Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 15(22)2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-38001576

RESUMEN

We have previously shown that the extracellular matrix and basement membrane protein Nidogen1 (NID1) is secreted by more malignant, mesenchymal-like CRC cells and induces the epithelial-mesenchymal transition (EMT) and promotes the migration and invasion of less malignant, epithelial-like CRC cells. Here, we performed a comprehensive bioinformatics analysis of multiple datasets derived from CRC patients and showed that elevated expression of NID1 and the genes ITGA3, ITGB1, and ITGAV, which encode NID1 receptors, is associated with poor prognosis and advanced tumor stage. Accordingly, the expression of NID1, ITGA3, ITGB1, and ITGAV was associated with an EMT signature, which included SNAIL/SNAI1, an EMT-inducing transcription factor. In CRC cells, ectopic SNAIL expression induced NID1 and SNAIL occupancy was detected at an E-box upstream of the NID1 transcription start site. Therefore, NID1 represents a direct target of SNAIL. Ectopic expression of NID1 or treatment with NID1-containing medium endowed non-metastatic CRC cells with the capacity to form lung metastases after xenotransplantation into mice. Suppression of the NID1 receptor ITGAV decreased cell viability, particularly in CMS/consensus molecular subtype 4 CRC cells. Taken together, our results show that NID1 is a direct target of EMT-TF SNAIL and is associated with and promotes CRC progression and metastasis. Furthermore, the NID1 receptor ITGAV represents a candidate therapeutic target in CMS4 colorectal tumors.

2.
Int J Biol Sci ; 18(14): 5415-5437, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36147476

RESUMEN

The CSF1 receptor (CSF1R) encoding mRNA represents a direct target of miR-34a. However, the in vivo relevance of the suppression of CSF1R by miR-34a for intestinal tumor suppression mediated by the p53/miR-34a pathway has remained unknown. Here, Apc Min/+ mice with intestinal-epithelial cell (IEC)-specific deletions of Mir34a showed increased formation of adenomas and decreased survival, whereas deletion of Csf1r decreased adenoma formation and increased survival. In adenomas deletion of Mir34a enhanced proliferation, STAT3 signaling, infiltration with fibroblasts, immune cells and microbes, and tumor stem cell abundance and decreased apoptosis. Deletion of Csf1r had the opposite effects. In addition, homeostasis of intestinal secretory and stem cells, and tumoroid formation were affected in opposite directions by deletion of Mir34a and CSF1R. Concomitant deletion of Csf1r and Mir34a neutralized the effects of the single deletions. mRNAs containing Mir34a seed-matching sites, which encode proteins related to EMT (epithelial-mesenchymal transition), stemness and Wnt signaling, were enriched after Mir34a inactivation in adenomas and derived tumoroids. Netrin-1/Ntn1 and Transgelin/Tagln were characterized as direct targets of Mir34a and Csf1r signaling. Mir34a-inactivation related expression signatures were associated with CMS4/CRISB+D, stage 4 CRCs and poor patient survival. In tumoroids the loss of Mir34a conferred resistance to 5-FU which was mediated by Csf1r. This study provides genetic evidence for a requirement of Mir34a-mediated Csf1r suppression for intestinal stem/secretory cell homeostasis and tumor suppression, and suggests that therapeutic targeting of CSF1R may be effective for the treatment of CRCs with defects in the p53/miR-34a pathway.


Asunto(s)
Adenoma , MicroARNs , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Adenoma/genética , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Fluorouracilo , Regulación Neoplásica de la Expresión Génica/genética , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Netrina-1/genética , Netrina-1/metabolismo , ARN Mensajero , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
3.
Mol Cancer ; 21(1): 120, 2022 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-35624466

RESUMEN

BACKGROUND: AP4 (TFAP4) encodes a basic helix-loop-helix leucine zipper (bHLH-LZ) transcription factor and is a direct target gene of the oncogenic transcription factor c-MYC. Here, we set out to determine the relevance of AP4 in human colorectal cancer (CRC) cells. METHODS: A CRISPR/Cas9 approach was employed to generate AP4-deficient CRC cell lines with inducible expression of c-MYC. Colony formation, ß-gal staining, immunofluorescence, comet and homologous recombination (HR) assays and RNA-Seq analysis were used to determine the effects of AP4 inactivation. qPCR and qChIP analyses was performed to validate differentially expressed AP4 targets. Expression data from CRC cohorts was subjected to bioinformatics analyses. Immunohistochemistry was used to evaluate AP4 targets in vivo. Ap4-deficient APCmin/+ mice were analyzed to determine conservation. Immunofluorescence, chromosome and micronuclei enumeration, MTT and colony formation assays were used to determine the effects of AP4 inactivation and target gene regulation on chromosomal instability (CIN) and drug sensitivity. RESULTS: Inactivation of AP4 in CRC cell lines resulted in increased spontaneous and c-MYC-induced DNA damage, chromosomal instability (CIN) and cellular senescence. AP4-deficient cells displayed increased expression of the long non-coding RNA MIR22HG, which encodes miR-22-3p and was directly repressed by AP4. Furthermore, Mediator of DNA damage Checkpoint 1 (MDC1), a central component of the DNA damage response and a known target of miR-22-3p, displayed decreased expression in AP4-deficient cells. Accordingly, MDC1 was directly induced by AP4 and indirectly by AP4-mediated repression of miR-22-3p. Adenomas and organoids from Ap4-deficient APCmin/+ mice displayed conservation of these regulations. Inhibition of miR-22-3p or ectopic MDC1 expression reversed the increased senescence, DNA damage, CIN and defective HR observed in AP4-deficient CRC cells. AP4-deficiency also sensitized CRC cells to 5-FU treatment, whereas ectopic AP4 conferred resistance to 5-FU in a miR-22-3p and MDC1-dependent manner. CONCLUSIONS: In summary, AP4, miR-22-3p and MDC1 form a conserved and coherent, regulatory feed-forward loop to promote DNA repair, which suppresses DNA damage, senescence and CIN, and contributes to 5-FU resistance. These findings explain how elevated AP4 expression contributes to development and chemo-resistance of colorectal cancer after c-MYC activation.


Asunto(s)
Neoplasias Colorrectales , MicroARNs , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular/genética , Inestabilidad Cromosómica , Neoplasias Colorrectales/genética , Daño del ADN , Fluorouracilo/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Factores de Transcripción/genética
4.
Nat Commun ; 9(1): 3573, 2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-30177706

RESUMEN

The gene encoding the transcription factor TFAP4/AP4 represents a direct target of the c-MYC oncoprotein. Here, we deleted Ap4 in ApcMin mice, a preclinical model of inherited colorectal cancer. Ap4 deficiency extends their average survival by 110 days and decreases the formation of intestinal adenomas and tumor-derived organoids. The effects of Ap4 deletion are presumably due to the reduced number of functional intestinal stem cells (ISCs) amenable to adenoma-initiating mutational events. Deletion of Ap4 also decreases the number of colonic stem cells and increases the number of Paneth cells. Expression profiling revealed that ISC signatures, as well as the Wnt/ß-catenin and Notch signaling pathways are downregulated in Ap4-deficient adenomas and intestinal organoids. AP4-associated signatures are conserved between murine adenomas and human colorectal cancer samples. Our results establish Ap4 as rate-limiting mediator of adenoma initiation, as well as regulator of intestinal and colonic stem cell and Paneth cell homeostasis.


Asunto(s)
Adenoma/genética , Colon/metabolismo , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Organoides/metabolismo , Células Madre/metabolismo , Factores de Transcripción/genética , Adenoma/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Colon/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Simulación por Computador , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Homeostasis , Humanos , Mucosa Intestinal/metabolismo , Neoplasias Intestinales/metabolismo , Neoplasias Intestinales/patología , Intestinos/patología , Ratones , Ratones Noqueados , Organoides/patología , Células de Paneth/patología , Receptores Notch/metabolismo , Células Madre/citología , Vía de Señalización Wnt
5.
Pancreas ; 40(1): 144-50, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20938367

RESUMEN

OBJECTIVES: RNA interference, an indispensable tool in functional genomics, can be induced by small interfering RNAs (siRNAs). Because of the transient nature of siRNA-mediated RNA interference, the continuous use of transfection reagents is mandatory. Because transfection reagents are expensive, cost-effective alternatives must be considered. In this study, we describe a polyethylenimine-based siRNA transfection protocol for pancreatic cancer cell lines. METHODS: For determination of polyethylenimine-based transfection efficiency, a FAM-labeled siRNA was transfected into several pancreatic cancer cell lines and subsequently analyzed by flow cytometry. The effective knockdown of 2 siRNAs was determined on the protein level by Western blot. Toxicity of the transfection reagent was analyzed by viability assays. RESULTS: Polyethylenimine can be used without overt cellular morphological changes, and toxicity is negligible in human and murine pancreatic cancer cell lines. Transfection efficiencies ranged between 83% and 98% in the cell lines used. The knockdown at the protein level was comparable to commercially available transfection reagents. Polyethylenimine and siRNA concentrations, incubation time, and cell density are determinates of the transfection efficiency. CONCLUSIONS: Polyethylenimine is a suitable and cost-effective alternative for transfecting siRNAs into pancreatic cancer cells.


Asunto(s)
Neoplasias Pancreáticas/genética , Polietileneimina/farmacología , ARN Interferente Pequeño/genética , Transfección/métodos , Animales , Recuento de Células , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Análisis Costo-Beneficio , Humanos , Ratones , FN-kappa B/metabolismo , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...