Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biomolecules ; 13(10)2023 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-37892208

RESUMO

The androgens/androgen receptor (AR) axis is the main therapeutic target in prostate cancer (PCa). However, while initially responsive, a subset of tumors loses AR expression through mechanisms putatively associated with epigenetic modifications. In this study, we assessed the link between the presence of CpG methylation in the 5'UTR and promoter regions of AR and loss of AR expression. Hence, we characterized and compared the methylation signature at CpG resolution of these regulatory regions in vitro, both at basal levels and following treatment with 5-aza-2-deoxycytidine (DAC) alone, or in combination with Trichostatin A (TSA). Our results showed heterogeneity in the methylation signature of AR negative cell lines and pinpointed the proximal promoter region as the most consistently methylated site in DU-145. Furthermore, this region was extremely resistant to the demethylating effects of DAC and was only significantly demethylated upon concomitant treatment with TSA. Nevertheless, no AR re-expression was detected at the mRNA or protein level. Importantly, after treatment, there was a significant increase in repressive histone marks at AR region 1 in DU-145 cells. Altogether, our data indicate that AR region 1 genomic availability is crucial for AR expression and that the inhibition of histone methyltransferases might hold promise for AR re-expression.


Assuntos
Androgênios , Neoplasias da Próstata , Masculino , Humanos , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Metilação de DNA , Linhagem Celular Tumoral , Regiões Promotoras Genéticas , Neoplasias da Próstata/metabolismo , Epigênese Genética/genética , Regulação Neoplásica da Expressão Gênica
2.
Exp Cell Res ; 418(2): 113282, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35841980

RESUMO

The Ser/Thr-protein phosphatase PP1 (PP1) is a positive regulator of the androgen receptor (AR), which suggests major roles for PP1 in prostate carcinogenesis. However, studies dedicated to the characterization of PP1 in PCa are currently scarce. Here we analyzed the expression and localization of the PP1 catalytic (PP1c) isoforms in formalin-fixed, paraffin-embedded prostate tissue samples, as well as in PCa cell lines. We also analyzed well-characterized PCa cohorts to determine their transcript levels, identify genetic alterations, and assess promoter methylation of PP1c-coding genes. We found that PP-1A was upregulated and relocalized towards the nucleus in PCa and that PPP1CA was frequently amplified in PCa, particularly in advanced stages. PP-1B was downregulated in PCa but upregulated in a subset of tumors with AR amplification. PP-1G transcript levels were found to be associated with Gleason score. PP1c-coding genes were rarely mutated in PCa and were not prone to regulation by promoter methylation. Protein phosphorylation, on the other hand, might be an important regulatory mechanism of PP1c isoforms' activity. Altogether, our results suggest differential expression, localization, and regulation of PP1c isoforms in PCa and support the need for investigating isoform-specific roles in prostate carcinogenesis in future studies.


Assuntos
Núcleo Celular , Neoplasias da Próstata , Carcinogênese/metabolismo , Núcleo Celular/metabolismo , Humanos , Masculino , Fosforilação , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína Fosfatase 1/genética , Proteína Fosfatase 1/metabolismo
3.
Epigenetics ; 17(5): 564-588, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34130596

RESUMO

Epigenome editing consists of fusing a predesigned DNA recognition unit to the catalytic domain of a chromatin modifying enzyme leading to the introduction or removal of an epigenetic mark at a specific locus. These platforms enabled the study of the mechanisms and roles of epigenetic changes in several research domains such as those addressing pathogenesis and progression of cancer. Despite the continued efforts required to overcome some limitations, which include specificity, off-target effects, efficacy, and longevity, these tools have been rapidly progressing and improving.Since prostate cancer is characterized by multiple genetic and epigenetic alterations that affect different signalling pathways, epigenetic editing constitutes a promising strategy to hamper cancer progression. Therefore, by modulating chromatin structure through epigenome editing, its conformation might be better understood and events that drive prostate carcinogenesis might be further unveiled.This review describes the different epigenome engineering tools, their mechanisms concerning gene's expression and regulation, highlighting the challenges and opportunities concerning prostate cancer research.


Assuntos
Edição de Genes , Neoplasias da Próstata , Sistemas CRISPR-Cas , Cromatina , Metilação de DNA , Epigênese Genética , Humanos , Masculino , Neoplasias da Próstata/genética
4.
Pharmaceuticals (Basel) ; 14(7)2021 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-34358096

RESUMO

Among the well-established alterations contributing to prostate cancer (PCa) pathogenesis, epigenetics is an important player in its development and aggressive disease state. Moreover, since no curative therapies are available for advanced stage disease, there is an urgent need for novel therapeutic strategies targeting this subset of patients. Thus, we aimed to evaluate the combined antineoplastic effects of DNA methylation inhibitor hydralazine and histone deacetylase inhibitors panobinostat and valproic acid in several prostate cell lines. The effect of these drugs was assessed in four PCa (LNCaP, 22Rv1, DU145 and PC-3) cell lines, as well as in non-malignant epithelial (RWPE-1) and stromal (WPMY-1) cell lines, using several assays to evaluate cell viability, apoptosis, proliferation, DNA damage and clonogenic potential. We found that exposure to each epidrug separately reduced viability of all PCa cells in a dose-dependent manner and that combined treatments led to synergic growth inhibitory effects, impacting also on colony formation, invasion, apoptotic and proliferation rates. Interestingly, antitumoral effects of combined treatment were particularly expressive in DU145 cells. We concluded that hydralazine and panobinostat attenuate malignant properties of PCa cells, constituting a potential therapeutic tool to counteract PCa progression.

5.
Biomedicines ; 9(8)2021 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-34440180

RESUMO

Advanced prostate cancers frequently develop resistance to androgen-deprivation therapy with serious implications for patient survival. Considering their importance in this type of neoplasia, epigenetic modifications have drawn attention as alternative treatment strategies. The aim of this study was to assess the antitumoral effects of the combination of hydralazine, a DNA methylation inhibitor, with enzalutamide, an antagonist of the androgen receptor, in prostate cancer cell lines. Several biological parameters, such as cell viability, proliferation, DNA damage, and apoptosis, as well as clonogenic and invasive potential, were evaluated. The individual treatments with hydralazine and enzalutamide exerted growth-inhibitory effects in prostate cancer cells and their combined treatment displayed synergistic effects. The combination of these two drugs was very effective in decreasing malignant features of prostate cancer and may become an alternative therapeutic option for prostate cancer patient management.

6.
EBioMedicine ; 69: 103462, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34229278

RESUMO

BACKGROUND: Gastric inflammation is a major risk factor for gastric cancer. Current endoscopic methods are not able to efficiently detect and characterize gastric inflammation, leading to a sub-optimal patients' care. New non-invasive methods are needed. Reflectance mucosal light analysis is of particular interest in this context. The aim of our study was to analyze reflectance light and specific autofluorescence signals, both in humans and in a mouse model of gastritis. METHODS: We recruited patients undergoing gastroendoscopic procedure during which reflectance was analysed with a multispectral camera. In parallel, the gastritis mouse model of Helicobacter pylori infection was used to investigate reflectance from ex vivo gastric samples using a spectrometer. In both cases, autofluorescence signals were measured using a confocal microscope. FINDINGS: In gastritis patients, reflectance modifications were significant in near-infrared spectrum, with a decrease between 610 and 725 nm and an increase between 750 and 840 nm. Autofluorescence was also modified, showing variations around 550 nm of emission. In H. pylori infected mice developing gastric inflammatory lesions, we observed significant reflectance modifications 18 months after infection, with increased intensity between 617 and 672 nm. Autofluorescence was significantly modified after 1, 3 and 6 months around 550 and 630 nm. Both in human and in mouse, these reflectance data can be considered as biomarkers and accurately predicted inflammatory state. INTERPRETATION: In this pilot study, using a practical measuring device, we identified in humans, modification of reflectance spectra in the visible spectrum and for the first time in near-infrared, associated with inflammatory gastric states. Furthermore, both in the mouse model and humans, we also observed modifications of autofluorescence associated with gastric inflammation. These innovative data pave the way to deeper validation studies on larger cohorts, for further development of an optical biopsy system to detect gastritis and finally to better surveil this important gastric cancer risk factor. FUNDING: The project was funded by the ANR EMMIE (ANR-15-CE17-0015) and the French Gastroenterology Society (SNFGE).


Assuntos
Gastrite/diagnóstico por imagem , Gastroscopia/métodos , Imagem Multimodal/métodos , Imagem Óptica/métodos , Adulto , Idoso , Animais , Feminino , Fluorescência , Gastrite/microbiologia , Gastrite/patologia , Helicobacter pylori/patogenicidade , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Imagem Multimodal/instrumentação , Imagem Óptica/instrumentação , Gravação em Vídeo/métodos
7.
Cancers (Basel) ; 12(10)2020 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-33050470

RESUMO

Novel treatment options are needed for testicular germ cell tumor (TGCT) patients, particularly important for those showing or developing cisplatin resistance, the major cause of cancer-related deaths. As TGCTs pathobiology is highly related to epigenetic (de)regulation, epidrugs are potentially effective therapies. Hence, we sought to explore, for the first time, the effect of the two most recently FDA-approved HDAC inhibitors (HDACis), belinostat and panobinostat, in (T)GCT cell lines including those resistant to cisplatin. In silico results were validated in 261 patient samples and differential expression of HDACs was also observed across cell lines. Belinostat and panobinostat reduced cell viability in both cisplatin-sensitive cells (NCCIT-P, 2102Ep-P, and NT2-P) and, importantly, also in matched cisplatin-resistant subclones (NCCIT-R, 2102Ep-R, and NT2-R), with IC50s in the low nanomolar range for all cell lines. Treatment of NCCIT-R with both drugs increased acetylation, induced cell cycle arrest, reduced proliferation, decreased Ki67 index, and increased p21, while increasing cell death by apoptosis, with upregulation of cleaved caspase 3. These findings support the effectiveness of HDACis for treating TGCT patients in general, including those developing cisplatin resistance. Future studies should explore them as single or combination agents.

8.
Pharmaceutics ; 12(5)2020 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-32365701

RESUMO

Epigenetic alterations, as a cancer hallmark, are associated with cancer initiation, progression and aggressiveness. Considering, however, that these alterations are reversible, drugs that target epigenetic machinery may have an inhibitory effect upon cancer treatment. The traditional drug discovery pathway is time-consuming and expensive, and thus, new and more effective strategies are required. Drug Repurposing (DR) comprises the discovery of a new medical indication for a drug that is approved for another indication, which has been recalled, that was not accepted or failed to prove efficacy. DR presents several advantages, mainly reduced resources, absence of the initial target discovery process and the reduced time necessary for the drug to be commercially available. There are numerous old drugs that are under study as repurposed epigenetic inhibitors which have demonstrated promising results in in vitro tumor models. Herein, we summarize the DR process and explore several repurposed drugs with different epigenetic targets that constitute promising candidates for cancer treatment, highlighting their mechanisms of action.

9.
Arthritis Res Ther ; 21(1): 46, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30717755

RESUMO

BACKGROUND: Spondyloarthritis (SpA) is a chronic inflammatory disease affecting primarily axial and peripheral joints and sometimes also extra-articular organs, such as the gut. Rats transgenic for HLA-B27 and human ß2-microglobulin (B27-Tg rat) develop clinical manifestations resembling human disease. In this model, it has been shown that CD103+ conventional dendritic cells (cDCs) exhibited altered functions, likely promoting SpA development. CD4- cDC subpopulation expressing XCR1, a chemokine receptor involved in their migration, have been described to be tolerogenic in steady state. Thus, in this study, we wished to examine the fate of XCR1+ cDCs in this animal model of SpA. METHODS: cDC populations were isolated from the spleen, mesenteric lymph nodes (MLN), and colonic lamina propria from B27-TG and control nontransgenic (NTG) and/or HLA-B7 transgenic rats after collagenase digestion and density gradient and characterized with flow cytometry or real-time PCR. Migration of cDCs from intestinal mucosa to MLN was assessed, using TLR-7 stimulation with Resiquimod. RESULTS: We observed a reduced frequency of cCD4- DCs in B27-Tg rats, as compared to control rats. Furthermore, such decrease was not due to excessive death of CD4- cDCs in B27-Tg rats. Interestingly, we observed a decrease frequency of the XCR1+ subpopulation among CD4- cDCs in the spleen, MLN, and lamina propria from B27-Tg rats. Finally, after TLR-7 stimulation, the migration of XCR1+ cDCs to MLN was proportionally reduced in B27-Tg rats. CONCLUSION: Our results demonstrate for the first time a decreased proportion of the tolerogenic XCR1+ cDC subpopulation in SpA target organs in B27-Tg rat, which may affect the maintenance of self-tolerance and control of inflammation.


Assuntos
Células Dendríticas/imunologia , Antígeno HLA-B27/imunologia , Receptores de Quimiocinas/imunologia , Espondilartrite/imunologia , Microglobulina beta-2/imunologia , Animais , Antígenos CD/imunologia , Movimento Celular/genética , Movimento Celular/imunologia , Células Cultivadas , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Antígeno HLA-B27/genética , Antígeno HLA-B27/metabolismo , Humanos , Tolerância Imunológica/genética , Tolerância Imunológica/imunologia , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Cadeias alfa de Integrinas/imunologia , Ratos Endogâmicos F344 , Ratos Transgênicos , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Espondilartrite/genética , Espondilartrite/metabolismo , Microglobulina beta-2/genética , Microglobulina beta-2/metabolismo
10.
J Oncol ; 2019: 5415761, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32082377

RESUMO

Helicobacter pylori infection causes chronic gastritis and is the major risk factor of gastric cancer. H. pylori induces a chronic inflammation-producing reactive oxygen species (ROS) which is a source of chromosome instabilities and contributes to the development of malignancy. H. pylori also promotes DNA hypermethylation, known to dysregulate essential genes that maintain genetic stability. The maintenance of telomere length by telomerase is essential for chromosome integrity. Telomerase reverse transcriptase (TERT) is the catalytic component of telomerase activity and an important target during host-pathogen interaction. We aimed to investigate the consequences of H. pylori on the regulation of TERT gene expression and telomerase activity. In vitro, hTERT mRNA levels and telomerase activity were analysed in H. pylori-infected human gastric epithelial cells. In addition, C57BL/6 and INS-GAS mice were used to investigate the influence of H. pylori-induced inflammation on TERT levels. Our data demonstrated that, in vitro, H. pylori inhibits TERT gene expression and decreases the telomerase activity. The exposure of cells to lycopene, an antioxidant compound, restores TERT levels in infected cells, indicating that ROS are implicated in this downregulation. In vivo, fewer TERT-positive cells are observed in gastric tissues of infected mice compared to uninfected, more predominantly in the vicinity of large aggregates of lymphocytes, suggesting an inflammation-mediated regulation. Furthermore, H. pylori appears to downregulate TERT gene expression through DNA hypermethylation as shown by the restoration of TERT transcript levels in cells treated with 5'-azacytidine, an inhibitor of DNA methylation. This was confirmed in infected mice, by PCR-methylation assay of the TERT gene promoter. Our data unraveled a novel way for H. pylori to promote genome instabilities through the inhibition of TERT levels and telomerase activity. This mechanism could play an important role in the early steps of gastric carcinogenesis.

12.
Helicobacter ; 22 Suppl 12017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28891130

RESUMO

Helicobacter pylori is responsible for the most commonly found infection in the world's population. It is the major risk factor for gastric cancer development. Numerous studies published over the last year provide new insights into the strategies employed by H. pylori to adapt to the extreme acidic conditions of the gastric environment, to establish persistent infection and to deregulate host functions, leading to gastric pathogenesis and cancer. In this review, we report recent data on the mechanisms involved in chemotaxis, on the essential role of nickel in acid resistance and gastric colonization, on the importance of adhesins and Hop proteins and on the role of CagPAI-components and CagA. Among the host functions, a special focus has been made on the escape from immune response, the ability of bacteria to induce genetic instability and modulate telomeres, the mechanism of autophagy and the deregulation of micro RNAs.


Assuntos
Infecções por Helicobacter/fisiopatologia , Helicobacter pylori/fisiologia , Helicobacter pylori/patogenicidade , Adaptação Fisiológica , Interações Hospedeiro-Patógeno , Humanos , Virulência , Fatores de Virulência/metabolismo
13.
Int J Surg Pathol ; 25(3): 262-265, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27708180

RESUMO

We report a case of metaplastic columnar epithelium in the mid-esophagus in a patient with history of caustic ingestion. A cardiac-type gastric phenotype, with early signs of intestinalization, was confirmed by immunohistochemistry studies (MUC5AC, MUC6, SOX2, and CDX2). Nonmetaplastic mucosa had histologic evidence of gastroesophageal reflux. In this case, esophageal reepithelization seems to have been modulated by acidic gastroesophageal reflux, which might activate transcription factors leading to phenotypic reprogramming of the regenerative epithelium. Most interestingly, it is a clinical example showcasing the origin of columnar metaplasia from stem cells located within the esophageal epithelium.


Assuntos
Queimaduras Químicas/patologia , Cáusticos/toxicidade , Células Epiteliais/patologia , Estenose Esofágica/induzido quimicamente , Esôfago/patologia , Idoso , Biomarcadores/análise , Humanos , Imuno-Histoquímica , Masculino , Metaplasia
14.
Sci Rep ; 6: 34860, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27721458

RESUMO

Dies1/VISTA induces embryonic stem-cell differentiation, via BMP-pathway, but also acts as inflammation regulator and immune-response modulator. Dies1 inhibition in a melanoma-mouse model led to increased tumour-infiltrating T-cells and decreased tumour growth, emphasizing Dies1 relevance in tumour-microenvironment. Dies1 is involved in cell de/differentiation, inflammation and cancer processes, which mimic those associated with Epithelial-to-Mesenchymal-Transition (EMT). Despite this axis linking Dies1 with EMT and cancer, its expression, modulation and relevance in these contexts is unknown. To address this, we analysed Dies1 expression, its regulation by promoter-methylation and miR-125a-5p overexpression, and its association with BMP-pathway downstream-effectors, in a TGFß1-induced EMT-model, cancer cell-lines and primary samples. We detected promoter-methylation as a mechanism controlling Dies1 expression in our EMT-model and in several cancer cell-lines. We showed that the relationship between Dies1 expression and BMP-pathway effectors observed in the EMT-model, was not present in all cell-lines, suggesting that Dies1 has other cell-specific effectors, beyond the BMP-pathway. We further demonstrated that: Dies1 expression loss is a recurrent event in GC, caused by promoter methylation and/or miR-125a-5p overexpression and; GC-microenvironment myofibroblasts overexpress Dies1. Our findings highlight Dies1 as a novel player in GC, with distinct roles within tumour cells and in the tumour-microenvironment.


Assuntos
Antígenos B7/genética , Epigênese Genética , Neoplasias Gástricas/genética , Linhagem Celular Tumoral , Metilação de DNA , Regulação para Baixo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Proteína 2 Inibidora de Diferenciação/genética , Proteínas Inibidoras de Diferenciação/genética , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas , Neoplasias Gástricas/patologia , Fator de Crescimento Transformador beta1/farmacologia , Microambiente Tumoral/genética
15.
Dis Markers ; 2016: 1532791, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27766003

RESUMO

Barrett's esophagus (BE) is the replacement of the normal esophageal squamous epithelium by a columnar lining epithelium. It is a premalignant condition for the development of adenocarcinoma of the esophagus and esophagogastric junction. BE is associated with gastroesophageal reflux which might change the expression profile of key transcription factors involved in the establishment of tissue differentiation, namely, SOX2 (associated with esophageal and gastric differentiation) and CDX2 (associated with intestinal differentiation). Here, we sought to characterize the expression profile of SOX2 and CDX2 in the sequential alterations of the esophageal mucosa towards adenocarcinoma and compare it with the well-established gastric and intestinal mucin profiles (MUC5AC, MUC6, and MUC2). We observed that SOX2 and CDX2 expression correlates with gastric and intestinal differentiation in BE, defined by morphological parameters and mucin expression. We show the presence of a complete intestinal profile in BE, without gastric mucins and without SOX2, and we observed an evolutionary modulation of the metaplastic phenotype by SOX2 and CDX2. We observed that adenocarcinomas harbor more frequently a mixed gastric and intestinal phenotype. In conclusion, our study establishes a role for transcription factors SOX2 and CDX2 in the progression from gastric to gastrointestinal differentiation in Barrett's metaplasia.


Assuntos
Adenocarcinoma/metabolismo , Esôfago de Barrett/metabolismo , Biomarcadores Tumorais/metabolismo , Fator de Transcrição CDX2/metabolismo , Mucosa Esofágica/metabolismo , Neoplasias Gastrointestinais/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Adenocarcinoma/patologia , Esôfago de Barrett/patologia , Biomarcadores Tumorais/genética , Fator de Transcrição CDX2/genética , Mucosa Esofágica/patologia , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Neoplasias Gastrointestinais/patologia , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Mucinas/genética , Mucinas/metabolismo , Fatores de Transcrição SOXB1/genética
16.
Mol Biol Evol ; 33(1): 93-108, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26420451

RESUMO

The human kallikrein (KLK) cluster, located at chromosome 19q13.3-13.4, encodes 15 serine proteases, including neighboring genes (KLK3, KLK2, KLK4, and KLK5) with key roles in the cascades of semen liquefaction, tooth enamel maturation, and skin desquamation. KLK2 and KLK3 were previously identified as targets of adaptive evolution in primates through different mechanisms linked to reproductive biology and, in humans, genome-wide scans of positive selection captured, a yet unexplored, evidence for KLK neutrality departure in East Asians. We perform a detailed evaluation of KLK3-KLK5 variability in the 1000 Genomes samples from East Asia, Europe, and Africa, which was sustained by our own sequencing. In East Asians, we singled out a 70-kb region surrounding KLK4 that combined unusual low levels of diversity, high frequency variants with significant levels of population differentiation (FST > 0.5) and fairly homogenous haplotypes given the large local recombination rates. Among these variants, rs1654556_G, rs198968_T, and rs17800874_A stand out for their location on putative regulatory regions and predicted functional effects, namely the introduction of several microRNA binding sites and a repressor motif. Our functional assays carried out in different cellular models showed that rs198968_T and rs17800874_A operate synergistically to reduce KLK4 expression and could be further assisted by rs1654556_G. Considering the previous findings that KLK4 inactivation causes enamel malformations in humans and mice, and that this gene is coexpressed in epidermal layers along with several substrates involved in either cell adhesion or keratinocyte differentiation, we propose KLK4 as another target of selection in East Asians correlated to tooth and epidermal morphological traits.


Assuntos
Povo Asiático/genética , Regulação para Baixo/genética , Variação Genética/genética , Calicreínas/genética , Evolução Molecular , Ásia Oriental , Humanos , Seleção Genética/genética
17.
Histopathology ; 66(3): 343-50, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25196071

RESUMO

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.


Assuntos
Proteínas de Homeodomínio/metabolismo , Mucosa Intestinal/patologia , Lesões Pré-Cancerosas/patologia , Fatores de Transcrição SOXB1/metabolismo , Neoplasias Gástricas/patologia , Fator de Transcrição CDX2 , Diferenciação Celular/fisiologia , Humanos , Imuno-Histoquímica , Mucosa Intestinal/metabolismo , Metaplasia/metabolismo , Metaplasia/patologia , Lesões Pré-Cancerosas/metabolismo , Neoplasias Gástricas/metabolismo
18.
BMC Cancer ; 14: 753, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25300947

RESUMO

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.


Assuntos
Adenocarcinoma/metabolismo , Biomarcadores Tumorais/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Neoplasias Gástricas/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/mortalidade , Adenocarcinoma/cirurgia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/genética , Fator de Transcrição CDX2 , Variações do Número de Cópias de DNA , Feminino , Amplificação de Genes , Proteínas de Homeodomínio/genética , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Fenótipo , Prognóstico , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Fatores de Transcrição SOXB1/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidade , Neoplasias Gástricas/cirurgia , Resultado do Tratamento , Adulto Jovem
19.
Carcinogenesis ; 33(10): 1985-92, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22791809

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Helicobacter pylori/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Neoplasias Gástricas/metabolismo , Transativadores/metabolismo , Animais , Fator de Transcrição CDX2 , Linhagem Celular Tumoral , Mucosa Gástrica/metabolismo , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Smad/genética
20.
Exp Parasitol ; 126(4): 526-35, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20547157

RESUMO

We have previously identified the expression of an estradiol (E2)-related molecule by Schistosoma haematobium total antigen (Sh). We now show that this molecule has an antagonistic effect of estradiol in vitro. Our results are consistent with the existence of an estrogenic molecule that antagonizes the activity of estradiol. We found evidence for this molecule as we identified and characterized by mass spectrometry new estrogenic molecules previously unknown, present in schistosome worm extracts and sera of Schistosoma-infected individuals. We also show that Sh is able to interact in vitro with estrogen receptor (ER), explaining how host endocrine system can favor the establishment of schistosomes. These findings highlight the exploitation of the host endocrine system by schistosomes and represent an additional regulatory component of schistosome development that defines a novel paradigm enabling host-parasite interactions. The identification of these molecules opens new ways for the development of alternative drugs to treat schistosomiasis.


Assuntos
Antagonistas de Estrogênios/isolamento & purificação , Estrogênios/isolamento & purificação , Receptores de Estrogênio/imunologia , Schistosoma haematobium/imunologia , Animais , Antígenos de Helmintos/química , Antígenos de Helmintos/imunologia , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Regulação para Baixo , Estradiol/imunologia , Antagonistas de Estrogênios/imunologia , Estrogênios/imunologia , Feminino , Humanos , Lactoferrina/antagonistas & inibidores , Lactoferrina/imunologia , Mesocricetus , Receptores de Estrogênio/antagonistas & inibidores , Schistosoma haematobium/genética , Esquistossomose Urinária/parasitologia , Esquistossomose Urinária/urina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA