Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Curr Issues Mol Biol ; 46(7): 6440-6471, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39057027

ABSTRACT

Despite recent campaigns for screening and the latest advances in cancer therapy and molecular biology, gastrointestinal (GI) neoplasms remain among the most frequent and lethal human tumors. Most GI neoplasms are sporadic, but there are some well-known familial syndromes associated with a significant risk of developing both benign and malignant GI tumors. Although some of these entities were described more than a century ago based on clinical grounds, the increasing molecular information obtained with high-throughput techniques has shed light on the pathogenesis of several of them. The vast amount of information gained from next-generation sequencing has led to the identification of some high-risk genetic variants, although others remain to be discovered. The opportunity for genetic assessment and counseling in these families has dramatically changed the management of these syndromes, though it has also resulted in significant psychological distress for the affected patients, especially those with indeterminate variants. Herein, we aim to summarize the most relevant hereditary cancer syndromes involving the stomach and colon, with an emphasis on new molecular findings, novel entities, and recent changes in the management of these patients.

2.
Br J Cancer ; 130(8): 1402-1413, 2024 May.
Article in English | MEDLINE | ID: mdl-38467828

ABSTRACT

BACKGROUND: Primary resistance to anti-EGFR therapies affects 40% of metastatic colorectal cancer patients harbouring wild-type RAS/RAF. YAP1 activation is associated with this resistance, prompting an investigation into AURKA's role in mediating YAP1 phosphorylation at Ser397, as observed in breast cancer. METHODS: We used transcriptomic analysis along with in vitro and in vivo models of RAS/RAF wild-type CRC to study YAP1 Ser397 phosphorylation as a potential biomarker for cetuximab resistance. We assessed cetuximab efficacy using CCK8 proliferation assays and cell cycle analysis. Additionally, we examined the effects of AURKA inhibition with alisertib and created a dominant-negative YAP1 Ser397 mutant to assess its impact on cancer stem cell features. RESULTS: The RAS/RAF wild-type CRC models exhibiting primary resistance to cetuximab prominently displayed elevated YAP1 phosphorylation at Ser397 primarily mediated by AURKA. AURKA-induced YAP1 phosphorylation was identified as a key trigger for cancer stem cell reprogramming. Consequently, we found that AURKA inhibition had the capacity to effectively restore cetuximab sensitivity and concurrently suppress the cancer stem cell phenotype. CONCLUSIONS: AURKA inhibition holds promise as a therapeutic approach to overcome cetuximab resistance in RAS/RAF wild-type colorectal cancer, offering a potential means to counter the development of cancer stem cell phenotypes associated with cetuximab resistance.


Subject(s)
Aurora Kinase A , Colorectal Neoplasms , Humans , Cetuximab/pharmacology , Cetuximab/metabolism , Aurora Kinase A/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Mutation , Proto-Oncogene Proteins p21(ras)/genetics
3.
Cell Mol Life Sci ; 80(6): 141, 2023 May 07.
Article in English | MEDLINE | ID: mdl-37149819

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a progressive, chronic, and neurodegenerative disease, and the most common cause of dementia worldwide. Currently, the mechanisms underlying the disease are far from being elucidated. Thus, the study of proteins involved in its pathogenesis would allow getting further insights into the disease and identifying new markers for AD diagnosis. METHODS: We aimed here to analyze protein dysregulation in AD brain by quantitative proteomics to identify novel proteins associated with the disease. 10-plex TMT (tandem mass tags)-based quantitative proteomics experiments were performed using frozen tissue samples from the left prefrontal cortex of AD patients and healthy individuals and vascular dementia (VD) and frontotemporal dementia (FTD) patients as controls (CT). LC-MS/MS analyses were performed using a Q Exactive mass spectrometer. RESULTS: In total, 3281 proteins were identified and quantified using MaxQuant. Among them, after statistical analysis with Perseus (p value < 0.05), 16 and 155 proteins were defined as upregulated and downregulated, respectively, in AD compared to CT (Healthy, FTD and VD) with an expression ratio ≥ 1.5 (upregulated) or ≤ 0.67 (downregulated). After bioinformatics analysis, ten dysregulated proteins were selected as more prone to be associated with AD, and their dysregulation in the disease was verified by qPCR, WB, immunohistochemistry (IHC), immunofluorescence (IF), pull-down, and/or ELISA, using tissue and plasma samples of AD patients, patients with other dementias, and healthy individuals. CONCLUSIONS: We identified and validated novel AD-associated proteins in brain tissue that should be of further interest for the study of the disease. Remarkably, PMP2 and SCRN3 were found to bind to amyloid-ß (Aß) fibers in vitro, and PMP2 to associate with Aß plaques by IF, whereas HECTD1 and SLC12A5 were identified as new potential blood-based biomarkers of the disease.


Subject(s)
Alzheimer Disease , Frontotemporal Dementia , Neurodegenerative Diseases , Humans , Alzheimer Disease/metabolism , Frontotemporal Dementia/genetics , Proteomics , Chromatography, Liquid , Tandem Mass Spectrometry , Amyloid beta-Peptides/metabolism , Prefrontal Cortex/metabolism , Biomarkers , tau Proteins/metabolism
4.
Int J Mol Sci ; 25(5)2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38473896

ABSTRACT

Gastric cancer (GC) is a heterogeneous disease, often diagnosed at advanced stages, with a 5-year survival rate of approximately 20%. Despite notable technological advancements in cancer research over the past decades, their impact on GC management and outcomes has been limited. Numerous molecular alterations have been identified in GC, leading to various molecular classifications, such as those developed by The Cancer Genome Atlas (TCGA) and the Asian Cancer Research Group (ACRG). Other authors have proposed alternative perspectives, including immune, proteomic, or epigenetic-based classifications. However, molecular stratification has not yet transitioned into clinical practice for GC, and little attention has been paid to alternative molecular classifications. In this review, we explore diverse molecular classifications in GC from a practical point of view, emphasizing their relationships with clinicopathological factors, prognosis, and therapeutic approaches. We have focused on classifications beyond those of TCGA and the ACRG, which have been less extensively reviewed previously. Additionally, we discuss the challenges that must be overcome to ensure their impact on patient treatment and prognosis. This review aims to serve as a practical framework to understand the molecular landscape of GC, facilitate the development of consensus molecular categories, and guide the design of innovative molecular studies in the field.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Proteomics
5.
PLoS Biol ; 18(6): e3000732, 2020 06.
Article in English | MEDLINE | ID: mdl-32603375

ABSTRACT

Coordination of gene expression with nutrient availability supports proliferation and homeostasis and is shaped by protein acetylation. Yet how physiological/pathological signals link acetylation to specific gene expression programs and whether such responses are cell-type-specific is unclear. AMP-activated protein kinase (AMPK) is a key energy sensor, activated by glucose limitation to resolve nutrient supply-demand imbalances, critical for diabetes and cancer. Unexpectedly, we show here that, in gastrointestinal cancer cells, glucose activates AMPK to selectively induce EP300, but not CREB-binding protein (CBP). Consequently, EP300 is redirected away from nuclear receptors that promote differentiation towards ß-catenin, a driver of proliferation and colorectal tumorigenesis. Importantly, blocking glycogen synthesis permits reactive oxygen species (ROS) accumulation and AMPK activation in response to glucose in previously nonresponsive cells. Notably, glycogen content and activity of the ROS/AMPK/EP300/ß-catenin axis are opposite in healthy versus tumor sections. Glycogen content reduction from healthy to tumor tissue may explain AMPK switching from tumor suppressor to activator during tumor evolution.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Colorectal Neoplasms/metabolism , E1A-Associated p300 Protein/metabolism , Glucose/pharmacology , Animals , CREB-Binding Protein/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Enzyme Activation/drug effects , Glycogen/metabolism , Mice, Inbred C57BL , Protein Binding/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , beta Catenin/metabolism
6.
Cytopathology ; 34(6): 630-633, 2023 11.
Article in English | MEDLINE | ID: mdl-37485960

ABSTRACT

OBJECTIVE: A case of intestinal spirochaetosis diagnosed in anal cytology is presented. METHODS: An anal liquid-based cytology was performed and stained with Papanicolau and Warthin-Starry stains. RESULTS: An apical filamentous "fringe" was identified in columnar cells groups. CONCLUSION: Anal cytology is a potential tool for intestinal spirochaetosis diagnosis.


Subject(s)
Anus Neoplasms , Papillomavirus Infections , Humans , Papillomavirus Infections/pathology , Cytodiagnosis , Anus Neoplasms/diagnosis , Anal Canal/pathology
7.
Int J Mol Sci ; 24(10)2023 May 12.
Article in English | MEDLINE | ID: mdl-37240002

ABSTRACT

Colitis-associated colorectal carcinoma (CAC) occurs in inflammatory bowel disease (IBD) because of the "chronic inflammation-dysplasia-cancer" carcinogenesis pathway characterized by p53 alterations in the early stages. Recently, gastric metaplasia (GM) has been described as the initial event of the serrated colorectal cancer (CRC) process, resulting from chronic stress on the colon mucosa. The aim of the study is to characterize CAC analyzing p53 alterations and microsatellite instability (MSI) to explore their relationship with GM using a series of CRC and the adjacent intestinal mucosa. Immunohistochemistry was performed to assess p53 alterations, MSI and MUC5AC expression as a surrogate for GM. The p53 mut-pattern was found in more than half of the CAC, most frequently stable (MSS) and MUC5AC negative. Only six tumors were unstable (MSI-H), being with p53 wt-pattern (p = 0.010) and MUC5AC positive (p = 0.005). MUC5AC staining was more frequently observed in intestinal mucosa, inflamed or with chronic changes, than in CAC, especially in those with p53 wt-pattern and MSS. Based on our results, we conclude that, as in the serrated pathway of CRC, in IBD GM occurs in inflamed mucosa, persists in those with chronic changes and disappears with the acquisition of p53 mutations.


Subject(s)
Colorectal Neoplasms , Inflammatory Bowel Diseases , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Colorectal Neoplasms/complications , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Microsatellite Instability , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Microsatellite Repeats , Mucin 5AC/genetics , Mucin 5AC/metabolism
8.
Br J Cancer ; 126(11): 1604-1615, 2022 06.
Article in English | MEDLINE | ID: mdl-35347323

ABSTRACT

BACKGROUND: Liver metastasis is the primary cause of colorectal cancer (CRC)-associated death. Aryl-hydrocarbon receptor-interacting protein (AIP), a putative positive intermediary in aryl-hydrocarbon receptor-mediated signalling, is overexpressed in highly metastatic human KM12SM CRC cells and other highly metastatic CRC cells. METHODS: Meta-analysis and immunohistochemistry were used to assess the relevance of AIP. Cellular functions and signalling mechanisms mediated by AIP were assessed by gain-of-function experiments and in vitro and in vivo experiments. RESULTS: A significant association of high AIP expression with poor CRC patients' survival was observed. Gain-of-function and quantitative proteomics experiments demonstrated that AIP increased tumorigenic and metastatic properties of isogenic KM12C (poorly metastatic) and KM12SM (highly metastatic to the liver) CRC cells. AIP overexpression dysregulated epithelial-to-mesenchymal (EMT) markers and induced several transcription factors and Cadherin-17 activation. The former induced the signalling activation of AKT, SRC and JNK kinases to increase adhesion, migration and invasion of CRC cells. In vivo, AIP expressing KM12 cells induced tumour growth and liver metastasis. Furthermore, KM12C (poorly metastatic) cells ectopically expressing AIP became metastatic to the liver. CONCLUSIONS: Our data reveal new roles for AIP in regulating proteins associated with cancer and metastasis to induce tumorigenic and metastatic properties in colon cancer cells driving liver metastasis.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Liver Neoplasms , Rectal Neoplasms , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Hydrocarbons , Immunohistochemistry , Liver Neoplasms/secondary , Neoplasm Metastasis
9.
J Proteome Res ; 20(11): 5115-5130, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34628858

ABSTRACT

New biomarkers of Alzheimer's disease (AD) with a diagnostic value in preclinical and prodromal stages are urgently needed. AD-related serum autoantibodies are potential candidate biomarkers. Here, we aimed at identifying AD-related serum autoantibodies using protein microarrays and mass spectrometry-based methods. To this end, an untargeted complementary screening using high-density (42,100 antigens) and low-density (384 antigens) planar protein-epitope signature tag (PrEST) arrays and an immunoprecipitation protocol coupled to mass spectrometry analysis were used for serum autoantibody profiling. From the untargeted screening phase, 377 antigens corresponding to 338 proteins were selected for validation. Out of them, IVD, CYFIP1, and ADD2 seroreactivity was validated using 128 sera from AD patients and controls by PrEST-suspension bead arrays, and ELISA or luminescence Halotag-based bead immunoassay using full-length recombinant proteins. Importantly, IVD, CYFIP1, and ADD2 showed in combination a noticeable AD diagnostic ability. Moreover, IVD protein abundance in the prefrontal cortex was significantly two-fold higher in AD patients than in controls by western blot and immunohistochemistry, whereas CYFIP1 and ADD2 were significantly down-regulated in AD patients. The panel of AD-related autoantigens identified by a comprehensive multiomics approach may provide new insights of the disease and should help in the blood-based diagnosis of Alzheimer's disease. Mass spectrometry raw data are available in the ProteomeXchange database with the access number PXD028392.


Subject(s)
Alzheimer Disease , Autoantibodies , Autoantigens , Biomarkers , Humans , Protein Array Analysis/methods
10.
Neuroendocrinology ; 111(1-2): 146-157, 2021.
Article in English | MEDLINE | ID: mdl-31991407

ABSTRACT

BACKGROUND: Galectin-1 (Gal-1) plays major roles in cancer by modulating different processes leading to tumor development and progression. In the last years, it has been suggested as a promising target for anticancer therapy. Recently, aflibercept has shown high affinity for Gal-1. Here, we investigated how aflibercept could exert its antitumor activity via Gal-1-driven pathways in neuroendocrine carcinomas (NECs). METHODS AND RESULTS: NEC tumor xenografts were used to assess the effect of aflibercept on Gal-1 functions. Aflibercept induced a significant reduction of Gal-1 at epithelial, stromal, and extracellular localizations in lung NEC, whereas this was not observed in colon NECs, which displayed low expression of Gal-1. Additionally, aflibercept significantly reduced p-VEGFR2 protein, extracellular matrix remodeling, epithelial-mesenchymal transition, and activation of cancer-associated fibroblast hampering cell invasion in lung NEC but not in colon NEC. Gal-1 screening in human NECs confirmed that pulmonary and pancreatic tumors displayed higher levels of Gal-1 than colon NECs, becoming good candidates to benefit from aflibercept treatment. CONCLUSIONS: The lack of validated predictive markers of aflibercept is a weakness for guaranteeing the best treatment management with this drug. This work provides new mechanistic insight of aflibercept depending on Gal-1. Thus, in tumors overexpressing Gal-1, aflibercept has not only an antiangiogenic effect but also prevents Gal-1-mediated tumor-stroma cross talk. The stronger aflibercept effect in tumors with high levels of Gal-1 points out this protein as a molecular marker to predict the efficacy of this agent not only for NECs but also for other tumors with high levels of this protein.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Galectin 1/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Angiogenesis Inhibitors/pharmacology , Animals , Carcinoma, Neuroendocrine/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Humans , Mice , Mice, Nude , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
Carcinogenesis ; 41(2): 203-213, 2020 04 22.
Article in English | MEDLINE | ID: mdl-31095674

ABSTRACT

Pancreatic adenocarcinoma upregulated factor (PAUF), also known as ZG16B, was previously found in the secretome of metastatic colorectal cancer cells. Here, we demonstrated the presence of PAUF at the intracellular level and its multiple effects on cancer progression. An initial decline of PAUF expression was observed at early stages of colorectal cancer followed by an increase at the metastatic site. PAUF was located at different cellular compartments: membrane-associated vesicles, endosomes, microtubule-associated vesicles, cell growth cones and the cell nucleus. PAUF loss in two colorectal cancer cell lines caused severe alterations in the cell phenotype and cell cycle, including tetraploidy, extensive genomic alterations, micronuclei and increased apoptosis. An exhaustive analysis of the PAUF interactome using different proteomic approaches revealed the presence of multiple components of the cell cycle, mitotic checkpoint, Wnt pathway and intracellular transport. Among the interacting proteins we found ZW10, a moonlighting protein with a dual function in membrane trafficking and mitosis. In addition, PAUF silencing was associated to APC loss and increased ß-catenin nuclear expression. Altogether, our results suggest that PAUF depletion increases aneuploidy, promotes apoptosis and activates the Wnt/ß-catenin pathway in colorectal cancer cells facilitating cancer progression. In summary, PAUF behaves as a multifunctional protein, with different roles in cancer progression according to the extra- or intracellular expression, suggesting a therapeutic value for colorectal cancer.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Colorectal Neoplasms/pathology , Lectins/metabolism , Liver Neoplasms/genetics , Microtubule-Associated Proteins/metabolism , Mitosis , Wnt Signaling Pathway , Adenomatous Polyposis Coli Protein/metabolism , Aneuploidy , Cell Line, Tumor , Colon/pathology , Colorectal Neoplasms/genetics , Disease Progression , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Intercellular Signaling Peptides and Proteins , Lectins/genetics , Liver/pathology , Liver Neoplasms/secondary , Protein Interaction Mapping , Proteomics , Rectum/pathology , Up-Regulation
12.
Neuroendocrinology ; 110(1-2): 50-62, 2020.
Article in English | MEDLINE | ID: mdl-31030198

ABSTRACT

BACKGROUND: Neuroendocrine carcinoma (NEC) is a rare and very aggressive tumor. It has been greatly understudied, and very little is known about optimal treatment strategy for patients with this disease. The purpose of this study was to evaluate in vivo whether anti-vascular endothelial growth factor (VEGF) drugs could be a therapeutic alternative for these tumors with a poor prognosis. METHODS: We have developed 2 xenograft models using either human cell line derived from lung (H460) or from colon (COLO320) NEC to assess the effect of 2 antiangiogenic drugs, aflibercept and bevacizumab, on tumor growth and their pathological characteristics. Additionally, tumors were subjected to immunohistochemistry staining and proteins were measured with Western blot and ELISA. RESULTS: Both aflibercept and bevacizumab showed significant antitumor activity (p < 0.001). In the H460 model, aflibercept resulted in 94% tumor growth inhibition (TGI) and bevacizumab treatment resulted in 72.2% TGI. Similarly, in the COLO320 model, aflibercept and bevacizumab resulted in 89.3 and 84% TGI, respectively. Moreover, antitumor activity occurs early after treatment initiation. Using Tumor Control Index score, which address the kinetics of tumor growth in a way comparable to the methods used in human clinical studies, we confirmed that both drugs inhibit significantly tumor growth. When tumor stabilization was evaluated, aflibercept shows higher ability to stabilize NEC tumors than bevacizumab. CONCLUSION: Results derived from this study strongly support anti-VEGF therapies, especially aflibercept, as a novel therapeutic option in NECs. Further studies are necessary, but our observations encourage the evaluation of antiangiogenics in clinical trials combined with standard chemotherapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Bevacizumab/pharmacology , Carcinoma, Neuroendocrine/drug therapy , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factors/antagonists & inhibitors , Cell Line, Tumor , Humans , Receptors, Vascular Endothelial Growth Factor
13.
Gut ; 68(12): 2129-2141, 2019 12.
Article in English | MEDLINE | ID: mdl-31366457

ABSTRACT

OBJECTIVE: Chronic inflammation is a risk factor in colorectal cancer (CRC) and reactive oxygen species (ROS) released by the inflamed stroma elicit DNA damage in epithelial cells. We sought to identify new drivers of ulcerative colitis (UC) and inflammatory CRC. DESIGN: The study uses samples from patients with UC, mouse models of colitis and CRC and mice deficient for the epithelial-to-mesenchymal transition factor ZEB1 and the DNA repair glycosylase N-methyl-purine glycosylase (MPG). Samples were analysed by immunostaining, qRT-PCR, chromatin immunoprecipitation assays, microbiota next-generation sequencing and ROS determination. RESULTS: ZEB1 was induced in the colonic epithelium of UC and of mouse models of colitis. Compared with wild-type counterparts, Zeb1-deficient mice were partially protected from experimental colitis and, in a model of inflammatory CRC, they developed fewer tumours and exhibited lower levels of DNA damage (8-oxo-dG) and higher expression of MPG. Knockdown of ZEB1 in CRC cells inhibited 8-oxo-dG induction by oxidative stress (H2O2) and inflammatory cytokines (interleukin (IL)1ß). ZEB1 bound directly to the MPG promoter whose expression inhibited. This molecular mechanism was validated at the genetic level and the crossing of Zeb1-deficient and Mpg-deficient mice reverted the reduced inflammation and tumourigenesis in the former. ZEB1 expression in CRC cells induced ROS and IL1ß production by macrophages that, in turn, lowered MPG in CRC cells thus amplifying a positive loop between both cells to promote DNA damage and inhibit DNA repair. CONCLUSIONS: ZEB1 promotes colitis and inflammatory CRC through the inhibition of MPG in epithelial cells, thus offering new therapeutic strategies to modulate inflammation and inflammatory cancer.


Subject(s)
Colitis, Ulcerative/genetics , Colonic Neoplasms/genetics , DNA Glycosylases/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms, Experimental , Zinc Finger E-box-Binding Homeobox 1/genetics , Animals , Biopsy , Cells, Cultured , Colitis, Ulcerative/complications , Colitis, Ulcerative/metabolism , Colonic Neoplasms/etiology , Colonic Neoplasms/pathology , DNA Glycosylases/metabolism , DNA Repair , Epithelial Cells/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Neoplasm/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Zinc Fingers
15.
Rev Esp Enferm Dig ; 111(7): 514-518, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31140285

ABSTRACT

INTRODUCTION AND OBJECTIVES: CDX2 is a specific transcription factor with a significant role in the early differentiation and maintenance of intestinal epithelial cells during gastrointestinal development and also as a tumor suppressor. The aim of this study was to assess the potential role of CDX2 expression as a prognostic predictor. MATERIAL AND METHODS: ninety-two of 206 (44.6%) patients with gastric carcinoma that underwent a curative surgery and had immunohistochemical staining for CDX2 were enrolled into the study; 51.1% were female and the average age was 74.07 years. Overall, 56.5% of tumors were of the intestinal type, 33.7% were diffuse and 9.8% were mixed; 23.9% were T1/T2, 76.1% were T3/T4 and lymph node metastases (N+) were identified in 69.6% of cases; 13% (12) were clinical stage I, 40.2% (37) were stage II and 46.7% (43) were stage III. RESULTS: a total of 68.5% (63) expressed CDX2. Our study suggests that CDX2 expression (HR = 0.339, p = 0.024) represents an independent risk marker together with the Lauren type (HR = 3.471, p = 0.022). There was association between a milder clinical stage and CDX2 expression (stage I) (p = 0.046). A significant difference was found in overall survival that favored patients with positive CDX2 expression (85.7% vs 65.5%, p = 0.012). CONCLUSION: our results confirm that CDX2 expression in gastric carcinoma is associated with improved prognosis, although further studies are needed to draw definitive conclusions.


Subject(s)
Adenocarcinoma/surgery , CDX2 Transcription Factor/physiology , Stomach Neoplasms/surgery , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Aged , CDX2 Transcription Factor/biosynthesis , Female , Humans , Male , Prognosis , Retrospective Studies , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Survival Rate
16.
J Proteome Res ; 17(1): 252-264, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29131639

ABSTRACT

We investigated new transcription and splicing factors associated with the metastatic phenotype in colorectal cancer. A concatenated tandem array of consensus transcription factor (TF)-response elements was used to pull down nuclear extracts in two different pairs of colorectal cancer cells, KM12SM/KM12C and SW620/480, genetically related but differing in metastatic ability. Proteins were analyzed by label-free LC-MS and quantified with MaxLFQ. We found 240 proteins showing a significant dysregulation in highly metastatic KM12SM cells relative to nonmetastatic KM12C cells and 257 proteins in metastatic SW620 versus SW480. In both cell lines there were similar alterations in genuine TFs and components of the splicing machinery like UPF1, TCF7L2/TCF-4, YBX1, or SRSF3. However, a significant number of alterations were cell-line specific. Functional silencing of MAFG, TFE3, TCF7L2/TCF-4, and SRSF3 in KM12 cells caused alterations in adhesion, survival, proliferation, migration, and liver homing, supporting their role in metastasis. Finally, we investigated the prognostic value of the altered TFs and splicing factors in cancer patients. SRSF3 and SFPQ showed significant prognostic value. We observed that SRSF3 displayed a gradual loss of expression associated with cancer progression. Loss of SRSF3 expression was significantly associated with poor survival and shorter disease-free survival, particularly in early stages, in colorectal cancer.


Subject(s)
Colorectal Neoplasms/chemistry , Neoplasm Metastasis , Proteomics/methods , RNA Splicing Factors/analysis , Transcription Factors/analysis , Cell Line, Tumor , Colorectal Neoplasms/pathology , Humans , Prognosis , Serine-Arginine Splicing Factors/analysis
17.
Mol Med ; 24(1): 64, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30567504

ABSTRACT

BACKGROUND: The multiple biological effects of vitamin D and its novel activities on inflammation and redox homeostasis have raised high expectations on its use as a therapeutic agent for multiple fibrogenic conditions. We have assessed the therapeutic effects of 1α,25-Dihydroxyvitamin D3, the biologically active form of vitamin D, in the context of lung fibrosis. METHODS: We have used representative cellular models for alveolar type II cells and human myofibroblasts. The extension of DNA damage and cellular senescence have been assessed by immunofluorescence, western-blot and senescence-associated ß-galactosidase activity. We have also set up a murine model for lung fibrosis by intraperitoneal injections of bleomycin. RESULTS: Vitamin D induces cellular senescence in bleomycin-treated alveolar epithelial type II cells and aggravates the lung pathology induced by bleomycin. These effects are probably due to an alteration of the cellular DNA double-strand breaks repair in bleomycin-treated cells. CONCLUSIONS: The detrimental effects of vitamin D in the presence of a DNA damaging agent might preclude its use as an antifibrogenic agent for pulmonary fibrosis characterized by DNA damage occurrence and cellular senescence.


Subject(s)
Cellular Senescence/drug effects , DNA Damage , Epithelial Cells/drug effects , Pulmonary Fibrosis/pathology , Vitamin D/analogs & derivatives , A549 Cells , Animals , Bleomycin , Epithelial Cells/metabolism , Female , Humans , Lung/drug effects , Lung/pathology , Mice, Inbred C57BL , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Pulmonary Fibrosis/chemically induced , Vitamin D/toxicity
18.
J Pathol ; 242(1): 39-51, 2017 05.
Article in English | MEDLINE | ID: mdl-28054337

ABSTRACT

The process of liver colonization in colorectal cancer remains poorly characterized. Here, we addressed the role of microRNA (miRNA) dysregulation in metastasis. We first compared miRNA expression profiles between colorectal cancer cell lines with different metastatic properties and then identified target proteins of the dysregulated miRNAs to establish their functions and prognostic value. We found that 38 miRNAs were differentially expressed between highly metastatic (KM12SM/SW620) and poorly metastatic (KM12C/SW480) cancer cell lines. After initial validation, we determined that three miRNAs (miR-424-3p, -503, and -1292) were overexpressed in metastatic colorectal cancer cell lines and human samples. Stable transduction of non-metastatic cells with each of the three miRNAs promoted metastatic properties in culture and increased liver colonization in vivo. Moreover, miR-424-3p and miR-1292 were associated with poor prognosis in human patients. A quantitative proteomic analysis of colorectal cancer cells transfected with miR-424-3p, miR-503, or miR-1292 identified alterations in 149, 129, or 121 proteins, respectively, with an extensive overlap of the target proteins of the three miRNAs. Importantly, down-regulation of two of these shared target proteins, CKB and UBA2, increased cell adhesion and proliferation in colorectal cancer cells. The capacity of distinct miRNAs to regulate the same mRNAs boosts the capacity of miRNAs to regulate cancer metastasis and underscores the necessity of targeting multiple miRNAs for effective cancer therapy. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Colorectal Neoplasms/genetics , MicroRNAs/genetics , Neoplasm Metastasis/genetics , RNA, Neoplasm/genetics , Animals , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Creatine Kinase, BB Form/biosynthesis , Creatine Kinase, BB Form/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Transplantation , Prognosis , Proteomics/methods , Ubiquitin-Activating Enzymes/biosynthesis , Ubiquitin-Activating Enzymes/genetics
19.
Rev Esp Enferm Dig ; 110(10): 674-675, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30168336

ABSTRACT

Anal adenocarcinomas account for approximately 10% of all anorectal tumours. We present the case of an adenocarcinoma of the proximal anal canal with exceptional immunohistochemistry as an incidental finding after haemorrhoidectomy, in a patient with endoscopically controlled serrated polyposis syndrome. Histopathological analysis of the specimen was a challenge in terms of differential diagnosis, which often determines the choice of therapeutic approach.


Subject(s)
Adenocarcinoma/pathology , Anus Neoplasms/pathology , Intestinal Polyposis/pathology , Adenocarcinoma/complications , Anus Neoplasms/complications , Female , Humans , Immunohistochemistry , Intestinal Polyposis/complications , Middle Aged , Syndrome
20.
Gut ; 66(4): 666-682, 2017 04.
Article in English | MEDLINE | ID: mdl-27965283

ABSTRACT

OBJECTIVE: Understand the role of ZEB1 in the tumour initiation and progression beyond inducing an epithelial-to-mesenchymal transition. DESIGN: Expression of the transcription factor ZEB1 associates with a worse prognosis in most cancers, including colorectal carcinomas (CRCs). The study uses survival analysis, in vivo mouse transgenic and xenograft models, gene expression arrays, immunostaining and gene and protein regulation assays. RESULTS: The poorer survival determined by ZEB1 in CRCs depended on simultaneous high levels of the Wnt antagonist DKK1, whose expression was transcriptionally activated by ZEB1. In cancer cells with mutant TP53, ZEB1 blocked the formation of senescence-associated heterochromatin foci at the onset of senescence by triggering a new regulatory cascade that involves the subsequent activation of DKK1, mutant p53, Mdm2 and CtBP to ultimately repress macroH2A1 (H2AFY). In a transgenic mouse model of colon cancer, partial downregulation of Zeb1 was sufficient to induce H2afy and to trigger in vivo tumour senescence, thus resulting in reduced tumour load and improved survival. The capacity of ZEB1 to induce tumourigenesis in a xenograft mouse model requires the repression of H2AFY by ZEB1. Lastly, the worst survival effect of ZEB1 in patients with CRC ultimately depends on low expression of H2AFY and of senescence-associated genes. CONCLUSIONS: The tumourigenic capacity of ZEB1 depends on its inhibition of cancer cell senescence through the activation of a herein identified new molecular pathway. These results set ZEB1 as a potential target in therapeutic strategies aimed at inducing senescence.


Subject(s)
Carcinogenesis/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Histones/genetics , Intercellular Signaling Peptides and Proteins/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Cell Line, Tumor , Cell Survival/genetics , Cellular Senescence/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Heterografts , Histones/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Transgenic , Mutation , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Survival Rate , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Wnt Signaling Pathway , Zinc Finger E-box-Binding Homeobox 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL