Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(14)2022 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-35886842

RESUMEN

Doxorubicin (doxo) remains the standard of care for patients with advanced soft tissue sarcoma (STS), even though response rates to doxo are only around 14% to 18%. We evaluated enapotamab vedotin (EnaV), an AXL-specific antibody-drug conjugate (ADC), in a panel of STS patient-derived xenografts (PDX). Eight models representing multiple STS subtypes were selected from our STS PDX platform (n = 45) by AXL immunostaining on archived passages. Models were expanded by unilateral transplantation of tumor tissue into the left flank of 20 NMRI nu/nu mice. Once tumors were established, mice were randomized into an EnaV treatment group, or a group treated with isotype control ADC. Treatment efficacy was assessed by tumor volume evaluation, survival analysis, and histological evaluation of tumors, and associated with AXL expression. EnaV demonstrated significant tumor growth delay, regression, and/or prolonged survival compared to isotype control ADC in 5/8 STS PDX models investigated. Experimental passages of responding models were all found positive for AXL at varying levels, but no linear relationship could be identified between the level of expression and level of response to EnaV. One model was found negative for AXL on experimental passage and did not respond to EnaV. This study provides a preclinical rationale for the evaluation of AXL-targeting ADCs in the treatment of AXL-expressing sarcomas.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Sarcoma , Neoplasias de los Tejidos Blandos , Animales , Ratones , Antineoplásicos/uso terapéutico , Modelos Animales de Enfermedad , Inmunoconjugados/uso terapéutico , Sarcoma/tratamiento farmacológico , Sarcoma/patología , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Neoplasias de los Tejidos Blandos/patología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
2.
PLoS Genet ; 9(5): e1003424, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23658527

RESUMEN

Canonical Wnt signaling plays a rate-limiting role in regulating self-renewal and differentiation in mouse embryonic stem cells (ESCs). We have previously shown that mutation in the Apc (adenomatous polyposis coli) tumor suppressor gene constitutively activates Wnt signaling in ESCs and inhibits their capacity to differentiate towards ecto-, meso-, and endodermal lineages. However, the underlying molecular and cellular mechanisms through which Wnt regulates lineage differentiation in mouse ESCs remain to date largely unknown. To this aim, we have derived and studied the gene expression profiles of several Apc-mutant ESC lines encoding for different levels of Wnt signaling activation. We found that down-regulation of Tcf3, a member of the Tcf/Lef family and a key player in the control of self-renewal and pluripotency, represents a specific and primary response to Wnt activation in ESCs. Accordingly, rescuing Tcf3 expression partially restored the neural defects observed in Apc-mutant ESCs, suggesting that Tcf3 down-regulation is a necessary step towards Wnt-mediated suppression of neural differentiation. We found that Tcf3 down-regulation in the context of constitutively active Wnt signaling does not result from promoter DNA methylation but is likely to be caused by a plethora of mechanisms at both the RNA and protein level as shown by the observed decrease in activating histone marks (H3K4me3 and H3-acetylation) and the upregulation of miR-211, a novel Wnt-regulated microRNA that targets Tcf3 and attenuates early neural differentiation in mouse ESCs. Our data show for the first time that Wnt signaling down-regulates Tcf3 expression, possibly at both the transcriptional and post-transcriptional levels, and thus highlight a novel mechanism through which Wnt signaling inhibits neuro-ectodermal lineage differentiation in mouse embryonic stem cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Células Madre Embrionarias/fisiología , Vía de Señalización Wnt , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Linaje de la Célula , Metilación de ADN , Regulación hacia Abajo , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Ratones , Mutación , Transcripción Genética
3.
Carcinogenesis ; 35(1): 2-13, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23955540

RESUMEN

Wnt signaling plays a central role in mammary stem cell (MaSC) homeostasis and in breast cancer. In particular, epigenetic alterations at different members of the Wnt pathway have been identified among triple-negative, basal-like breast cancers. Previously, we developed a mouse model for metaplastic breast adenocarcinoma, a subtype of triple-negative breast cancer, by targeting a hypomorphic mutations in the endogenous Apc gene (Apc (1572T/+)). Here, by employing the CD24 and CD29 cell surface antigens, we have identified a subpopulation of mammary cancer stem cells (MaCSCs) from Apc (1572T/+) capable of self-renewal and differentiation both in vivo and in vitro. Moreover, immunohistochemical analysis of micro- and macrolung metastases and preliminary intravenous transplantation assays suggest that the MaCSCs underlie metastasis at distant organ sites. Expression profiling of the normal and tumor cell subpopulations encompassing MaSCs and CSCs revealed that the normal stem cell compartment is more similar to tumor cells than to their own differentiated progenies. Accordingly, Wnt signaling appears to be active in both the normal and cancer stem cell compartments, although at different levels. By comparing normal with cancer mouse mammary compartments, we identified a MaCSC gene signature able to predict outcome in breast cancer in man. Overall, our data indicate that constitutive Wnt signaling activation affects self-renewal and differentiation of MaSCs leading to metaplasia and basal-like adenocarcinomas.


Asunto(s)
Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Células Madre Neoplásicas/patología , Vía de Señalización Wnt/fisiología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Antígeno CD24/metabolismo , Diferenciación Celular , Femenino , Humanos , Integrina beta1/metabolismo , Glándulas Mamarias Animales/citología , Ratones , Ratones Transgénicos , Células Madre Neoplásicas/metabolismo , Valor Predictivo de las Pruebas , Valores de Referencia , Transcriptoma , Proteína Wnt1/genética , Proteína Wnt1/metabolismo
4.
J Pathol ; 230(1): 48-58, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23288720

RESUMEN

Endometrioid endometrial cancer arises through a gradual series of histological changes, each accompanied by specific alterations in gene expression and activity. Activation of the Wnt-ß-catenin pathway and loss of PTEN activity are frequently observed in endometrial cancers. However, the specific roles played by alterations in these pathways in the initiation and progression of endometrial cancer are currently unclear. Here, we investigated the effects of loss of Pten and Apc gene function in the mouse endometrium by employing tissue-specific and inducible mutant alleles, followed by immunohistochemical (IHC) and loss of heterozygosity (LOH) analysis of their corresponding cancerous lesions. Loss of the Apc function in the endometrium leads to cytoplasmic and nuclear ß-catenin accumulation in association with uterine hyperplasia and squamous cell metaplasia, but without malignant transformation. Loss of Pten function also resulted in squamous metaplasia but, in contrast to loss of Apc function, it initiates endometrial cancer. On the other hand, loss of Apc function in the endometrium accelerates Pten-driven endometrial tumourigenesis. Analysis of compound heterozygous mice confirmed that somatic loss of the wild-type Pten allele represents the rate-limiting initiation step in endometrial cancer. Simultaneous loss of Pten and Apc resulted in endometrial cancer characterized by earlier onset and a more aggressive malignant behaviour. These observations are indicative of the synergistic action between the Wnt-ß-catenin and Pten signalling pathways in endometrial cancer onset and progression.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Neoplasias Endometriales/metabolismo , Fosfohidrolasa PTEN/metabolismo , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo , Animales , Carcinoma de Células Escamosas/patología , Progresión de la Enfermedad , Neoplasias Endometriales/patología , Femenino , Eliminación de Gen , Silenciador del Gen , Genes APC/fisiología , Pérdida de Heterocigocidad/genética , Masculino , Ratones , Ratones Noqueados , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Útero/anomalías , Útero/metabolismo
5.
Carcinogenesis ; 34(11): 2629-38, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23764752

RESUMEN

Whereas aberrant activation of canonical Wnt/ß-catenin signaling underlies the majority of colorectal cancer cases, the contribution of non-canonical Wnt signaling is unclear. As enhanced expression of the most extensively studied non-canonical Wnt ligand WNT5A is observed in various diseases including colon cancer, WNT5A is gaining attention nowadays. Numerous in vitro studies suggest modulating capacities of WNT5A on proliferation, differentiation, migration and invasion, affecting tumor and non-mutant cells. However, a possible contribution of WNT5A to colorectal cancer remains to be elucidated. We have analyzed WNT5A expression in colorectal cancer profiling data sets, altered WNT5A expression in colon cancer cells and used our inducible Wnt5a transgenic mouse model to gain more insight into the role of WNT5A in intestinal cancer. We observed that increased WNT5A expression is associated with poor prognosis of colorectal cancer patients. WNT5A knockdown in human colon cancer cells caused reduced directional migration, deregulated focal adhesion site formation and reduced invasion, whereas Wnt5a administration promoted the directional migration of colon cancer cells. Despite these observed protumorigenic activities of WNT5A, the induction of Wnt5a expression in intestinal tumors of Apc1638N mice was not sufficient to augment malignancy or metastasis by itself. In conclusion, WNT5A promotes adhesion sites to form in a focal fashion and promotes the directional migration and invasion of colon cancer cells. Although these activities appear insufficient by themselves to augment malignancy or metastasis in Apc1638N mice, they might explain the poor colon cancer prognosis associated with enhanced WNT5A expression.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/fisiología , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/patología , Neoplasias del Colon/patología , Intestinos/patología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Wnt/metabolismo , Animales , Apoptosis , Western Blotting , Adhesión Celular , Transformación Celular Neoplásica/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Adhesiones Focales , Humanos , Técnicas para Inmunoenzimas , Mucosa Intestinal/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Invasividad Neoplásica , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Proteínas Wnt/genética , Proteína Wnt-5a
6.
Dev Biol ; 369(1): 91-100, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22691362

RESUMEN

Wnt5a is essential during embryonic development, as indicated by mouse Wnt5a knockout embryos displaying outgrowth defects of multiple structures including the gut. The dynamics of Wnt5a involvement in these processes is unclear, and perinatal lethality of Wnt5a knockout embryos has hampered investigation of Wnt5a during postnatal stages in vivo. Although in vitro studies have suggested a relevant role for Wnt5a postnatally, solid evidence for a significant impact of Wnt5a within the complexity of an adult organism is lacking. We generated a tightly-regulated inducible Wnt5a transgenic mouse model and investigated the effects of Wnt5a induction during different time-frames of embryonic development and in adult mice, focusing on the gastrointestinal tract. When induced in embryos from 10.5 dpc onwards, Wnt5a expression led to severe outgrowth defects affecting the gastrointestinal tracts, limbs, facial structures and tails, closely resembling the defects observed in Wnt5a knockout mice. However, Wnt5a induction from 13.5 dpc onwards did not cause this phenotype, indicating that the most critical period for Wnt5a in embryonic development is prior to 13.5 dpc. In adult mice, induced Wnt5a expression did not reveal abnormalities, providing the first in vivo evidence that Wnt5a has no major impact on mouse intestinal homeostasis postnatally. Protein expression of Wnt5a receptor Ror2 was strongly reduced in adult intestine compared to embryonic stages. Moreover, we uncovered a regulatory process where induction of Wnt5a causes downregulation of its receptor Ror2. Taken together, our results indicate a role for Wnt5a during a restricted time-frame of embryonic development, but suggest no impact during homeostatic postnatal stages.


Asunto(s)
Envejecimiento/genética , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Intestinos/embriología , Proteínas Wnt/metabolismo , Envejecimiento/efectos de los fármacos , Animales , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Desarrollo Embrionario/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Mucosa Intestinal/metabolismo , Intestinos/citología , Intestinos/efectos de los fármacos , Ratones , Ratones Transgénicos , Modelos Animales , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tetraciclina/farmacología , Proteínas Wnt/genética , Proteína Wnt-5a
7.
BMC Dev Biol ; 12: 18, 2012 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-22690876

RESUMEN

BACKGROUND: To facilitate the in vivo study of esophageal (stem) cell biology in homeostasis and cancer, novel mouse models are necessary to elicit expression of candidate genes in a tissue-specific and inducible fashion. To this aim, we developed and studied a mouse model to allow labeling of esophageal cells with the histone 2B-GFP (H2B-GFP) fusion protein. RESULTS: First, we generated a transgenic mouse model expressing the reverse tetracycline transactivator rtTA2-M2 under control of the promoter (ED-L2) of the Epstein-Barr virus (EBV) gene encoding the latent membrane protein-1 (LMP-1). The newly generated ED-L2-rtTA2-M2 (ED-L2-rtTA) mice were then bred with the previously developed tetO-HIST1H2BJ/GFP (tetO-H2B-GFP) model to assess inducibility and tissue-specificity. Expression of the H2B-GFP fusion protein was observed upon doxycycline induction but was restricted to the terminally differentiated cells above the basal cell layer. To achieve expression in the basal compartment of the esophagus, we subsequently employed a different transgenic model expressing the reverse transactivator rtTA2S-M2 under the control of the ubiquitous, methylation-free CpG island of the human hnRNPA2B1-CBX3 gene (hnRNP-rtTA). Upon doxycycline administration to the compound hnRNP-rtTA/tetO-H2B-GFP mice, near-complete labeling of all esophageal cells was achieved. Pulse-chase experiments confirmed that complete turnover of the esophageal epithelium in the adult mouse is achieved within 7-10 days. CONCLUSIONS: We show that the esophagus-specific promoter ED-L2 is expressed only in the differentiated cells above the basal layer. Moreover, we confirmed that esophageal turn-over in the adult mouse does not exceed 7-10 days.


Asunto(s)
Esófago/metabolismo , Ingeniería Genética , Activación Transcripcional , Animales , Núcleo Celular/metabolismo , Proteínas Cromosómicas no Histona/genética , Epitelio/metabolismo , Esófago/citología , Femenino , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Histonas/biosíntesis , Histonas/genética , Humanos , Queratinas/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Transactivadores/genética
8.
PLoS Genet ; 5(7): e1000547, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19578404

RESUMEN

Germline mutations in the adenomatous polyposis coli (APC) gene are responsible for familial adenomatous polyposis (FAP), an autosomal dominant hereditary predisposition to the development of multiple colorectal adenomas and of a broad spectrum of extra-intestinal tumors. Moreover, somatic APC mutations play a rate-limiting and initiating role in the majority of sporadic colorectal cancers. Notwithstanding its multifunctional nature, the main tumor suppressing activity of the APC gene resides in its ability to regulate Wnt/beta-catenin signaling. Notably, genotype-phenotype correlations have been established at the APC gene between the length and stability of the truncated proteins encoded by different mutant alleles, the corresponding levels of Wnt/beta-catenin signaling activity they encode for, and the incidence and distribution of intestinal and extra-intestinal tumors. Here, we report a novel mouse model, Apc1572T, obtained by targeting a truncated mutation at codon 1572 in the endogenous Apc gene. This hypomorphic mutant allele results in intermediate levels of Wnt/beta-catenin signaling activation when compared with other Apc mutations associated with multifocal intestinal tumors. Notwithstanding the constitutive nature of the mutation, Apc(+/1572T) mice have no predisposition to intestinal cancer but develop multifocal mammary adenocarcinomas and subsequent pulmonary metastases in both genders. The histology of the Apc1572T primary mammary tumours is highly heterogeneous with luminal, myoepithelial, and squamous lineages and is reminiscent of metaplastic carcinoma of the breast in humans. The striking phenotype of Apc(+/1572T) mice suggests that specific dosages of Wnt/beta-catenin signaling activity differentially affect tissue homeostasis and initiate tumorigenesis in an organ-specific fashion.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/genética , Marcación de Gen , Mutación de Línea Germinal , Neoplasias Intestinales/genética , Neoplasias Mamarias Animales/genética , Poliposis Adenomatosa del Colon/metabolismo , Poliposis Adenomatosa del Colon/patología , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Masculino , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense , Especificidad de Órganos , Transducción de Señal , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
9.
Gut ; 60(9): 1204-12, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21307168

RESUMEN

Objective Deregulation of the Wnt signalling pathway by mutations in the Apc or ß-catenin genes underlies colorectal carcinogenesis. As a result, ß-catenin stabilises, translocates to the nucleus, and activates gene transcription. Intestinal tumours show a heterogeneous pattern of nuclear ß-catenin, with the highest levels observed at the invasion front. Activation of receptor tyrosine kinases in these tumour areas by growth factors expressed by surrounding stromal cells phosphorylate ß-catenin at tyrosine residues, which is thought to increase ß-catenin nuclear translocation and tumour invasiveness. This study investigates the relevance of ß-catenin tyrosine phosphorylation for Wnt signalling and intestinal tumorigenesis in vivo. Design A conditional knock-in mouse model was generated into which the phospho-mimicking Y654E modification in the endogenous ß-catenin gene was introduced. Results This study provided in vivo evidence that ß-catenin(E654) is characterised by reduced affinity for cadherins, increased signalling and strongly increased phosphorylation at serine 675 by protein kinase A (PKA). In addition, homozygosity for the ß-catenin(E654) targeted allele caused embryonic lethality, whereas heterozygosity predisposed to intestinal tumour development, and strongly enhanced Apc-driven intestinal tumour initiation associated with increased nuclear accumulation of ßcatenin. Surprisingly, the expression of ß-catenin(E654) did not affect histological grade or induce tumour invasiveness. Conclusions A thus far unknown mechanism was uncovered in which Y654 phosphorylation of ß-catenin facilitates additional phosphorylation at serine 675 by PKA. In addition, in contrast to the current belief that ß-catenin Y654 phosphorylation increases tumour progression to a more invasive phenotype, these results show that it rather increases tumour initiation by enhancing Wnt signalling.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Neoplasias Colorrectales/metabolismo , Proteínas Wnt/fisiología , beta Catenina/metabolismo , Adenoma/genética , Adenoma/metabolismo , Animales , Células COS , Cadherinas/metabolismo , Membrana Celular/metabolismo , Transformación Celular Neoplásica/genética , Chlorocebus aethiops , Neoplasias Colorrectales/genética , Proteínas Quinasas Dependientes de AMP Cíclico/farmacología , Pérdida del Embrión/genética , Técnicas de Sustitución del Gen , Genes APC , Genotipo , Heterocigoto , Homocigoto , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Fosforilación/fisiología
10.
J Pathol ; 221(4): 379-89, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20549647

RESUMEN

Accumulating evidence has suggested that tumours have a hierarchical organization in which only the cancer stem cells (CSCs) have tumour-initiating properties. Several surface antigens have been employed to isolate CSCs from various malignancies, although not from oesophageal adenocarcinoma (EA). We tested whether Barrett's oesophagus (BE) and EA might serve as a model for the CSC concept. In vivo assays were performed by transplantation of serially diluted bulk EA cells into NOD-SCID mice to establish the presence and frequency of tumour-initiating cells. These were found to be present as ca. 1 in 64 000 cells. The transplanted tumours fully recapitulated the primary lesions. Subsequently, a panel of previously established CSC markers was employed for immunohistochemistry. CD24, CD29 and CD44 showed heterogeneous staining in EA. Nuclear beta-catenin accumulation increased during progression from metaplasia to dysplasia and was often observed in the basal compartment with CD24 and CD29 staining. However, the overall staining patterns were not such to clearly point out specific candidate markers. Accordingly, all markers were employed to sort the corresponding subpopulations of cancer cells and transplant them at low multiplicities in NOD-SCID mice. No increased tumour-initiating capacity of sorted EA cells was observed upon transplantation. These results indicate that tumour-initiating cells are present in EA, thus reflecting a hierarchical organization. However, antibodies directed against novel surface antigens are needed to detect subpopulations enriched for CSCs in EA by transplantation assays.


Asunto(s)
Adenocarcinoma/patología , Esófago de Barrett/patología , Biomarcadores de Tumor/metabolismo , Neoplasias Esofágicas/patología , Células Madre Neoplásicas/patología , Adenocarcinoma/metabolismo , Animales , Esófago de Barrett/metabolismo , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Progresión de la Enfermedad , Neoplasias Esofágicas/metabolismo , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias , Células Madre Neoplásicas/metabolismo , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología
11.
Cancer Res ; 81(7): 1775-1787, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33531370

RESUMEN

Although immune checkpoint blockade (ICB) has shown remarkable clinical benefit in a subset of patients with melanoma and lung cancer, most patients experience no durable benefit. The receptor tyrosine kinase AXL is commonly implicated in therapy resistance and may serve as a marker for therapy-refractory tumors, for example in melanoma, as we previously demonstrated. Here, we show that enapotamab vedotin (EnaV), an antibody-drug conjugate targeting AXL, effectively targets tumors that display insensitivity to immunotherapy or tumor-specific T cells in several melanoma and lung cancer models. In addition to its direct tumor cell killing activity, EnaV treatment induced an inflammatory response and immunogenic cell death in tumor cells and promoted the induction of a memory-like phenotype in cytotoxic T cells. Combining EnaV with tumor-specific T cells proved superior to either treatment alone in models of melanoma and lung cancer and induced ICB benefit in models otherwise insensitive to anti-PD-1 treatment. Our findings indicate that targeting AXL-expressing, immunotherapy-resistant tumors with EnaV causes an immune-stimulating tumor microenvironment and enhances sensitivity to ICB, warranting further investigation of this treatment combination. SIGNIFICANCE: These findings show that targeting AXL-positive tumor fractions with an antibody-drug conjugate enhances antitumor immunity in several humanized tumor models of melanoma and lung cancer.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoconjugados/uso terapéutico , Neoplasias Pulmonares/terapia , Melanoma/terapia , Proteínas Proto-Oncogénicas/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Terapia Combinada , Resistencia a Antineoplásicos/inmunología , Sinergismo Farmacológico , Células HEK293 , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Inmunoconjugados/administración & dosificación , Inmunoterapia , Neoplasias Pulmonares/patología , Masculino , Melanoma/patología , Ratones , Ratones Desnudos , Ratones Transgénicos , Terapia Molecular Dirigida/métodos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Tirosina Quinasa del Receptor Axl
12.
Hum Mutat ; 31(11): 1205-15, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20725929

RESUMEN

The MUTYH gene encodes a DNA glycosylase involved in base excision repair (BER). Biallelic pathogenic MUTYH variants have been associated with colorectal polyposis and cancer. The pathogenicity of a few variants is beyond doubt, including c.536A4G/p.Tyr179Cys and c.1187G4A/p.Gly396Asp (previously c.494A4G/p.Tyr165Cys and c.1145G4A/p.Gly382Asp).However, for a substantial fraction of the detected variants, the clinical significance remains uncertain,compromising molecular diagnostics and thereby genetic counseling. We have established an interactive MUTYH gene sequence variant database (www.lovd.nl/MUTYH) with the aim of collecting and sharing MUTYH genotype and phenotype data worldwide. To support standard variant description, we chose NM_001128425.1 as the reference sequence. The database includes records with variants per individual, linked to available phenotype and geographic origin data as well as records with in vitro functional and in silico test data. As of April 2010, the database contains 1968 published and 423 unpublished submitted entries, and 230 and 61 unique variants,respectively. This open-access repository allows all involved to quickly share all variants encountered and communicate potential consequences, which will be especially useful to classify variants of uncertain significance.


Asunto(s)
ADN Glicosilasas/genética , Bases de Datos Genéticas , Variación Genética , Poliposis Adenomatosa del Colon/genética , Empalme Alternativo , Secuencia de Aminoácidos , Secuencia de Bases , ADN/genética , ADN Glicosilasas/química , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Datos de Secuencia Molecular , Mutación , Países Bajos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estructura Terciaria de Proteína
13.
Genesis ; 47(1): 7-13, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18942097

RESUMEN

To develop a sensitive and inducible system to study intestinal biology, we generated a transgenic mouse model expressing the reverse tetracycline transactivator rtTA2-M2 under control of the 12.4 kb murine Villin promoter. The newly generated Villin-rtTA2-M2 mice were then bred with the previously developed tetO-HIST1H2BJ/GFP model to assess inducibility and tissue-specificity. Expression of the histone H2B-GFP fusion protein was observed exclusively upon doxycycline induction and was uniformly distributed throughout the intestinal epithelium. The Villin-rtTA2-M2 was also found to drive transgene expression in the developing mouse intestine. Furthermore, we could detect transgene expression in the proximal tubules of the kidney and in a population of alleged gastric progenitor cells. By administering different concentrations of doxycycline, we show that the Villin-rtTA2-M2 system drives transgene expression in a dosage-dependent fashion. Thus, we have generated a novel doxycycline-inducible mouse model, providing a valuable tool to study the effect of different gene dosages on intestinal physiology and pathology.


Asunto(s)
Doxiciclina/farmacología , Expresión Génica/efectos de los fármacos , Ingeniería Genética/métodos , Intestinos/fisiología , Animales , Genes Reporteros/genética , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Ratones , Ratones Transgénicos , Especificidad de Órganos , Investigación , Volumetría , Transgenes/genética
14.
Am J Pathol ; 172(5): 1363-80, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18403596

RESUMEN

Expression profiling is a well established tool for the genome-wide analysis of human cancers. However, the high sensitivity of this approach combined with the well known cellular and molecular heterogeneity of cancer often result in extremely complex expression signatures that are difficult to interpret functionally. The majority of sporadic colorectal cancers are triggered by mutations in the adenomatous polyposis coli (APC) tumor suppressor gene, leading to the constitutive activation of the Wnt/beta-catenin signaling pathway and formation of adenomas. Despite this common genetic basis, colorectal cancers are very heterogeneous in their degree of differentiation, growth rate, and malignancy potential. Here, we applied a cross-species comparison of expression profiles of intestinal polyps derived from hereditary colorectal cancer patients carrying APC germline mutations and from mice carrying a targeted inactivating mutation in the mouse homologue Apc. This comparative approach resulted in the establishment of a conserved signature of 166 genes that were differentially expressed between adenomas and normal intestinal mucosa in both species. Functional analyses of the conserved genes revealed a general increase in cell proliferation and the activation of the Wnt/beta-catenin signaling pathway. Moreover, the conserved signature was able to resolve expression profiles from hereditary polyposis patients carrying APC germline mutations from those with bi-allelic inactivation of the MYH gene, supporting the usefulness of such comparisons to discriminate among patients with distinct genetic defects.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Poliposis Adenomatosa del Colon/metabolismo , Transformación Celular Neoplásica/metabolismo , Neoplasias Colorrectales/patología , Pólipos Intestinales/metabolismo , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Transformación Celular Neoplásica/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Perfilación de la Expresión Génica , Humanos , Pólipos Intestinales/genética , Pólipos Intestinales/patología , Ratones , Mutación , Transducción de Señal , Especificidad de la Especie
15.
Acta Histochem ; 109(4): 266-72, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17445872

RESUMEN

Long-standing ulcerative colitis (UC) has been associated with a high risk of developing colonic adenocarcinoma. Importantly, both low- and high-grade dysplasia are strongly related to the presence or development of malignancy. The canonical Wnt/beta-catenin signaling pathway is of crucial importance in cancer development and progression, but its role in UC-related carcinogenesis remains to be determined. We evaluated the immunolabeling patterns of beta-catenin, as well as the products of Wnt-associated cancer genes E-cadherin, cyclin D1 and c-myc, along the dysplasia-carcinoma pathway in UC. For this purpose, immunohistochemistry (IHC) was performed on 18 adenocarcinomas and 17 dysplasias, derived from 21 patients. We found that intracellular beta-catenin accumulation, the hallmark of Wnt signaling activation, is observed in dysplasia, together with enhanced labeling of nuclear protein cyclin D1 and reduction of membranous labeling of E-cadherin. c-myc displayed moderate immunolabeling in the (pre)malignant lesions. Thus, the Wnt pathway is activated in early stages of malignant progression in UC. Furthermore, upregulation of the oncogene cyclin D1 and downregulation of tumor suppressor E-cadherin also occurs in the (pre)neoplastic state. This may contribute to the high potential for malignant degeneration of dysplasia in UC-related colitis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/patología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Proteínas Wnt/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Transformación Celular Neoplásica/genética , Colitis Ulcerosa/genética , Neoplasias del Colon/genética , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Transducción de Señal , Proteínas Wnt/genética
16.
J Clin Oncol ; 23(13): 3074-8, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15860866

RESUMEN

PURPOSE: TGFBR16A is a tumor susceptibility gene that increases breast, colon, and ovarian cancer risk. Fourteen percent of the general population carries TGFBR16A, and TGFBR16A homozygotes have a greater than 100% increased colon cancer risk compared with noncarriers. Low-penetrance genes such as TGFBR16A may account for a sizable proportion of familial colorectal cancer occurrences. To test this hypothesis, we determined whether TGFBR16A contributes to a proportion of mismatch repair (MMR) gene mutation-negative hereditary nonpolyposis colorectal cancer (HNPCC) patients. PATIENTS AND METHODS: A case-case study was performed of 208 index patients with HNPCC meeting the Amsterdam criteria. Patients were examined for mutations and genomic rearrangements in the MLH1, MSH2, and MSH6 genes and genotyped for TGFBR16A. Tumor microsatellite instability status was available for 95 patients. RESULTS: A total of 144 patients (69.2%) carried a deleterious mutation and were classified as positive for MMR gene mutation; 64 patients (30.8%) had no evidence of mutations and were classified as MMR negative. TGFBR16A allelic frequency was significantly higher among MMR-negative patients (0.195) than among MMR-positive patients (0.104; P = .011). The proportion of TGFBR16A homozygotes was nine-fold higher among MMR-negative (6.3%) than among MMR-positive patients (0.7%; P = .032). The highest TGFBR16A allelic frequency was found among MMR-negative patients with tumors exhibiting no microsatellite instability (0.211), and the lowest frequency was found among MMR-positive patients with tumors exhibiting microsatellite instability (0.121); the difference was not statistically significant (P = .17). CONCLUSION: TGFBR16A may be causally responsible for a proportion of HNPCC occurrences.


Asunto(s)
Receptores de Activinas Tipo I/genética , Neoplasias Colorrectales/genética , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Receptores de Factores de Crecimiento Transformadores beta/genética , Adulto , Disparidad de Par Base , Estudios de Casos y Controles , Análisis Mutacional de ADN , Reparación del ADN , Femenino , Genotipo , Humanos , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Proteínas Serina-Treonina Quinasas , Receptor Tipo I de Factor de Crecimiento Transformador beta
17.
Virchows Arch ; 449(6): 716-21, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17091253

RESUMEN

Long-standing ulcerative colitis is associated with an elevated risk of developing colonic adenocarcinoma. A very limited group of patients present with multiple synchronous cancers. This could be due to either a multifocal presentation of the same neoplastic clone or different tumors arising in a large area of polyclonal dysplastic colonic mucosa ("field cancerization"). Here, we describe a patient with long-standing colitis and three different tumors in the rectosigmoid part of the large bowel. Clonal evaluation of the lesions was performed by array-based comparative genomic hybridization. These three neoplasms showed a comparable pattern of genomic alterations characterized by gains of chromosomes 12, 13, and 20. Noteworthy, dysplastic mucosa distal to the three cancers displayed a completely different pattern of genomic changes indicating that different cell lineages were present. In addition, all three carcinomas were microsatellite stable and revealed identical immunoprofiles for several cancer-associated genes. We conclude that these three multifocal tumors must have originated from the same preneoplastic lineage.


Asunto(s)
Adenocarcinoma/genética , Colitis Ulcerosa/complicaciones , Neoplasias del Colon/genética , Adenocarcinoma/etiología , Anciano , Anciano de 80 o más Años , Aberraciones Cromosómicas , Neoplasias del Colon/etiología , Femenino , Humanos , Inestabilidad de Microsatélites , Hibridación de Ácido Nucleico
18.
Eur J Cancer ; 68: 114-124, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27750112

RESUMEN

INTRODUCTION: S100a4 is a calcium-binding protein belonging to the family of S100-proteins, highly expressed in different stromal cell types. S100A4 has been reported as a prognostic marker in colorectal cancer in association with tumour progression and metastasis. METHODS: In this study, we analysed the in vivo role of S100a4 in intestinal tumour initiation and progression using different transgenic and knockout mouse models. RESULTS: We found that genetic ablation or overexpression of S100a4 in both Apc- and Smad4-mutant mice do not affect tumour initiation in the intestinal tract. In contrast, S100a4 epithelial overexpression in Apc1638N/+/KRASV12G mice increases the dissemination of intestinal tumour cells to the liver, in agreement with its role in tumour metastasis. Moreover, we report a novel role for S100a4 in desmoid formation where S100a4 deficiency results in a significant reduction of the tumour burden characteristic of the Apc1638N model. In agreement with these results, S100a4 appears to be co-expressed together with mesenchymal stem cell (MSC) markers in desmoid tumours from Apc1638N/+ mice, as well as from sporadic and hereditary human desmoids. CONCLUSION: Our data provide the first report on the in vivo role of S100a4 in intestinal tumourigenesis and describe a new role for S100a4 in the aetiology of desmoids formation.


Asunto(s)
Carcinogénesis/genética , Neoplasias Colorrectales/genética , Fibromatosis Agresiva/genética , Proteína de Unión al Calcio S100A4/genética , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Neoplasias Colorrectales/metabolismo , Técnicas de Sustitución del Gen , Humanos , Neoplasias Intestinales/genética , Ratones , Ratones Noqueados , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína de Unión al Calcio S100A4/metabolismo , Proteína Smad4/genética
19.
Cell Stem Cell ; 19(1): 38-51, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27292189

RESUMEN

The intestinal stem cell niche provides cues that actively maintain gut homeostasis. Dysregulation of these cues may compromise intestinal regeneration upon tissue insult and/or promote tumor growth. Here, we identify secreted phospholipases A2 (sPLA2s) as stem cell niche factors with context-dependent functions in the digestive tract. We show that group IIA sPLA2, a known genetic modifier of mouse intestinal tumorigenesis, is expressed by Paneth cells in the small intestine, while group X sPLA2 is expressed by Paneth/goblet-like cells in the colon. During homeostasis, group IIA/X sPLA2s inhibit Wnt signaling through intracellular activation of Yap1. However, upon inflammation they are secreted into the intestinal lumen, where they promote prostaglandin synthesis and Wnt signaling. Genetic ablation of both sPLA2s improves recovery from inflammation but increases colon cancer susceptibility due to release of their homeostatic Wnt-inhibitory role. This "trade-off" effect suggests sPLA2s have important functions as genetic modifiers of inflammation and colon cancer.


Asunto(s)
Fosfolipasas A2 Grupo II/metabolismo , Fosfolipasas A2 Grupo X/metabolismo , Homeostasis , Inflamación/patología , Neoplasias Intestinales/enzimología , Neoplasias Intestinales/patología , Intestinos/patología , Nicho de Células Madre , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular , Diferenciación Celular , Linaje de la Célula , Dinoprostona/biosíntesis , Inflamación/enzimología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Neoplasias Intestinales/genética , Espacio Intracelular/metabolismo , Ratones Endogámicos C57BL , Organoides/metabolismo , Células de Paneth/enzimología , Células de Paneth/patología , Fosfoproteínas/metabolismo , Fosforilación , Células Madre/patología , Vía de Señalización Wnt , Proteínas Señalizadoras YAP
20.
J Mol Diagn ; 5(3): 160-7, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12876206

RESUMEN

At present, limiting factors in the use of tissue microarrays (TMAs) for high-throughput analysis relate to the visual evaluation of the staining patterns of each of the individual cores in the array and to the subsequent input of the results into a database. Such a database is essential to correlate the data with tumor type and outcome, and to evaluate the performance against other markers achieved in separate experiments. So far, these steps are mostly performed by hand, and consequently are time-consuming and potentially prone to bias and errors, respectively. This paper describes the use of a high-resolution flat-bed scanner for digitization of TMAs with a resolution of about 5 x 5 micro m(2). The arrays are acquired, the positions of the tissue cores are automatically determined, and measurement data including the images of the individual cores are archived. The program provides digital zooming of arrays for interactive verification of the results and rapid linkage of individual core images to data sets of other markers derived from the same array. Performance of the system was compared to manual classification for a representative set of arrays containing colorectal tumors stained with different markers.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Automatización , Neoplasias Colorrectales Hereditarias sin Poliposis/patología , Humanos , Procesamiento de Imagen Asistido por Computador
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA