Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Pathol ; 262(3): 347-361, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38235615

RESUMEN

Partial epithelial-mesenchymal transition (p-EMT) has recently been identified as a hybrid state consisting of cells with both epithelial and mesenchymal characteristics and is associated with the migration, metastasis, and chemoresistance of cancer cells. Here, we describe the induction of p-EMT in starved colorectal cancer (CRC) cells and identify a p-EMT gene signature that can predict prognosis. Functional characterisation of starvation-induced p-EMT in HCT116, DLD1, and HT29 cells showed changes in proliferation, morphology, and drug sensitivity, supported by in vivo studies using the chorioallantoic membrane model. An EMT-specific quantitative polymerase chain reaction (qPCR) array was used to screen for deregulated genes, leading to the establishment of an in silico gene signature that was correlated with poor disease-free survival in CRC patients along with the CRC consensus molecular subtype CMS4. Among the significantly deregulated p-EMT genes, a triple-gene signature consisting of SERPINE1, SOX10, and epidermal growth factor receptor (EGFR) was identified. Starvation-induced p-EMT was characterised by increased migratory potential and chemoresistance, as well as E-cadherin processing and internalisation. Both gene signature and E-cadherin alterations could be reversed by the proteasomal inhibitor MG132. Spatially resolving EGFR expression with high-resolution immunofluorescence imaging identified a proliferation stop in starved CRC cells caused by EGFR internalisation. In conclusion, we have gained insight into a previously undiscovered EMT mechanism that may become relevant when tumour cells are under nutrient stress, as seen in early stages of metastasis. Targeting this process of tumour cell dissemination might help to prevent EMT and overcome drug resistance. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias Colorrectales , Humanos , Neoplasias Colorrectales/patología , Proliferación Celular , Transición Epitelial-Mesenquimal/genética , Receptores ErbB , Línea Celular Tumoral , Cadherinas/genética , Cadherinas/metabolismo , Movimiento Celular
2.
Cell Mol Life Sci ; 79(8): 423, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35838828

RESUMEN

In cancer, the activating transcription factor 2 (ATF2) has pleiotropic functions in cellular responses to growth stimuli, damage, or inflammation. Due to only limited studies, the significance of ATF2 in colorectal cancer (CRC) is not well understood. We report that low ATF2 levels correlated with worse prognosis and tumor aggressiveness in CRC patients. NanoString gene expression and ChIP analysis confirmed trophoblast cell surface antigen 2 (TROP2) as a novel inhibitory ATF2 target gene. This inverse correlation was further observed in primary human tumor tissues. Immunostainings revealed that high intratumoral heterogeneity for ATF2 and TROP2 expression was sustained also in liver metastasis. Mechanistically, our in vitro data of CRISPR/Cas9-generated ATF2 knockout (KO) clones revealed that high TROP2 levels were critical for cell de-adhesion and increased cell migration without triggering EMT. TROP2 was enriched in filopodia and displaced Paxillin from adherens junctions. In vivo imaging, micro-computer tomography, and immunostainings verified that an ATF2KO/TROP2high status triggered tumor invasiveness in in vivo mouse and chicken xenograft models. In silico analysis provided direct support that ATF2low/TROP2high expression status defined high-risk CRC patients. Finally, our data demonstrate that ATF2 acts as a tumor suppressor by inhibiting the cancer driver TROP2. Therapeutic TROP2 targeting might prevent particularly the first steps in metastasis, i.e., the de-adhesion and invasion of colon cancer cells.


Asunto(s)
Factor de Transcripción Activador 2 , Antígenos de Neoplasias , Neoplasias Colorrectales , Factor de Transcripción Activador 2/genética , Factor de Transcripción Activador 2/metabolismo , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral/metabolismo , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Humanos , Ratones , Regulación hacia Arriba
3.
Carcinogenesis ; 41(9): 1219-1228, 2020 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-32628753

RESUMEN

AKR1B1 and AKR1B10, members of the aldo-keto reductase family of enzymes that participate in the polyol pathway of aldehyde metabolism, are aberrantly expressed in colon cancer. We previously showed that high expression of AKR1B1 (AKR1B1HIGH) was associated with enhanced motility, inflammation and poor clinical outcome in colon cancer patients. Using publicly available datasets and ex vivo gene expression analysis (n = 51, Ankara cohort), we have validated our previous in silico finding that AKR1B1HIGH was associated with worse overall survival (OS) compared with patients with low expression of AKR1B1 (AKR1B1LOW) samples. A combined signature of AKR1B1HIGH and AKR1B10LOW was significantly associated with worse recurrence-free survival (RFS) in microsatellite stable (MSS) patients and in patients with distal colon tumors as well as a higher mesenchymal signature when compared with AKR1B1LOW/AKR1B10HIGH tumors. When the patients were stratified according to consensus molecular subtypes (CMS), AKR1B1HIGH/AKR1B10LOW samples were primarily classified as CMS4 with predominantly mesenchymal characteristics while AKR1B1LOW/AKR1B10HIGH samples were primarily classified as CMS3 which is associated with metabolic deregulation. Reverse Phase Protein Array carried out using protein samples from the Ankara cohort indicated that AKR1B1HIGH/AKR1B10LOW tumors showed aberrant activation of metabolic pathways. Western blot analysis of AKR1B1HIGH/AKR1B10LOW colon cancer cell lines also suggested aberrant activation of nutrient-sensing pathways. Collectively, our data suggest that the AKR1B1HIGH/AKR1B10LOW signature may be predictive of poor prognosis, aberrant activation of metabolic pathways, and can be considered as a novel biomarker for colon cancer prognostication.


Asunto(s)
Aldehído Reductasa/metabolismo , Aldo-Ceto Reductasas/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias del Colon/patología , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Aldehído Reductasa/genética , Aldo-Ceto Reductasas/genética , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Estudios de Cohortes , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Humanos , Pronóstico , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Tasa de Supervivencia , Células Tumorales Cultivadas
4.
Biophys Chem ; 288: 106855, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35849958

RESUMEN

Lipoxygenases (LOX) are a family lipid oxygenating enzymes that can generate bioactive lipids of clinical relevance from polyunsaturated fatty acids. Most LOXs display a Ca2+-dependent association with membranes for their activity. Nanodiscs (ND) are stable self-assembled discoidal fragments of lipid bilayers that can mimic the plasma membrane. In this study, we evaluated the association of mammalian 15-LOXs (ALOX15 and ALOX15B) and soybean LOX-1 with NDs (LOX-ND), their enzymatic activities and inhibition. Mammalian LOXs associated with NDs showed better retention of enzymatic function compared to soybean LOX-1. Treatment of both LOX-NDs and free enzymes with the pan-LOX inhibitor nordihydroguaiaretic acid (NDGA) showed an approximately 5-fold more effective inhibition of the enzymes associated with NDs compared to the free form. NDs are easy to generate membrane mimics that can be used as an effective tool to determine enzymatic function and inhibition of membrane associated proteins.


Asunto(s)
Inhibidores de la Lipooxigenasa , Lipooxigenasas , Animales , Inhibidores de la Lipooxigenasa/química , Inhibidores de la Lipooxigenasa/farmacología , Lipooxigenasas/química , Lipooxigenasas/metabolismo , Mamíferos/metabolismo , Receptores Depuradores de Clase E
5.
Sci Rep ; 11(1): 13637, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34211032

RESUMEN

Recently, there has been a resurgence of interest in metabolic rewiring of tumors to identify clinically relevant genes. However, most of these studies have had either focused on individual tumors, or are too general, providing a broad outlook on overall changes. In this study, we have first curated an extensive list of genes encoding metabolic enzymes and metabolite transporters relevant to carbohydrate, fatty acid and amino acid oxidation and biosynthesis. Next, we have used publicly available transcriptomic data for 20 different tumor types from The Cancer Genome Atlas Network (TCGA) and focused on differential expression of these genes between tumor and adjacent normal tissue. Our study revealed major transcriptional alterations in genes that are involved in central metabolism. Most tumors exhibit upregulation in carbohydrate and amino acid transporters, increased glycolysis and pentose phosphate pathway, and decreased fatty acid and amino acid oxidation. On the other hand, the expression of genes of the tricarboxylic acid cycle, anaplerotic reactions and electron transport chain differed between tumors. Although most transcriptomic alterations were conserved across many tumor types suggesting the initiation of common regulatory programs, expression changes unique to specific tumors were also identified, which can provide gene expression fingerprints as potential biomarkers or drug targets. Our study also emphasizes the value of transcriptomic data in the deeper understanding of metabolic changes in diseases.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Transcriptoma , Aminoácidos/metabolismo , Metabolismo de los Hidratos de Carbono , Ciclo del Ácido Cítrico , Ácidos Grasos/metabolismo , Humanos , Redes y Vías Metabólicas , Neoplasias/metabolismo
6.
FEBS J ; 287(12): 2504-2523, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31762160

RESUMEN

Intestinal epithelial cells are derived from stem cells at the crypts that undergo differentiation into transit-amplifying cells, which in turn form terminally differentiated enterocytes as these cells reach the villus. Extensive alterations in both transcriptional and translational programs occur during differentiation, which can induce the activation of cellular stress responses such as ER stress-related unfolded protein response (UPR) and autophagy, particularly in the cells that are already committed to becoming absorptive cells. Using an epithelial cell model of enterocyte differentiation, we report a mechanistic study connecting enterocyte differentiation to UPR and autophagy. We report that differentiated colon epithelial cells showed increased cytosolic Ca2+ levels and activation of all three pathways of UPR: inositol-requiring enzyme 1 (IRE1), protein kinase RNA-like ER kinase, and activating transcription factor 6 (ATF6) compared to the undifferentiated cells. Enhanced UPR in the differentiated cells was accompanied by the induction of autophagy as evidenced by increased ratio of light chain 3 II/I, upregulation of Beclin-1, and downregulation of p62. We show for the first time that mechanistically, the upregulation of hepatocyte nuclear factor 4α (HNF4α) during differentiation led to increased promoter binding and transcriptional upregulation of two major proteins of UPR: X-box binding protein-1 and ATF6, implicating HNF4α as a key regulator of UPR response during differentiation. Integrating wet-lab with in silico analyses, the present study links differentiation to cellular stress responses, and highlights the importance of transcription factor signaling and cross-talk between the cellular events in the regulation of intestinal cell differentiation.


Asunto(s)
Estrés del Retículo Endoplásmico/genética , Células Epiteliales/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Mucosa Intestinal/metabolismo , Diferenciación Celular , Factor Nuclear 4 del Hepatocito/metabolismo , Humanos , Células Tumorales Cultivadas
7.
Cell Signal ; 64: 109410, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31487531

RESUMEN

Short chain fatty acids (SCFAs) are produced by commensal bacteria in the gut and are known to reduce inflammation through transcriptional inhibition of cytokines and inflammatory proteins such as cyclooxygenase-2 (COX-2). Butyrate is a SCFA that was reported to alter the mRNA stability of inflammatory genes by increasiing the expression of the RNA binding protein (RBP) Tristetraprolin (TTP). We have hypothesized that butyrate may regulate gene expression post-transcriptionally through global effects on the expression or cytoplasmic translocation of RBPs. Using bioinformatics analyses of publicly available microarray data as well as colon cancer cell lines treated with sodium butyrate, we have observed that butyrate treatment led to a general reduction in expression of several (but not all) RBPs and inhibition in the cytosolic translocation of HuR, a well-known stabilizing RBP. This was reflected in reduced NanoLuc reporter activity of several different AU-rich element (ARE) sequences in the presence of butyrate; this suppression was retained even when HuR was overexpressed. Mechanistically, we have shown that reduced activity of HuR was related to decreased phosphorylation of p38 and MK2 and enhanced phosphorylation of Chk2. As a proof of concept, we show butyrate-mediated inhibition in binding of HuR to the 3'UTR of COX-2 mRNA resulting in reduced mRNA and protein levels of the inflammatory gene. Overall, our data suggest that butyrate can reduce the expression of inflammatory genes not only by transcriptional regulation, but also by post-transcriptional regulation via inhibition of mRNA stabilizing proteins.


Asunto(s)
Butiratos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/genética , Proteínas de Unión al ARN/genética , Células CACO-2 , Ciclooxigenasa 2/metabolismo , Proteína 1 Similar a ELAV/genética , Células HCT116 , Células HT29 , Humanos , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/metabolismo , Transcripción Genética/efectos de los fármacos
8.
Sci Rep ; 8(1): 14828, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30287873

RESUMEN

Dimethyl sulfoxide (DMSO) is a small molecule with polar, aprotic and amphiphilic properties. It serves as a solvent for many polar and nonpolar molecules and continues to be one of the most used solvents (vehicle) in medical applications and scientific research. To better understand the cellular effects of DMSO within the concentration range commonly used as a vehicle (0.1-1.5%, v/v) for cellular treatments, we applied Attenuated Total Reflectance (ATR) Fourier Transform Infrared (FT-IR) spectroscopy to DMSO treated and untreated epithelial colon cancer cells. Both unsupervised (Principal Component Analysis-PCA) and supervised (Linear Discriminant Analysis-LDA) pattern recognition/modelling algorithms applied to the IR data revealed total segregation and prominent differences between DMSO treated and untreated cells at whole, lipid and nucleic acid regions. Several of these data were supported by other independent techniques. Further IR data analyses of macromolecular profile indicated comprehensive alterations especially in proteins and nucleic acids. Protein secondary structure analysis showed predominance of ß-sheet over α-helix in DMSO treated cells. We also observed for the first time, a reduction in nucleic acid level upon DMSO treatment accompanied by the formation of Z-DNA. Molecular docking and binding free energy studies indicated a stabilization of Z-DNA in the presence of DMSO. This alternate DNA form may be related with the specific actions of DMSO on gene expression, differentiation, and epigenetic alterations. Using analytical tools combined with molecular and cellular biology techniques, our data indicate that even at very low concentrations, DMSO induces a number of changes in all macromolecules, which may affect experimental outcomes where DMSO is used as a solvent.


Asunto(s)
Neoplasias del Colon/patología , Dimetilsulfóxido/metabolismo , Células Epiteliales/fisiología , Algoritmos , Neoplasias del Colon/metabolismo , Simulación por Computador , ADN de Forma Z/metabolismo , Células HCT116 , Humanos , Simulación del Acoplamiento Molecular , Complejos Multiproteicos/metabolismo , Análisis de Componente Principal , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Receptores de Reconocimiento de Patrones/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA