), and (iii). ECM proteases (MMP-1, -10). The major downregulated genes are DeltaNp63 and PUMILIO, two potential markers for the maintenance of keratinocyte stem cells. NHEK were found to be more resistant than SCC to UVB-induced apoptosis and this resistance was mainly because of the protection from cell death by secreted survival factors, since it can be transferred from NHEK to SCC cultures by the conditioned medium. Whereas the response of keratinocytes to UVB involved regulation of key checkpoint genes (p53, MDM2, p21(Cip1), DeltaNp63), as well as antiapoptotic and DNA repair-related genes - no or little regulation of these genes was observed in SCC. The effect of UVB on NHEK and SCC resulted in upregulation of 251 and 127 genes, respectively, and downregulation of 322 genes in NHEK and 117 genes in SCC. To further analyse these changes, we used a novel unsupervised coupled two-way clustering method that allowed the identification of groups of genes that clearly partitioned keratinocytes from SCC, including a group of genes whose constitutive expression levels were similar before UVB. This allowed the identification of discriminating genes not otherwise revealed by simple static comparison in the absence of UVB irradiation. The implication of the changes in gene profile in keratinocytes for epithelial cancer is discussed.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/radioterapia , Genoma Humano , Queratinocitos/efectos de la radiación , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/radioterapia , Terapia Ultravioleta , Apoptosis/genética , Apoptosis/efectos de la radiación , Carcinoma de Células Escamosas/patología , Transformación Celular Neoplásica , Reparación del ADN/genética , Reparación del ADN/efectos de la radiación , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Cutáneas/patología
13.
FASEB J ; 18(2): 400-2, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14656992

RESUMEN

The synthetic immunomodulator AS101[ammonium trichloro(dioxoethylene-o,o')tellurate] was previously found to protect cancer patients from chemotherapy-induced bone marrow toxicity and alopecia. Here we show that AS101 induces hair growth in nude and normal mice. AS101 possesses the dual ability to both induce anagen and retard spontaneous catagen in the C57BL/6 mouse model. Anagen induced by AS101 is mediated by keratinocyte growth factor (KGF), as it is abrogated both in nude mice co-treated with AS101 plus neutralizing anti KGF antibodies and in AS101-treated transgenic mice expressing a dominant-negative KGF receptor transgene in basal keratinocytes. AS101 up-regulates KGF expression by activating the ras signaling pathway in cultured fibroblasts. AS101-induced delayed catagen is associated with inhibition of terminal differentiation marker expression both in nude and C57BL/6 mice epidermal follicular keratinocytes and in cultures of primary mouse follicular keratinocytes induced to differentiate. This activity is associated with relatively sustained elevation of p21waf. Delayed expression of terminal differentiation markers was not induced by AS101 in follicular keratinocytes from p21waf knockout mice. Because similar results were obtained with cultures of primary human keratinocytes and fibroblasts, preliminary case report studies revealed substantial hair growth when AS101 was topically applied on three adolescents who had remained alopeciac 1-2 years after chemotherapy. The results emphasize the unique mode of action of AS101 and highlight its potential clinical use for treating certain types of alopecia.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Etilenos/farmacología , Factores de Crecimiento de Fibroblastos/metabolismo , Cabello/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Proteínas ras/metabolismo , Adyuvantes Inmunológicos/farmacología , Alopecia/tratamiento farmacológico , Animales , Factor 7 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Fibroblastos , Cabello/crecimiento & desarrollo , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Telurio/farmacología
14.
Acta Histochem ; 116(1): 197-203, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23978330

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers because of late symptoms and resistance to chemotherapy and radiation therapy. We have investigated the appearance of c-kit, a stem cell marker, in both normal adult pancreatic tissue and in cancerous tissue. Apart from some very pale staining of islets of Langerhans, normal pancreas was devoid of staining with antibodies to c-kit. In contrast, in cancerous tissue that still preserves the overall integrity of the pancreatic tissue, there was a clear labeling in islets of Langerhans, which seemed to be co-localized with insulin containing ß cells. In other cases, where the pancreatic tissue was completely deteriorated, intensive labeling was clearly evident in remnants of both the exocrine and the endocrine tissues. The duct cells of the adenocarcinoma were moderately but clearly labeled with antibodies to c-kit. In contrast, in metastasis of PDAC, very intensive labeling of c-kit was evident. The location of KRAS, which is strongly associated with PDAC, was also analyzed at the initial stages of the disease, when islets of Langerhans still preserve their integrity to a large extent. KRAS was found exclusively in islets of Langerhans and overlapped in its location with insulin and c-kit expressing cells. It is suggested that the modulation of the expression of c-kit, visualized by antibodies to the oncogene molecule, may play an important role in the formation and progression of PDAC. The absence of c-kit in normal pancreas and its appearance in PDAC is probably due to a mutational event, which probably allows conversion of the ß cells into cancer stem cells (CSC). Co-expression of both c-kit and KRAS, typical markers for CSC with overlapping with insulin in islets of Langerhans, strongly support the notion that ß-cells play a central role in the development of PDAC. The use of specific drugs that can attenuate the kinase activity of c-kit or target KRAS expressing cancer cells should be tested in order to attenuate the progression of this lethal disease.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma Ductal Pancreático/enzimología , Células Madre Neoplásicas/enzimología , Neoplasias Pancreáticas/enzimología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Anciano , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Progresión de la Enfermedad , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Páncreas/enzimología , Páncreas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas c-kit/genética
15.
Acta Histochem ; 115(4): 330-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23092806

RESUMEN

LGR5 and Nanog were recently characterized as stem cell markers in various embryonic, adult and cancer stem cells. However, there are no data on their precise localization in the normal adult ovary, which may be important for the initial steps of development of ovarian cancer, the most lethal gynecological cancer. We analyzed by immunocytochemistry the precise localization of these markers in normal ovary (11 specimens, age range 43-76), in borderline specimens (12 specimens), and in serous ovarian cancer (12 specimens of stage II) which comprises the vast majority (80%) of all ovarian cancer. Surprisingly, we revealed that both Nanog and LGR5 are clearly localized in the epithelial cells of the normal ovary. However, in 5 of 12 ovaries there was no labeling at all, while in 3 ovaries staining of Nanog was more prominent with only weak labeling of LGR5. In addition, we found in 3 of 11 ovaries clear labeling in foci of both LGR5 and Nanog antibodies, with partial overlapping. Occasionally, we also found in the stroma foci labeled by either Nanog or LGR5 antibodies. In general, the stroma area of tissue sections labeled with LGR5 was much greater than that labeled with Nanog. In borderline tumors a significant portion of the specimens (7 of 12) was labeled exclusively with Nanog and not with LGR5. In ovarian carcinomas almost 100% of the cells were exclusively labeled only with Nanog (6 of 12 of the specimens) with no labeling of LGR5. These data may suggest the potential of ovaries from postmenopausal women, which express Nanog, to undergo transformation, since Nanog was shown to be oncogenic. We conclude that Nanog, which probably plays an important role in ovarian embryonic development, may be partially silenced in fertile and post-menopausal women, but is re-expressed in ovarian cancer, probably by epigenetic activation of Nanog gene expression. Expression of Nanog and LGR5 in normal ovaries and in borderline tumors may assist in the early detection and improved prognosis of ovarian cancer. Moreover, targeting of Nanog by inhibitory miRNA or other means may assist in treating this disease.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas de Homeodominio/metabolismo , Neoplasias Ováricas/patología , Ovario/patología , Receptores Acoplados a Proteínas G/metabolismo , Adulto , Anciano , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Persona de Mediana Edad , Proteína Homeótica Nanog , Clasificación del Tumor , Neoplasias Ováricas/metabolismo , Ovario/metabolismo , Posmenopausia
16.
Acta Histochem ; 115(4): 320-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23098761

RESUMEN

One paradigm of cancer development claims that cancer emerges at the niche of tissue stem cells and these cells continue to proliferate in the tumor as cancer stem cells. LGR5, a membrane receptor, was recently found to be a marker of normal colon stem cells in colon polyps and is also expressed in colon cancer stem cells. Nanog, an embryonic stem cell nuclear factor, is expressed in several embryonic tissues, but Nanog expression is not well documented in cancerous stem cells. Our aim was to examine whether both LGR5 and Nanog are expressed in the same clusters of colon stem cells or cancer stem cells, using immunocytochemistry with specific antibodies to each antigen. We analyzed this aspect using paraffin embedded tumor tissue sections obtained from 18 polyps and 36 colon cancer specimens at stages I-IV. Antibodies to LGR5 revealed membrane and cytoplasm immunostaining of scattered labeled cells in normal crypts, with no labeling of Nanog. However, in close proximity to the tumors, staining to LGR5 was much more intensive in the crypts, including that of the epithelial cells. In cancer tissue, positive LGR5 clusters of stem cells were observed mainly in poorly differentiated tumors and in only a few scattered cells in the highly differentiated tumors. In contrast, antibodies to Nanog mainly stained the growing edges of carcinoma cells, leaving the poorly differentiated tumor cells unlabeled, including the clustered stem cells that could be detected even by direct morphological examination. In polyp tissues, scattered labeled cells were immunostained with antibodies to Nanog and to a much lesser extent with antibodies to LGR5. We conclude that expression of LGR5 is probably specific to stem cells of poorly differentiated tumors, whereas Nanog is mainly expressed at the edges of highly differentiated tumors. However, some of the cell layers adjacent to the carcinoma cell layers that still remained undifferentiated, expressed mainly Nanog with only a few cells labeled with antibodies to LGR5. Considering the different sites and pattern of expression in the tumor, our data imply that targeting the clustered stem cells expressing LGR5 in poorly differentiated colon cancer may require different strategies than targeting the stem cells expressing Nanog in the highly differentiated tumors. Alternatively, combined application of specific inhibitory miRNAs to Nanog and to LGR5 expression may assist therapeutically.


Asunto(s)
Neoplasias del Colon/patología , Proteínas de Homeodominio/metabolismo , Células Madre Neoplásicas/patología , Receptores Acoplados a Proteínas G/metabolismo , Diferenciación Celular , Colon/citología , Colon/patología , Neoplasias del Colon/fisiopatología , Humanos , Proteína Homeótica Nanog , Clasificación del Tumor
17.
Cancer Biol Ther ; 13(4): 198-205, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22236966

RESUMEN

Vimentin, a mesenchymal marker, is frequently overexpressed in epithelial carcinomas undergoing epithelial to mesenchymal transition (EMT), a condition correlated with invasiveness and poor prognosis. Therefore, vimentin is a potential molecular target for anticancer therapy. Emerging studies in experimental models underscore the functions of homeodomain-interacting protein kinase 2 (HIPK2) as potential oncosuppressor by acting as transcriptional corepressor or catalytic activator of molecules involved in apoptosis and response to antitumor drugs. However, an involvement of HIPK2 in limiting tumor invasion remains to be elucidated. This study, by starting with a microarray analysis, demonstrates that HIPK2 downregulates vimentin expression in invasive, vimentin-positive, MDA-MB-231 breast cancer cells and in the non-invasive MCF7 breast cancer cells subjected to chemical hypoxia, a drive for mesenchymal shift and tumor invasion. At functional level, vimentin downregulation by HIPK2 correlates with inhibition of breast tumor cell invasion. Together, these data show that vimentin is a novel target for HIPK2 repressor function and that HIPK2-mediated vimentin downregulation can contribute to inhibition of breast cancer cells invasion that might be applied in clinical therapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas Portadoras/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Vimentina/metabolismo , Apoptosis/fisiología , Neoplasias de la Mama/genética , Proteínas Portadoras/genética , Línea Celular Tumoral , Movimiento Celular/fisiología , Regulación hacia Abajo , Femenino , Humanos , Invasividad Neoplásica , Proteínas Serina-Treonina Quinasas/genética , Transfección , Vimentina/genética
18.
Cell Cycle ; 10(10): 1679-89, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21508668

RESUMEN

Absence of p53 expression or expression of mutant p53 (mtp53) are common in human cancers and are associated with increased cancer resistance to chemo- and radiotherapy. Therefore, significant efforts towards pharmaceutical reactivation of defective p53 pathways are underway. We previously reported that, in HIPK2 knockdown background, p53 undergoes misfolding with inhibition of DNA binding and transcriptional activities that correlate with increased chemoresistance, and that zinc rescues wild-type p53 activity. Zinc has a crucial role in the biology of p53, in that p53 binds to DNA through a structurally complex domain stabilized by zinc atom. In this study, we explored the role of zinc in p53 reactivation in mutant p53-expressing cancer cells. We found that zinc re-established chemosensitivity in breast cancer SKBR3 (expressing R175H mutation) and glioblastoma U373MG (expressing R273H mutation) cell lines. Biochemical studies showed that zinc partly induced the transition of mutant p53 protein (reactive to conformation-sensitive PAb240 antibody for mutant conformation) into a functional conformation (reactive to conformation-sensitive PAb1620 antibody for wild-type conformation). Zinc-mediated p53 reactivation also reduced the mtp53/p73 interaction restoring both wtp53 and p73 binding to target gene promoters by ChIP assay with in vivo induction of wtp53 target gene expression, which rendered mutant p53 cells more prone to drug killing in vitro. Finally, zinc administration in U373MG tumor xenografts increased drug-induced tumor regression in vivo, which correlated with increased wild-type p53 protein conformation. These results show that the use of zinc might restore drug sensitivity and inhibit tumor growth by reactivating mutant p53.


Asunto(s)
Antineoplásicos/farmacología , Proteína p53 Supresora de Tumor/química , Zinc/fisiología , Animales , Antineoplásicos/uso terapéutico , Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , Mutación , Proteínas Nucleares/metabolismo , Conformación Proteica , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Trasplante Heterólogo , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Zinc/farmacología
19.
Oncotarget ; 2(12): 1191-202, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22202117

RESUMEN

Hypoxia-inducible factor 1 (HIF-1), the major transcription factor specifically activated during hypoxia, regulates genes involved in critical aspects of cancer biology, including angiogenesis, cell proliferation, glycolysis and invasion. The HIF-1a subunit is stabilized by low oxygen, genetic alteration and cobaltous ions, and its over-expression correlates with drug resistance and increased cancer mortality in various cancer types, therefore representing an important anticancer target. Zinc supplementation has been shown to counteract the hypoxic phenotype in cancer cells, in vitro and in vivo, hence, understanding the molecular pathways modulated by zinc under hypoxia may provide the basis for reprogramming signalling pathways for anticancer therapy. Here we performed genome-wide analyses of colon cancer cells treated with combinations of cobalt, zinc and anticancer drug and evaluated the effect of zinc on gene expression patterns. Using Principal Component Analysis we found that zinc markedly reverted the cobalt-induced changes of gene expression, with reactivation of the drug-induced transcription of pro-apoptotic genes. We conclude that the hypoxia pathway is a potential therapeutic target addressed by zinc that also influences tumor cell response to anticancer drug.


Asunto(s)
Antineoplásicos/farmacología , Cobalto/farmacología , Neoplasias del Colon/metabolismo , Regulación Neoplásica de la Expresión Génica , Zinc/farmacología , Apoptosis/efectos de los fármacos , Hipoxia de la Célula , Línea Celular Tumoral , Neoplasias del Colon/genética , Doxorrubicina/farmacología , Estudio de Asociación del Genoma Completo , Humanos , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Oxígeno/análisis , Análisis de Componente Principal , Transducción de Señal
20.
FEBS Lett ; 584(19): 4253-8, 2010 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-20849851

RESUMEN

Homeodomain-interacting protein kinase-2 (HIPK2) is a crucial regulator of p53 apoptotic function by phosphorylating serine 46 (Ser46) in response to DNA damage. In tumors with wild-type p53, its tumor suppressor function is often impaired by MDM2 overexpression that targets p53 for proteasomal degradation. Likewise, MDM2 targets HIPK2 for protein degradation impairing p53-apoptotic function. Here we report that zinc antagonised MDM2-induced HIPK2 degradation as well as p53 ubiquitination. The zinc inhibitory effect on MDM2 activity leads to HIPK2-induced p53Ser46 phosphorylation and p53 pro-apoptotic transcriptional activity. These results suggest that zinc derivatives are potential molecules to target the MDM2-induced HIPK2/p53 inhibition.


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Proteínas Portadoras/genética , Línea Celular , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Genes p53 , Células HeLa , Humanos , Modelos Biológicos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/genética , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Ubiquitinación/efectos de los fármacos , Zinc/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA