Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
EMBO Rep ; 21(4): e48938, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32052574

RESUMEN

Intestinal stem cells (ISCs) fuel the lifelong self-renewal of the intestinal tract and are paramount for epithelial repair. In this context, the Wnt pathway component LGR5 is the most consensual ISC marker to date. Still, the effort to better understand ISC identity and regulation remains a challenge. We have generated a Mex3a knockout mouse model and show that this RNA-binding protein is crucial for the maintenance of the Lgr5+ ISC pool, as its absence disrupts epithelial turnover during postnatal development and stereotypical organoid maturation ex vivo. Transcriptomic profiling of intestinal crypts reveals that Mex3a deletion induces the peroxisome proliferator-activated receptor (PPAR) pathway, along with a decrease in Wnt signalling and loss of the Lgr5+ stem cell signature. Furthermore, we identify PPARγ activity as a molecular intermediate of MEX3A-mediated regulation. We also show that high PPARγ signalling impairs Lgr5+ ISC function, thus uncovering a new layer of post-transcriptional regulation that critically contributes to intestinal homeostasis.


Asunto(s)
Mucosa Intestinal , Células Madre , Animales , Intestinos , Ratones , Organoides , Receptores Acoplados a Proteínas G/genética , Vía de Señalización Wnt
2.
Int J Mol Sci ; 23(3)2022 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-35162954

RESUMEN

Mesothelin (MSLN) overexpression (OE) is a frequent finding in ovarian carcinomas and increases cell survival and tumor aggressiveness. Since cancer stem cells (CSCs) contribute to pathogenesis, chemoresistance and malignant behavior in ovarian cancer (OC), we hypothesized that MSLN expression could be creating a favorable environment that nurtures CSCs. In this study, we analyzed the expression of MSLN and CSC markers SOX2 and ALDH1 by immunohistochemistry (IHC) in different model systems: primary high-grade serous carcinomas (HGSCs) and OC cell lines, including cell lines that were genetically engineered for MSLN expression by either CRISPR-Cas9-mediated knockout (Δ) or lentivirus-mediated OE. Cell lines, wild type and genetically engineered, were evaluated in 2D and 3D culture conditions and xenografted in nude mice. We observed that MSLN was widely expressed in HGSC, and restricted expression was observed in OC cell lines. In contrast, SOX2 and ALDH1 expression was limited in all tissue and cell models. Most importantly, the expression of CSC markers was independent of MSLN expression, and manipulation of MSLN expression did not affect CSC markers. In conclusion, MSLN expression is not involved in driving the CSC phenotype.


Asunto(s)
Familia de Aldehído Deshidrogenasa 1/metabolismo , Cistadenocarcinoma Seroso/patología , Mesotelina/metabolismo , Neoplasias Ováricas/patología , Retinal-Deshidrogenasa/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Cistadenocarcinoma Seroso/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Neoplasias Ováricas/metabolismo , Estudios Retrospectivos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Lab Invest ; 100(1): 120-134, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31641225

RESUMEN

Flourescence-based multiplex immunohistochemistry (mIHC) combined with multispectral imaging and digital image analysis (DIA) is a quantitative high-resolution method for determination of protein expression in tissue. We applied this method for five biomarkers (CDX2, SOX2, SOX9, E-cadherin, and ß-catenin) using tissue microarrays of a Norwegian unselected series of primary colorectal cancer. The data were compared with previously obtained chromogenic IHC data of the same tissue cores, visually assessed by the Allred method. We found comparable results between the methods, although confirmed that DIA offered improved resolution to differentiate cases with high and low protein expression. However, we experienced inherent challenges with digital image analysis of membrane staining, which was better assessed visually. DIA and mIHC enabled quantitative analysis of biomarker coexpression on the same tissue section at the single-cell level, revealing a strong negative correlation between the differentiation markers CDX2 and SOX2. Both methods confirmed known prognostic associations for CDX2, but DIA improved data visualization and detection of clinicopathological and biological associations. In summary, mIHC combined with DIA is an efficient and reliable method to evaluate protein expression in tissue, here shown to recapitulate and improve detection of known clinicopathological and survival associations for the emerging biomarker CDX2, and is therefore a candidate approach to standardize CDX2 detection in pathology laboratories.


Asunto(s)
Biomarcadores de Tumor/análisis , Técnica del Anticuerpo Fluorescente , Interpretación de Imagen Asistida por Computador , Biomarcadores de Tumor/metabolismo , Factor de Transcripción CDX2/metabolismo , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/metabolismo , Humanos , Factores de Transcripción SOXB1/metabolismo , Análisis de Matrices Tisulares
4.
Gastric Cancer ; 23(5): 811-823, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32215766

RESUMEN

BACKGROUND: Gastric cancer is the fifth most common cancer and the third cause of global cancer mortality. CDX2 is an intestinal differentiation marker with prognostic value in gastric cancer and transcriptionally regulates the expression of glycoprotein A33 (GPA33) and liver intestine cadherin (LI-cadherin). METHODS: This study evaluated the clinical significance of the combined expression of CDX2 and its targets GPA33 and LI-cadherin in gastric cancer by fluorescence-based multiplex immunohistochemistry together with digital image analysis and chromogenic immunohistochemistry in 329 gastric cancer samples arranged in tissue microarrays. Additionally, publicly available RNA-seq expression data from 354 gastric cancer samples from the TCGA database were used to validate the immunohistochemistry results. RESULTS: Expression of the three markers (CDX2, GPA33, and LI-cadherin) was strongly correlated, defining an intestinal differentiation panel. Low or negative protein expression of the intestinal differentiation panel identified patients with particularly poor overall survival, irrespective of the methodology used, and was validated in the independent series at the RNA-seq level. CONCLUSIONS: Expression of the intestinal differentiation panel (CDX2, GPA33, and LI-cadherin) defines a set of biomarkers with a strong biological rationale and favourable impact for prognostication of gastric cancer patients.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Biomarcadores de Tumor/metabolismo , Factor de Transcripción CDX2/metabolismo , Cadherinas/metabolismo , Intestinos/citología , Glicoproteínas de Membrana/metabolismo , Neoplasias Gástricas/patología , Anciano , Diferenciación Celular , Femenino , Estudios de Seguimiento , Humanos , Masculino , Pronóstico , Estudios Retrospectivos , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/cirugía , Tasa de Supervivencia
5.
Nucleic Acids Res ; 45(13): e123, 2017 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-28472465

RESUMEN

Tetracycline-based inducible systems provide powerful methods for functional studies where gene expression can be controlled. However, the lack of tight control of the inducible system, leading to leakiness and adverse effects caused by undesirable tetracycline dosage requirements, has proven to be a limitation. Here, we report that the combined use of genome editing tools and last generation Tet-On systems can resolve these issues. Our principle is based on precise integration of inducible transcriptional elements (coined PrIITE) targeted to: (i) exons of an endogenous gene of interest (GOI) and (ii) a safe harbor locus. Using PrIITE cells harboring a GFP reporter or CDX2 transcription factor, we demonstrate discrete inducibility of gene expression with complete abrogation of leakiness. CDX2 PrIITE cells generated by this approach uncovered novel CDX2 downstream effector genes. Our results provide a strategy for characterization of dose-dependent effector functions of essential genes that require absence of endogenous gene expression.


Asunto(s)
Edición Génica/métodos , Regulación de la Expresión Génica , Factor de Transcripción CDX2/antagonistas & inhibidores , Factor de Transcripción CDX2/genética , Línea Celular , Exones , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Marcación de Gen , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Humanos , Tetraciclina
6.
Int J Mol Sci ; 19(7)2018 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-30011875

RESUMEN

Optimal research results rely on the selection of cellular models capable of recapitulating the characteristics of primary tumours from which they originate. The expression of mucins (MUC16 and MUC1) and truncated O-glycans (Tn, STn and T) represents a characteristic footprint of serous ovarian carcinomas (SOCs). Therefore, selecting ovarian cancer (OVCA) cell lines that reflect this phenotype is crucial to explore the putative biological role of these biomarkers in the SOC setting. Here, we investigated a panel of OVCA cell lines commonly used as SOC models, and tested whether, when cultured in 2D and 3D conditions, these recapitulate the mucin and O-glycan expression profiles of SOCs. We further explored the role of truncating the O-glycosylation capacity in OVCAR3 cells through knockout of the COSMC chaperone, using in vitro and in vivo assays. We found that the majority of OVCA cell lines of serous origin do not share the mucin and truncated O-glycan footprint of SOCs, although 3D cultures showed a higher resemblance. We also found that genetic truncation of the O-glycosylation capacity of OVCAR3 cells did not enhance oncogenic features either in vitro or in vivo. This study underscores the importance of well-characterized cellular models to study specific features of ovarian cancer.


Asunto(s)
Antígeno Ca-125/metabolismo , Cistadenocarcinoma Seroso/metabolismo , Proteínas de la Membrana/metabolismo , Mucina-1/metabolismo , Neoplasias Ováricas/metabolismo , Polisacáridos/metabolismo , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Antígeno Ca-125/genética , Línea Celular Tumoral , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología , Femenino , Perfilación de la Expresión Génica , Glicosilación , Humanos , Proteínas de la Membrana/genética , Ratones Desnudos , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mucina-1/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Fenotipo , Trasplante Heterólogo
7.
Biochim Biophys Acta ; 1852(9): 1928-39, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26144047

RESUMEN

Helicobacter pylori exploits host glycoconjugates to colonize the gastric niche. Infection can persist for decades promoting chronic inflammation, and in a subset of individuals lesions can silently progress to cancer. This study shows that H. pylori chronic infection and gastric tissue inflammation result in a remodeling of the gastric glycophenotype with increased expression of sialyl-Lewis a/x antigens due to transcriptional up-regulation of the B3GNT5, B3GALT5, and FUT3 genes. We observed that H. pylori infected individuals present a marked gastric local pro-inflammatory signature with significantly higher TNF-α levels and demonstrated that TNF-induced activation of the NF-kappaB pathway results in B3GNT5 transcriptional up-regulation. Furthermore, we show that this gastric glycosylation shift, characterized by increased sialylation patterns, favors SabA-mediated H. pylori attachment to human inflamed gastric mucosa. This study provides novel clinically relevant insights into the regulatory mechanisms underlying H. pylori modulation of host glycosylation machinery, and phenotypic alterations crucial for life-long infection. Moreover, the biosynthetic pathways here identified as responsible for gastric mucosa increased sialylation, in response to H. pylori infection, can be exploited as drug targets for hindering bacteria adhesion and counteract the infection chronicity.

8.
Glycobiology ; 25(11): 1172-82, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26201951

RESUMEN

The MUC16 mucin is overexpressed and aberrantly glycosylated in ovarian carcinomas. Immunodetection of circulating MUC16 is one of the most used cancer biomarker assays, but existing antibodies to MUC16 fail to distinguish normal and aberrant cancer glycoforms. Although all antibodies react with the tandem-repeat region, their epitopes appear to be conformational dependent and not definable by a short peptide. Aberrant glycoforms of MUC16 may constitute promising targets for diagnostic and immunotherapeutic intervention, and it is important to develop well-defined immunogens for induction of potent MUC16 immunity. Here, we developed a MUC16 vaccine based on a 1.7TR (264 aa) expressed in Escherichia coli and in vitro enzymatically glycosylated to generate the aberrant cancer-associated glycoform Tn. This vaccine elicited a potent serum IgG response in mice and we identified two major immunodominant linear peptide epitopes within the tandem repeat. We developed one monoclonal antibody, 5E11, reactive with a minimum epitope with the sequence FNTTER. This sequence contains potential N- and O-glycosylation sites and, interestingly, glycosylation blocked binding of 5E11. In immunochemistry of ovarian benign and cancer lesions, 5E11 showed similar reactivity as traditional MUC16 antibodies, suggesting that the epitope is not efficiently glycosylated. The study provides a vaccine design and immunodominant MUC16 TR epitopes.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/inmunología , Antígeno Ca-125/inmunología , Epítopos/inmunología , Proteínas de la Membrana/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales de Origen Murino/química , Antígeno Ca-125/química , Células CHO , Cricetinae , Cricetulus , Epítopos/química , Femenino , Humanos , Proteínas de la Membrana/química , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular
9.
Lab Invest ; 95(7): 718-27, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25867765

RESUMEN

De novo expression of Sialyl-Tn (STn) antigen is one of the most common features of intestinal metaplasia (IM) and gastric carcinomas, and its biosynthesis has been mostly attributed to ST6GalNAc-I activity. However, the regulation of this glycosyltransferase expression is not elucidated. In IM lesions and in the intestine, CDX2 homeobox transcription factor is co-expressed with STn and ST6GalNAc-I. We therefore hypothesized that CDX2 might induce STn expression by positive regulation of ST6GalNAc-I. We showed that ST6GalNAc-I transcript levels and CDX2 have a coordinated expression upon Caco-2 in vitro differentiation, and overexpression of CDX2 in MKN45 gastric cells increases ST6GalNAc-I transcript levels. Nine putative CDX-binding sites in the ST6GalNAc-I-regulatory sequence were identified and analyzed by chromatin immunoprecipitation in Caco-2 cells and in IM. The results showed that CDX2 protein is recruited to all regions, being the most proximal sites preferentially occupied in vivo. Luciferase assays demonstrated that CDX2 is able to transactivate ST6GalNac-I-regulatory region. The induction was stronger for the regions mapped in the neighbourhood of ATG start codon and site-directed mutagenesis of these sites confirmed their importance. In conclusion, we show that CDX2 transcriptionally regulates ST6GalNAc-I gene expression, specifically in the preneoplastic IM lesion.


Asunto(s)
Antígenos CD/metabolismo , Proteínas de Homeodominio/metabolismo , Enfermedades Intestinales/metabolismo , Lesiones Precancerosas/metabolismo , Sialiltransferasas/metabolismo , Antígenos CD/genética , Secuencia de Bases , Factor de Transcripción CDX2 , Células CACO-2 , Diferenciación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Metaplasia/metabolismo , Datos de Secuencia Molecular , Secuencias Reguladoras de Ácidos Nucleicos , Sialiltransferasas/genética
10.
Histopathology ; 66(3): 343-50, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25196071

RESUMEN

AIMS: Intestinal metaplasia (IM), which results from de-novo expression of CDX2, and dysplasia are precursor lesions of gastric cancer that are associated with an increased risk for cancer development. There is much evidence suggesting a role for the transcription factor SOX2 in gastric differentiation. The aim of this study was to attempt to establish the relationship of SOX2 with CDX2 and with the differentiation reprogramming that characterizes gastric carcinogenesis, to assess their involvement in IM and dysplasia. METHODS AND RESULTS: Characterization of gastric (SOX2, MUC5AC, and MUC6) and intestinal (CDX2 and MUC2) markers in normal gastric mucosa, in 55 foci of IM and in 26 foci of dysplasia, was performed by immunohistochemistry. SOX2 was expressed in the normal gastric mucosa, in the presumptive stem cell compartment, and was maintained in 7% of the complete (MUC5AC-negative) and 85% of the incomplete (MUC5AC-positive) IM subtypes. Twelve per cent of the dysplastic lesions expressed SOX2, and the association with MUC5AC was lost. CDX2 was present in all IMs and dysplastic lesions. CONCLUSIONS: SOX2 is associated with gastric differentiation in incomplete IM and is lost in the progression to dysplasia, whereas CDX2 is acquired de novo in IM and maintained in dysplasia. This suggests that the balance between gastric and intestinal differentiation programmes impacts on the gastric carcinogenesis cascade progression.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Mucosa Intestinal/patología , Lesiones Precancerosas/patología , Factores de Transcripción SOXB1/metabolismo , Neoplasias Gástricas/patología , Factor de Transcripción CDX2 , Diferenciación Celular/fisiología , Humanos , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Metaplasia/metabolismo , Metaplasia/patología , Lesiones Precancerosas/metabolismo , Neoplasias Gástricas/metabolismo
11.
J Infect Dis ; 210(2): 183-91, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24459192

RESUMEN

BACKGROUND: Noroviruses (NoVs) represent a considerable public health burden. Despite their enormous genetic diversity, most outbreaks are due to the single GII.4 genotype, but the reasons for this are poorly understood. NoVs use histo-blood group antigens (HBGAs) as attachment factors. Since HBGAs are present in saliva, binding of strains to saliva is commonly used as a surrogate for recognition of the gut surface by specific strains, although the relationship between saliva and gut tissue expression of HBGAs is not well defined. METHODS: The presence of fucosylated HBGAs in saliva and stomach biopsy specimens, as well as that of genogroup I.1 and genogroup II.4 virus-like particles, were compared in a series of 109 donors from Portugal. RESULTS: An overall good concordance between HBGA expression in saliva and stomach surface mucosa was observed. However, unexpected mucosal expression of α(1,2)fucosylated epitopes in nonsecretor individuals was frequently detected, allowing for GII.4 attachment. Although all individuals were infected with Helicobacter pylori, abnormal expression of α(1,2)fucosylated motifs and binding of GII.4 virus-like particles in nonsecretors' mucosa were associated with positivity for the H. pylori CagA virulence factor. CONCLUSIONS: Infection by CagA-positive H. pylori induces expression of GII.4 attachment factors in nonsecretors' mucosa, expanding the host range of these strains and thereby possibly contributing to their epidemiological dominance.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Antígenos de Grupos Sanguíneos/análisis , Mucosa Gástrica/química , Mucosa Gástrica/virología , Norovirus/aislamiento & purificación , Saliva/química , Saliva/virología , Genotipo , Voluntarios Sanos , Infecciones por Helicobacter/microbiología , Helicobacter pylori/aislamiento & purificación , Especificidad del Huésped , Interacciones Huésped-Patógeno , Humanos , Norovirus/clasificación , Norovirus/genética , Portugal , Receptores Virales/análisis , Factores de Virulencia/metabolismo
12.
J Proteome Res ; 13(7): 3349-59, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24850311

RESUMEN

The most used cancer serum biomarker is the CA125 immunoassay for ovarian cancer that detects the mucin glycoprotein MUC16. Several monoclonal antibodies (mAbs) including OC125 and M11 are used in CA125 assays. However, despite considerable efforts, our knowledge of the molecular characteristics of the recognized epitopes and the role played by glycosylation has remained elusive. Here a comprehensive set of recombinant MUC16 tandem repeats (TRs) expressed in glycoengineered mammalian cells and E. coli, together with overlapping peptides, was used to probe antigen-binding epitopes. We present a complete analysis of N- and O-glycosylation sites of a MUC16 TR expressed in CHO cells and demonstrate that neither N- nor O-glycosylation appear to substantially influence binding of OC125 and M11 mAbs. A series of successive N- and C-terminal truncations of a MUC16 TR construct expressed in E. coli narrowed down the epitopes for OC125 and M11 to a segment containing parts of two consecutive SEA domains with a linker. Thus, a complete SEA domain is not required. These findings suggest that binding epitopes of mAbs OC125 and M11 are dependent on conformation but not on glycosylation. The availability of recombinant TR constructs with and without aberrant glycosylation now opens the way for vaccine studies.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/química , Antígeno Ca-125/inmunología , Proteínas de la Membrana/inmunología , Animales , Antígeno Ca-125/química , Antígeno Ca-125/metabolismo , Células CHO , Cricetinae , Cricetulus , Mapeo Epitopo , Glicosilación , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Unión Proteica , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína
13.
BMC Cancer ; 14: 753, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25300947

RESUMEN

BACKGROUND: Gastric cancer remains a serious health concern worldwide. Patients would greatly benefit from the discovery of new biomarkers that predict outcome more accurately and allow better treatment and follow-up decisions. Here, we used a retrospective, observational study to assess the expression and prognostic value of the transcription factors SOX2 and CDX2 in gastric cancer. METHODS: SOX2, CDX2, MUC5AC and MUC2 expression were assessed in 201 gastric tumors by immunohistochemistry. SOX2 and CDX2 expression were crossed with clinicopathological and follow-up data to determine their impact on tumor behavior and outcome. Moreover, SOX2 locus copy number status was assessed by FISH (N = 21) and Copy Number Variation Assay (N = 62). RESULTS: SOX2 was expressed in 52% of the gastric tumors and was significantly associated with male gender, T stage and N stage. Moreover, SOX2 expression predicted poorer patient survival, and the combination with CDX2 defined two molecular phenotypes, SOX2+CDX2- versus SOX2-CDX2+, that predict the worst and the best long-term patients' outcome. These profiles combined with clinicopathological parameters stratify the prognosis of patients with intestinal and expanding tumors and in those without signs of venous invasion. Finally, SOX2 locus copy number gains were found in 93% of the samples reaching the amplification threshold in 14% and significantly associating with protein expression. CONCLUSIONS: We showed, for the first time, that SOX2 combined with CDX2 expression profile in gastric cancer segregate patients into different prognostic groups, complementing the clinicopathological information. We further demonstrate a molecular mechanism for SOX2 expression in a subset of gastric cancer cases.


Asunto(s)
Adenocarcinoma/metabolismo , Biomarcadores de Tumor/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción SOXB1/metabolismo , Neoplasias Gástricas/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Adenocarcinoma/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Factor de Transcripción CDX2 , Variaciones en el Número de Copia de ADN , Femenino , Amplificación de Genes , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Fenotipo , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Factores de Transcripción SOXB1/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/cirugía , Resultado del Tratamiento , Adulto Joven
14.
J Proteome Res ; 12(3): 1454-66, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23312025

RESUMEN

Gastric cancer is preceded by a carcinogenesis pathway that includes gastritis caused by Helicobacter pylori infection, chronic atrophic gastritis that may progress to intestinal metaplasia (IM), dysplasia, and ultimately gastric carcinoma of the more common intestinal subtype. The identification of glycosylation changes in circulating serum proteins in patients with precursor lesions of gastric cancer is of high interest and represents a source of putative new biomarkers for early diagnosis and intervention. This study applies a glycoproteomic approach to identify altered glycoproteins expressing the simple mucin-type carbohydrate antigens T and STn in the serum of patients with gastritis, IM (complete and incomplete subtypes), and control healthy individuals. The immunohistochemistry analysis of the gastric mucosa of these patients showed expression of T and STn antigens in gastric lesions, with STn being expressed only in IM. The serum glycoproteomic analysis using 2D-gel electrophoresis, Western blot, and MALDI-TOF/TOF mass spectrometry led to the identification of circulating proteins carrying these altered glycans. One of the glycoproteins identified was plasminogen, a protein that has been reported to play a role in H. pylori chronic infection of the gastric mucosa and is involved in extracellular matrix modeling and degradation. Plasminogen was further characterized and showed to carry STn antigens in patients with gastritis and IM. These results provide evidence of serum proteins displaying abnormal O-glycosylation in patients with precursor lesions of gastric carcinoma and include a panel of putative targets for the non-invasive clinical diagnosis of individuals with gastritis and IM.


Asunto(s)
Carbohidratos/sangre , Lesiones Precancerosas/sangre , Proteómica , Neoplasias Gástricas/sangre , Western Blotting , Cromatografía Líquida de Alta Presión , Electroforesis en Gel Bidimensional , Glicosilación , Humanos , Inmunohistoquímica , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
15.
Carcinogenesis ; 33(10): 1985-92, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22791809

RESUMEN

Helicobacter pylori infection is the main risk factor for intestinal metaplasia (IM) and gastric cancer development. IM is a pre-neoplastic lesion, induced by the transcription factor CDX2, where the gastric mucosa is converted to an intestinal phenotype. We previously demonstrated that key elements of the bone morphogenetic protein (BMP) pathway co-localize with CDX2 in IM and upregulate CDX2 expression in gastric cell lines. These observations, together with the hypothesis that CDX2 could be repressed by SOX2, led us to test whether H. pylori, through BMPs, SOX2 and CDX2 could participate in a molecular network critical for the development of IM. AGS cells with and without SMAD4 knock-down were co-cultured with H. pylori or BMP2 to assess the expression of BMP pathway members as well as CDX2 and SOX2 by qPCR and western blot. Proximity ligation assay (PLA) was also performed to evaluate SMAD proteins interaction. Immunohistochemistry and western blot were performed in gastric samples from mice infected with Helicobacter spp. to measure Smad4, pSmad1/5/8, Cdx2 and Sox2 expression in vivo. Increased expression and activity of the BMP pathway accompanied by CDX2 upregulation and SOX2 downregulation were observed in AGS cells co-cultured with H. pylori or BMP2. These effects were impaired by downregulation of the BMP pathway. Finally, infected mice present BMP pathway upregulation, focal Cdx2 expression and decreased Sox2. These results provide a novel link between H. pylori infection and the BMP pathway in the regulation of intestinal and gastric-specific genes that might be relevant for gastric IM.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Helicobacter pylori/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción SOXB1/metabolismo , Neoplasias Gástricas/metabolismo , Transactivadores/metabolismo , Animales , Factor de Transcripción CDX2 , Línea Celular Tumoral , Mucosa Gástrica/metabolismo , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Smad/genética
16.
J Cell Mol Med ; 16(7): 1474-84, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21883895

RESUMEN

Mucin glycoproteins are major secreted or membrane-bound molecules that, in cancer, show modifications in both the mucin proteins expression and in the O-glycosylation profile, generating some of the most relevant tumour markers in clinical use for decades. Thus far, the identification of these biomarkers has been based on the detection of either the protein or the O-glycan modifications. We therefore aimed to identify the combined mucin and O-glycan features, that is, specific glycoforms, in an attempt to increase specificity of these cancer biomarkers. Using in situ proximity ligation assays (PLA) based on existing monoclonal antibodies directed to MUC1, MUC2, MUC5AC and MUC6 mucins and to cancer-associated carbohydrate antigens Tn, Sialyl-Tn (STn), T, Sialyl-Le(a) (SLe(a)) and Sialyl-Le(x) (SLe(x)) we screened a series of 28 mucinous adenocarcinomas from different locations (stomach, ampulla of Vater, colon, lung, breast and ovary) to detect specific mucin glycoforms. We detected Tn/STn/SLe(a)/SLe(x)-MUC1 and STn/SLe(a)/SLe(x)-MUC2 glycoforms in ≥50% of the cases, with a variable distribution among organs. Some new glycoforms-T/SLe(a)-MUC2, STn/T/SLe(a) SLe(x)-MUC5AC and STn/T/SLe(a)/SLe(x)-MUC6-were identified for the first time in the present study in a variable percentage of cases from different organs. In conclusion, application of the PLA technique allowed sensitive detection of specific aberrant mucin glycoforms in cancer, increasing specificity to the use of antibodies either to the mucin protein backbone or to the O-glycan haptens alone.


Asunto(s)
Biomarcadores de Tumor/análisis , Mucina 5AC/análisis , Mucina-1/análisis , Mucina 2/análisis , Mucina 6/análisis , Neoplasias/diagnóstico , Adenocarcinoma Mucinoso/metabolismo , Adenocarcinoma Mucinoso/patología , Antígenos de Carbohidratos Asociados a Tumores/análisis , Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Mama/patología , Antígeno CA-19-9 , Colon/patología , Técnica del Anticuerpo Fluorescente , Gangliósidos/análisis , Gangliósidos/metabolismo , Glicosilación , Humanos , Inmunohistoquímica , Pulmón/patología , Neoplasias/patología , Oligosacáridos/análisis , Oligosacáridos/metabolismo , Antígeno Sialil Lewis X
17.
Gut ; 60(3): 290-8, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21148572

RESUMEN

BACKGROUND AND AIMS: Intestinal metaplasia (IM) is a gastric preneoplastic lesion that appears following Helicobacter pylori infection and confers an increased risk for development of cancer. It is induced by gastric expression of the intestine-specific transcription factor CDX2. The regulatory mechanisms involved in triggering and maintaining gastric CDX2 expression have not been fully elucidated. The Cdx2(+/-) mouse develops intestinal polyps with gastric differentiation and total loss of Cdx2 expression in the absence of structural loss of the second allele, suggesting a regulatory defect. This putative haplo-insufficiency, together with the apparent stability of IM, led to the hypothesis that CDX2 regulates its own expression through an autoregulatory loop in both contexts. METHODS: Gastrointestinal cell lines were co-transfected with wild-type or mutated Cdx2 promoter constructs and CDX2 expression vector for luciferase assays. Transfection experiments were also used to assess endogenous CDX2 autoregulation, evaluated by RT-PCR, qPCR and western blotting. Chromatin immunoprecipitation was performed in a cell line, mouse ileum and human IM. RESULTS: CDX2 binds to and transactivates its own promoter and positively regulates its expression in gastrointestinal human carcinoma cell lines. Furthermore, CDX2 is bound to its promoter in the mouse ileum and in human gastric IM, providing a major contribution to understanding the relevance of this autoregulatory pathway in vivo. CONCLUSION: The results of this study demonstrate another layer of complexity in CDX2 regulation by an effective autoregulatory loop which may have a major impact on the stability of human IM, possibly resulting in the inevitable progression of the gastric carcinogenesis pathway.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Lesiones Precancerosas/metabolismo , Neoplasias Gástricas/metabolismo , Estómago/patología , Adenocarcinoma/metabolismo , Animales , Factor de Transcripción CDX2 , Regulación de la Expresión Génica/genética , Proteínas de Homeodominio/genética , Homeostasis/genética , Homeostasis/fisiología , Humanos , Íleon/metabolismo , Metaplasia/metabolismo , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Mutación Puntual , Lesiones Precancerosas/genética , Regiones Promotoras Genéticas/genética , Neoplasias Gástricas/genética , Transfección , Células Tumorales Cultivadas
18.
Cancers (Basel) ; 14(11)2022 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-35681562

RESUMEN

SARS-CoV-2 pandemics have been massively characterized on a global scale by the rapid generation of in-depth genomic information. The main entry gate of SARS-CoV-2 in human cells is the angiotensin-converting enzyme 2 (ACE2) receptor. The expression of this protein has been reported in several human tissues, suggesting a correlation between SARS-CoV-2 organotropism and ACE2 distribution. In this study, we selected (a series of) 90 patients who were submitted to surgery for tumor removal between the beginning of the SARS-CoV-2 pandemic and the closure of operating rooms (by the end of March 2020) in two different countries-Portugal and Brazil. We evaluated the expressions of ACE2 and furin (another important factor for virus internalization) in colon (n = 60), gastric (n = 19), and thyroid (n = 11) carcinomas. In a subseries of cases with PCR results for SARS-CoV-2 detection in the peri-operatory window (n = 18), we performed different methodological approaches for viral detections in patient tumor samples. Our results show that colon and gastric carcinomas display favorable microenvironments to SARS-CoV-2 tropism, presenting high expression levels of ACE2 and furin. From the subseries of 18 cases, 11 tested positive via PCR detection performed in tumor blocks; however, a direct association between the ACE2 expression and SARS-CoV-2 infection was not demonstrated in cancer cells using histology-based techniques, such as immunohistochemistry or in situ hybridization. This study raises the possibility of ACE2-mediated viral tropism in cancer tissues to be clarified in future studies.

19.
Nat Commun ; 13(1): 4755, 2022 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-35963857

RESUMEN

Determining the levels of protein-protein interactions is essential for the analysis of signaling within the cell, characterization of mutation effects, protein function and activation in health and disease, among others. Herein, we describe MolBoolean - a method to detect interactions between endogenous proteins in various subcellular compartments, utilizing antibody-DNA conjugates for identification and signal amplification. In contrast to proximity ligation assays, MolBoolean simultaneously indicates the relative abundances of protein A and B not interacting with each other, as well as the pool of A and B proteins that are proximal enough to be considered an AB complex. MolBoolean is applicable both in fixed cells and tissue sections. The specific and quantifiable data that the method generates provide opportunities for both diagnostic use and medical research.


Asunto(s)
Mapeo de Interacción de Proteínas , Proteínas , Mapeo de Interacción de Proteínas/métodos , Proteínas/metabolismo , Transducción de Señal
20.
J Clin Invest ; 118(6): 2325-36, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18483624

RESUMEN

Chronic Helicobacter pylori infection is recognized as a cause of gastric cancer. H. pylori adhesion to gastric cells is mediated by bacterial adhesins such as sialic acid-binding adhesin (SabA), which binds the carbohydrate structure sialyl-Lewis x. Sialyl-Lewis x expression in the gastric epithelium is induced during persistent H. pylori infection, suggesting that H. pylori modulates host cell glycosylation patterns for enhanced adhesion. Here, we evaluate changes in the glycosylation-related gene expression profile of a human gastric carcinoma cell line following H. pylori infection. We observed that H. pylori significantly altered expression of 168 of the 1,031 human genes tested by microarray, and the extent of these alterations was associated with the pathogenicity of the H. pylori strain. A highly pathogenic strain altered expression of several genes involved in glycan biosynthesis, in particular that encoding beta3 GlcNAc T5 (beta3GnT5), a GlcNAc transferase essential for the biosynthesis of Lewis antigens. beta3GnT5 induction was specific to infection with highly pathogenic strains of H. pylori carrying a cluster of genes known as the cag pathogenicity island, and was dependent on CagA and CagE. Further, beta3GnT5 overexpression in human gastric carcinoma cell lines led to increased sialyl-Lewis x expression and H. pylori adhesion. This study identifies what we believe to be a novel mechanism by which H. pylori modulates the biosynthesis of the SabA ligand in gastric cells, thereby strengthening the epithelial attachment necessary to achieve successful colonization.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Helicobacter pylori/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Oligosacáridos/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/microbiología , Secuencia de Carbohidratos , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Glicosilación , Humanos , Ligandos , Modelos Biológicos , Datos de Secuencia Molecular , Filogenia , Antígeno Sialil Lewis X , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA