Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Pathol Clin Res ; 9(1): 73-85, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36349502

RESUMEN

Tumor stem cells play a pivotal role in carcinogenesis and metastatic spread in colorectal cancer (CRC). Olfactomedin 4 (OLFM4) is co-expressed with the established stem cell marker leucine-rich repeat-containing G protein-coupled receptor 5 at the bottom of intestinal crypts and has been suggested as a surrogate for cancer stemness and a biomarker in gastrointestinal tumors associated with prognosis. Therefore, it was the aim of the present study to clarify whether OLFM4 is involved in carcinogenesis and metastatic spread in CRC. We used a combined approach of functional assays using forced OLFM4 overexpression in human CRC cell lines, xenograft mice, and an immunohistochemical approach using patient tissues to investigate the impact of OLFM4 on stemness, canonical Wnt signaling, properties of metastasis and differentiation as well as prognosis. OLFM4 expression correlated weakly with tumor grade in one patient cohort (metastasis collection: p = 0.05; pooled analysis of metastasis collection and survival collection: p = 0.19) and paralleled the expression of differentiation markers (FABP2, MUC2, and CK20) (p = 0.002) but did not correlate with stemness-associated markers. Further analyses in CRC cells lines as well as xenograft mice including forced overexpression of OLFM4 revealed that OLFM4 neither altered the expression of markers of stemness nor epithelial-mesenchymal transition, nor did OLFM4 itself drive proliferation, migration, or colony formation, which are all prerequisites of carcinogenesis and tumor progression. In line with this, we found no significant correlation between OLFM4 expression, metastasis, and patient survival. In summary, expression of OLFM4 in human CRC seems to be characteristic of differentiation marker expression in CRC but is not a driver of carcinogenesis nor metastatic spread.


Asunto(s)
Antígenos de Diferenciación , Neoplasias Colorrectales , Factor Estimulante de Colonias de Granulocitos , Animales , Humanos , Ratones , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Transición Epitelial-Mesenquimal/genética , Factor Estimulante de Colonias de Granulocitos/genética , Factor Estimulante de Colonias de Granulocitos/metabolismo , Células Madre Neoplásicas/metabolismo
2.
Genes Dev ; 29(10): 1074-86, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25956904

RESUMEN

Colorectal cancer (CRC) remains a major contributor to cancer-related mortality. LIN28A and LIN28B are highly related RNA-binding protein paralogs that regulate biogenesis of let-7 microRNAs and influence development, metabolism, tissue regeneration, and oncogenesis. Here we demonstrate that overexpression of either LIN28 paralog cooperates with the Wnt pathway to promote invasive intestinal adenocarcinoma in murine models. When LIN28 alone is induced genetically, half of the resulting tumors harbor Ctnnb1 (ß-catenin) mutation. When overexpressed in Apc(Min/+) mice, LIN28 accelerates tumor formation and enhances proliferation and invasiveness. In conditional genetic models, enforced expression of a LIN28-resistant form of the let-7 microRNA reduces LIN28-induced tumor burden, while silencing of LIN28 expression reduces tumor volume and increases tumor differentiation, indicating that LIN28 contributes to tumor maintenance. We detected aberrant expression of LIN28A and/or LIN28B in 38% of a large series of human CRC samples (n = 595), where LIN28 expression levels were associated with invasive tumor growth. Our late-stage CRC murine models and analysis of primary human tumors demonstrate prominent roles for both LIN28 paralogs in promoting CRC growth and progression and implicate the LIN28/let-7 pathway as a therapeutic target.


Asunto(s)
Adenocarcinoma/fisiopatología , Neoplasias Colorrectales/fisiopatología , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Invasividad Neoplásica/genética , Invasividad Neoplásica/fisiopatología , Proteínas de Unión al ARN/genética
4.
Nature ; 514(7523): 508-12, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25174708

RESUMEN

Several features common to a Western lifestyle, including obesity and low levels of physical activity, are known risk factors for gastrointestinal cancers. There is substantial evidence suggesting that diet markedly affects the composition of the intestinal microbiota. Moreover, there is now unequivocal evidence linking dysbiosis to cancer development. However, the mechanisms by which high-fat diet (HFD)-mediated changes in the microbial community affect the severity of tumorigenesis in the gut remain to be determined. Here we demonstrate that an HFD promotes tumour progression in the small intestine of genetically susceptible, K-ras(G12Dint), mice independently of obesity. HFD consumption, in conjunction with K-ras mutation, mediated a shift in the composition of the gut microbiota, and this shift was associated with a decrease in Paneth-cell-mediated antimicrobial host defence that compromised dendritic cell recruitment and MHC class II molecule presentation in the gut-associated lymphoid tissues. When butyrate was administered to HFD-fed K-ras(G12Dint) mice, dendritic cell recruitment in the gut-associated lymphoid tissues was normalized, and tumour progression was attenuated. Importantly, deficiency in MYD88, a signalling adaptor for pattern recognition receptors and Toll-like receptors, blocked tumour progression. The transfer of faecal samples from HFD-fed mice with intestinal tumours to healthy adult K-ras(G12Dint) mice was sufficient to transmit disease in the absence of an HFD. Furthermore, treatment with antibiotics completely blocked HFD-induced tumour progression, suggesting that distinct shifts in the microbiota have a pivotal role in aggravating disease. Collectively, these data underscore the importance of the reciprocal interaction between host and environmental factors in selecting a microbiota that favours carcinogenesis, and they suggest that tumorigenesis is transmissible among genetically predisposed individuals.


Asunto(s)
Carcinogénesis/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/efectos adversos , Disbiosis/inducido químicamente , Disbiosis/microbiología , Neoplasias Intestinales/microbiología , Obesidad , Animales , Antibacterianos/farmacología , Butiratos/farmacología , Progresión de la Enfermedad , Mucosa Intestinal/inmunología , Neoplasias Intestinales/inducido químicamente , Intestinos/efectos de los fármacos , Intestinos/microbiología , Ratones , Obesidad/inducido químicamente , Obesidad/microbiología , Prebióticos
5.
J Clin Invest ; 124(4): 1853-67, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24642471

RESUMEN

Members of the miR-34 family are induced by the tumor suppressor p53 and are known to inhibit epithelial-to-mesenchymal transition (EMT) and therefore presumably suppress the early phases of metastasis. Here, we determined that exposure of human colorectal cancer (CRC) cells to the cytokine IL-6 activates the oncogenic STAT3 transcription factor, which directly represses the MIR34A gene via a conserved STAT3-binding site in the first intron. Repression of MIR34A was required for IL-6-induced EMT and invasion. Furthermore, we identified the IL-6 receptor (IL-6R), which mediates IL-6-dependent STAT3 activation, as a conserved, direct miR-34a target. The resulting IL-6R/STAT3/miR-34a feedback loop was present in primary colorectal tumors as well as CRC, breast, and prostate cancer cell lines and associated with a mesenchymal phenotype. An active IL-6R/STAT3/miR-34a loop was necessary for EMT, invasion, and metastasis of CRC cell lines and was associated with nodal and distant metastasis in CRC patient samples. p53 activation in CRC cells interfered with IL-6-induced invasion and migration via miR-34a-dependent downregulation of IL6R expression. In Mir34a-deficient mice, colitis-associated intestinal tumors displayed upregulation of p-STAT3, IL-6R, and SNAIL and progressed to invasive carcinomas, which was not observed in WT animals. Collectively, our data indicate that p53-dependent expression of miR-34a suppresses tumor progression by inhibiting a IL-6R/STAT3/miR-34a feedback loop.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Retroalimentación Fisiológica , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Invasividad Neoplásica , Metástasis de la Neoplasia , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo
6.
J Gastroenterol ; 49(4): 618-27, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24566894

RESUMEN

Every cancer cell is "different"--within one and the same tumor, between different lesions originating from the same tumor, among different patients suffering from the same tumor type, and certainly between different tumor types. The complexity of tumor development, with its genetic, phenotypic and functional heterogeneity and plasticity within tumors and between primary tumors and metastases, underlies the unpredictable influences and stimuli of a tumor-associated inflammatory microenvironment, immune response, mechanical and metabolic stress, therapy-induced inflammation or interaction with microbiota. The stochastic and context dependent nature of these factors accounts for the difficulties to investigate the impact of resulting cell plasticity on tumor development, and justifies the challenge to prevent tumor recurrence. The emerging concept of cell plasticity and reciprocity (to change the phenotype by processing signals from the environment) throws more light on the actual complexity of tumor heterogeneity than can be expected solely from a unidirectional, classical cancer stem cell (CSC) model. To date, it remains widely unclear to what extent cell plasticity impacts tumor development, and it is difficult to assess by current methods. As a high tumor plasticity is likely to predict a poor outcome for patients, the future therapeutic challenge will be the development of personalized treatment strategies to predict and finally prevent cell plasticity in patients.


Asunto(s)
Mediadores de Inflamación , Neoplasias/patología , Células Madre Neoplásicas/patología , Fenotipo , Microambiente Tumoral , Desdiferenciación Celular , Epigénesis Genética , Humanos , Modelos Biológicos , Neoplasias/genética , Neoplasias/fisiopatología , Neoplasias/terapia
7.
Cell Stem Cell ; 12(6): 761-73, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23665120

RESUMEN

The Adenomatous Polyposis Coli (APC) gene is mutated in the majority of colorectal cancers (CRCs). Loss of APC leads to constitutively active WNT signaling, hyperproliferation, and tumorigenesis. Identification of pathways that facilitate tumorigenesis after APC loss is important for therapeutic development. Here, we show that RAC1 is a critical mediator of tumorigenesis after APC loss. We find that RAC1 is required for expansion of the LGR5 intestinal stem cell (ISC) signature, progenitor hyperproliferation, and transformation. Mechanistically, RAC1-driven ROS and NF-κB signaling mediate these processes. Together, these data highlight that ROS production and NF-κB activation triggered by RAC1 are critical events in CRC initiation.


Asunto(s)
Neoplasias Colorrectales/patología , Intestino Delgado/citología , FN-kappa B/metabolismo , Neuropéptidos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Células Madre/citología , Proteínas Wnt/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Intestino Delgado/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Células Madre/metabolismo
8.
Cancer Cell ; 23(1): 93-106, 2013 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-23273920

RESUMEN

Loss of p53 is considered to allow progression of colorectal tumors from the adenoma to the carcinoma stage. Using mice with an intestinal epithelial cell (IEC)-specific p53 deletion, we demonstrate that loss of p53 alone is insufficient to initiate intestinal tumorigenesis but markedly enhances carcinogen-induced tumor incidence and leads to invasive cancer and lymph node metastasis. Whereas p53 controls DNA damage and IEC survival during the initiation stage, loss of p53 during tumor progression is associated with increased intestinal permeability, causing formation of an NF-κB-dependent inflammatory microenvironment and the induction of epithelial-mesenchymal transition. Thus, we propose a p53-controlled tumor-suppressive function that is independent of its well-established role in cell-cycle regulation, apoptosis, and senescence.


Asunto(s)
Carcinógenos/toxicidad , Neoplasias Colorrectales/patología , Ganglios Linfáticos/patología , Microambiente Tumoral , Proteína p53 Supresora de Tumor/genética , Adenoma/inducido químicamente , Adenoma/genética , Adenoma/patología , Animales , Carcinoma/inducido químicamente , Carcinoma/genética , Carcinoma/patología , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Modelos Animales de Enfermedad , Ratones , Mutagénesis Sitio-Dirigida , Invasividad Neoplásica/genética , Metástasis de la Neoplasia
9.
Cell ; 152(1-2): 25-38, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23273993

RESUMEN

Cell-type plasticity within a tumor has recently been suggested to cause a bidirectional conversion between tumor-initiating stem cells and nonstem cells triggered by an inflammatory stroma. NF-κB represents a key transcription factor within the inflammatory tumor microenvironment. However, NF-κB's function in tumor-initiating cells has not been examined yet. Using a genetic model of intestinal epithelial cell (IEC)-restricted constitutive Wnt-activation, which comprises the most common event in the initiation of colon cancer, we demonstrate that NF-κB modulates Wnt signaling and show that IEC-specific ablation of RelA/p65 retards crypt stem cell expansion. In contrast, elevated NF-κB signaling enhances Wnt activation and induces dedifferentiation of nonstem cells that acquire tumor-initiating capacity. Thus, our data support the concept of bidirectional conversion and highlight the importance of inflammatory signaling for dedifferentiation and generation of tumor-initiating cells in vivo.


Asunto(s)
Desdiferenciación Celular , Transformación Celular Neoplásica , Neoplasias del Colon/patología , Células Madre Neoplásicas/patología , Animales , Colon/patología , Células Epiteliales/patología , Femenino , Humanos , Masculino , Ratones , FN-kappa B/metabolismo , Vía de Señalización Wnt
10.
Proc Natl Acad Sci U S A ; 108(16): 6567-72, 2011 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-21464320

RESUMEN

Loss of IκB kinase (IKK) ß-dependent NF-κB signaling in hematopoietic cells is associated with increased granulopoiesis. Here we identify a regulatory cytokine loop that causes neutrophilia in Ikkß-deficient mice. TNF-α-dependent apoptosis of myeloid progenitor cells leads to the release of IL-1ß, which promotes Th17 polarization of peripheral CD4(+) T cells. Although the elevation of IL-17 and the consecutive induction of granulocyte colony-stimulating factor compensate for the loss of myeloid progenitor cells, the facilitated induction of Th17 cells renders Ikkß-deficient animals more susceptible to the development of experimental autoimmune encephalitis. These results unravel so far unanticipated direct and indirect functions for IKKß in myeloid progenitor survival and maintenance of innate and Th17 immunity and raise concerns about long-term IKKß inhibition in IL-17-mediated diseases.


Asunto(s)
Quinasa I-kappa B/inmunología , Células Progenitoras Mieloides/inmunología , Mielopoyesis/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Apoptosis/genética , Apoptosis/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Encefalitis/genética , Encefalitis/inmunología , Encefalitis/metabolismo , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Interleucina-17/biosíntesis , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Ratones , Ratones Noqueados , Células Progenitoras Mieloides/metabolismo , Mielopoyesis/genética , FN-kappa B/genética , FN-kappa B/inmunología , FN-kappa B/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Células Th17/inmunología , Células Th17/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
11.
Clin Exp Metastasis ; 28(1): 75-90, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20981475

RESUMEN

Several data of the past years clearly indicated that the fusion of tumor cells and tumor cells or tumor cells and normal cells can give rise to hybrids cells exhibited novel properties such as an increased malignancy, drug resistance, or resistance to apoptosis. In the present study we characterized hybrid cells derived from spontaneous fusion events between the breast epithelial cell line M13SV1-EGFP-Neo and two breast cancer cell lines: HS578T-Hyg and MDA-MB-435-Hyg. Short-tandem-repeat analysis revealed an overlap of parental alleles in all hybrid cells indicating that hybrid cells originated from real cell fusion events. RealTime-PCR-array gene expression data provided evidence that each hybrid cell clone exhibited a unique gene expression pattern, resulting in a specific resistance of hybrid clones towards chemotherapeutic drugs, such as doxorubicin and paclitaxel, as well as a specific migratory behavior of hybrid clones towards EGF. For instance, M13MDA435-4 hybrids showed a marked resistance towards etoposide, doxorubicin and paclitaxel, whereas hybrid clones M13MDA-435-1 and -2 were only resistant towards etoposide. Likewise, all investigated M13MDA435 hybrids responded to EGF with an increased migratory activity, whereas the migration of parental MDA-MB-435-Hyg cells was blocked by EGF, suggesting that M13MDA435 hybrids may have acquired a new motility pathway. Similar findings have been obtained for M13HS hybrids. We conclude from our data that they further support the hypothesis that cell fusion could give rise to drug resistant and migratory active tumor (hybrid) cells in cancer.


Asunto(s)
Neoplasias de la Mama/patología , Fusión Celular , Células Epiteliales/patología , Neoplasias de la Mama/tratamiento farmacológico , Movimiento Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Etopósido/farmacología , Femenino , Citometría de Flujo , Fluorouracilo/uso terapéutico , Perfilación de la Expresión Génica , Humanos , Células Híbridas/patología , Paclitaxel/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Relación Estructura-Actividad , Células Tumorales Cultivadas
12.
Med Hypotheses ; 73(4): 542-7, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19564079

RESUMEN

Within the past 10-15 years our knowledge about cancer and how cancer cells might originate has changed dramatically. It is now generally believed that a tumor has its origin in cancer stem cells (CSCs), which originated either from transformed tissue stem cells or transformed progenitor cells that have regained self-renewal activity. CSCs share several characteristics of normal stem cells, such as self-renewal capacity, slow cell cycle activity, differentiation capacity, possessing an enhanced resistance towards cytotoxic agents and radiation, as well as tissue restoration capacity. Due to the increased drug and radiation resistance and slow cell cycle activity concomitant with tumor initiation capacity it is generally assumed that recurrent cancers originate from first line therapy surviving CSCs. But how does the CSC hypothesis explain "oncogenic resistance", which describes the phenomenon that most recurrent cancers are characterized by both an increased malignancy as well as resistance towards first line cancer therapy. To us, "oncogenic resistance" can not be simply attributed to the resistance properties of normal CSCs. If so, the recurring tumor should be treatable by first line therapy, which is mostly not the case. Thus, we conclude that "oncogenic resistance" demands a new type of tumor initiating cells, the so-called recurrence CSCs (rCSCs). This type of tumor initiating cell originates during first line therapy and is characterized by giving rise to first line therapy resistant and highly malignant progenies. Because several characteristics of "oncogenic resistance", such as increased drug resistance, increased resistance to apoptosis and an enhanced malignancy have been linked to cell fusion we further conclude that rCSCs might originate from this cellular event. However, which cell types have to fuse with each other to ultimately give rise to rCSCs is not clear. In any case, tumor tissues, particularly those being destructed by first line therapy comprise of a variety of fusogenic cells including tumor cells and CSCs as well as recruited monocytes/macrophages and bone marrow-derived stem cells. The fusogenic properties of these cells concomitant with phenotypic heterogeneity, which is also a property of cell fusion, will then lead to the origin of rCSCs. In accordance with Darwinian evolution only those cells will survive that can resist best to the selection pressure first line therapy.


Asunto(s)
Fusión Celular , Transformación Celular Neoplásica/patología , Modelos Biológicos , Neoplasias/patología , Neoplasias/fisiopatología , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Animales , Humanos
13.
Cancer Cell ; 15(2): 91-102, 2009 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-19185844

RESUMEN

Although gastrointestinal cancers are frequently associated with chronic inflammation, the underlying molecular links have not been comprehensively deciphered. Using loss- and gain-of-function mice in a colitis-associated cancer model, we establish here a link comprising the gp130/Stat3 transcription factor signaling axis. Mutagen-induced tumor growth and multiplicity are reduced following intestinal epithelial cell (IEC)-specific Stat3 ablation, while its hyperactivation promotes tumor incidence and growth. Conversely, IEC-specific Stat3 deficiency enhances susceptibility to chemically induced epithelial damage and subsequent mucosal inflammation, while excessive Stat3 activation confers resistance to colitis. Stat3 has the capacity to mediate IL-6- and IL-11-dependent IEC survival and to promote proliferation through G1 and G2/M cell-cycle progression as the common tumor cell-autonomous mechanism that bridges chronic inflammation to tumor promotion.


Asunto(s)
Ciclo Celular/fisiología , Supervivencia Celular/fisiología , Colitis , Receptor gp130 de Citocinas/metabolismo , Enterocitos/fisiología , Neoplasias , Factor de Transcripción STAT3/metabolismo , Animales , Apoptosis/fisiología , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Colitis/complicaciones , Colitis/inmunología , Colitis/patología , Receptor gp130 de Citocinas/genética , Enterocitos/citología , Enterocitos/patología , Humanos , Inflamación/inmunología , Interleucina-11/inmunología , Interleucina-6/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Ratones , Neoplasias/etiología , Neoplasias/inmunología , Neoplasias/patología , Regeneración/fisiología , Factor de Transcripción STAT3/genética , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...