Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 108
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Curr Top Microbiol Immunol ; 444: 157-184, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38231218

RESUMEN

Gastric cancer remains an important global health burden. Helicobacter pylori is the major etiological factor in gastric cancer, infecting the stomach of almost half of the population worldwide. Recent progress in microbiome research offered a new perspective on the complexity of the microbial communities of the stomach. Still, the role of the microbiome of the stomach beyond H. pylori in gastric carcinogenesis is not well understood and requires deeper investigation. The gastric bacterial communities of gastric cancer patients are distinct from those of patients without cancer, but the microbial alterations that occur along the process of gastric carcinogenesis, and the mechanisms through which microorganisms influence cancer progression still need to be clarified. Except for Epstein-Barr virus, the potential significance of the virome and of the mycobiome in gastric cancer have received less attention. This chapter updates the current knowledge regarding the gastric microbiome, including bacteria, viruses, and fungi, within the context of H. pylori-mediated carcinogenesis. It also reviews the possible roles of the local gastric microbiota, as well as the microbial communities of the oral and gut ecosystems, as biomarkers for gastric cancer detection. Finally, it discusses future perspectives and acknowledges limitations in the area of microbiome research in the gastric cancer setting, to which further research efforts should be directed. These will be fundamental not only to increase our current understanding of host-microbial interactions but also to facilitate translation of the findings into innovative preventive, diagnostic, and therapeutic strategies to decrease the global burden of gastric cancer.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Helicobacter pylori , Microbiota , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/etiología , Helicobacter pylori/genética , Herpesvirus Humano 4/genética , Carcinogénesis
2.
Cell Commun Signal ; 22(1): 152, 2024 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-38414029

RESUMEN

BACKGROUND: Germline mutations of E-cadherin contribute to hereditary diffuse gastric cancer (HDGC) and congenital malformations, such as oral facial clefts (OFC). However, the molecular mechanisms through which E-cadherin loss-of-function triggers distinct clinical outcomes remain unknown. We postulate that E-cadherin-mediated disorders result from abnormal interactions with the extracellular matrix and consequent aberrant intracellular signalling, affecting the coordination of cell migration. METHODS: Herein, we developed in vivo and in vitro models of E-cadherin mutants associated with either OFC or HDGC. Using a Drosophila approach, we addressed the impact of the different variants in cell morphology and migration ability. By combining gap closure migration assays and time-lapse microscopy, we further investigated the migration pattern of cells expressing OFC or HDGC variants. The adhesion profile of the variants was evaluated using high-throughput ECM arrays, whereas RNA sequencing technology was explored for identification of genes involved in aberrant cell motility. RESULTS: We have demonstrated that cells expressing OFC variants exhibit an excessive motility performance and irregular leading edges, which prevent the coordinated movement of the epithelial monolayer. Importantly, we found that OFC variants promote cell adhesion to a wider variety of extracellular matrices than HDGC variants, suggesting higher plasticity in response to different microenvironments. We unveiled a distinct transcriptomic profile in the OFC setting and pinpointed REG1A as a putative regulator of this outcome. Consistent with this, specific RNAi-mediated inhibition of REG1A shifted the migration pattern of OFC expressing cells, leading to slower wound closure with coordinated leading edges. CONCLUSIONS: We provide evidence that E-cadherin variants associated with OFC activate aberrant signalling pathways that support dynamic rearrangements of cells towards improved adaptability to the microenvironment. This proficiency results in abnormal tissue shaping and movement, possibly underlying the development of orofacial malformations.


Asunto(s)
Adenocarcinoma , Neoplasias Gástricas , Humanos , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular , Movimiento Celular , Mutación de Línea Germinal , Litostatina/genética , Neoplasias Gástricas/metabolismo , Microambiente Tumoral , Animales , Drosophila melanogaster
3.
Helicobacter ; 29(1): e13031, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37997013

RESUMEN

BACKGROUND: Outer membrane vesicles (OMVs) are spontaneously released by Gram-negative bacteria and influence bacteria-host interactions by acting as a delivery system for bacterial components and by interacting directly with host cells. Helicobacter pylori, a pathogenic bacterium that chronically colonizes the human stomach, also sheds OMVs, and their impact on bacterial-mediated diseases is still being elucidated. MATERIALS AND METHODS: Transcriptomic profiling of the human gastric cell line MKN74 upon challenge with H. pylori OMVs compared to control and infected cells was performed using the Ion AmpliSeq™ Transcriptome Human Gene Expression Panel to understand the gene expression changes that human gastric epithelial cells might undergo when exposed to H. pylori OMVs. RESULTS: H. pylori OMVs per se modify the gene expression profile of gastric epithelial cells, adding another layer of (gene) regulation to the already complex host-bacteria interaction. The most enriched pathways include those related to amino acid metabolism, mitogen-activated protein kinase signaling, autophagy, and ferroptosis, whereas the cell cycle, DNA replication, and DNA repair were the most downregulated. The transcriptomic changes induced by OMVs were mostly similar to those induced by the parental bacteria, likely amplifying the effects of the bacterium itself. CONCLUSIONS: Our data provide a valuable portrayal of the transcriptomic remodeling of gastric cells induced by H. pylori OMVs. It demonstrates the breadth of cellular pathways and genes affected by OMVs, most previously unreported, which can be further dissected for the underlying molecular mediators and explored to understand the pathobiology of the full spectrum of H. pylori-mediated diseases.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Humanos , Helicobacter pylori/fisiología , Transcriptoma , Infecciones por Helicobacter/microbiología , Estómago , Perfilación de la Expresión Génica
4.
J Infect Dis ; 226(12): 2226-2237, 2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36173814

RESUMEN

BACKGROUND: Helicobacter pylori infection induces cellular phenotypes relevant for cancer progression, namely cell motility and invasion. We hypothesized that the extracellular matrix (ECM) could be involved in these deleterious effects. METHODS: Microarrays were used to uncover ECM interactors in cells infected with H. pylori. LAMC2, encoding laminin γ2, was selected as a candidate gene and its expression was assessed in vitro and in vivo. The role of LAMC2 was investigated by small interference RNA (siRNA) combined with a set of functional assays. Laminin γ2 and E-cadherin expression patterns were evaluated in gastric cancer cases. RESULTS: Laminin γ2 was found significantly overexpressed in gastric cancer cells infected with H. pylori. This finding was validated in vitro by infection with clinical isolates and in vivo by using gastric biopsies of infected and noninfected individuals. We showed that laminin γ2 overexpression is dependent on the bacterial type IV secretion system and on the CagA. Functionally, laminin γ2 promotes cell invasion and resistance to apoptosis, through modulation of Src, JNK, and AKT activity. These effects were abrogated in cells with functional E-cadherin. CONCLUSIONS: These data highlight laminin γ2 and its downstream effectors as potential therapeutic targets, and the value of H. pylori eradication to delay gastric cancer onset and progression.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Neoplasias Gástricas , Humanos , Helicobacter pylori/genética , Laminina/metabolismo , Infecciones por Helicobacter/microbiología , Línea Celular Tumoral , Cadherinas/metabolismo , Antígenos Bacterianos/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
5.
Crit Rev Microbiol ; 48(3): 376-396, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-34569892

RESUMEN

Helicobacter pylori is the major component of the gastric microbiome of infected individuals and one of the aetiological factors of chronic gastritis, peptic ulcer disease and gastric cancer. The increasing resistance to antibiotics worldwide has made the treatment of H. pylori infection a challenge. As a way to overhaul the efficacy of currently used H. pylori antibiotic-based eradication therapies, alternative treatment strategies are being devised. These include probiotics and prebiotics as adjuvants in H. pylori treatment, antimicrobial peptides as alternatives to antibiotics, photodynamic therapy ingestible devices, microparticles and nanoparticles applied as drug delivery systems, vaccines, natural products, and phage therapy. This review provides an updated synopsis of these emerging H. pylori control strategies and discusses the advantages, hurdles, and challenges associated with their development and implementation. An effective human vaccine would be a major achievement although, until now, projects regarding vaccine development have failed or were discontinued. Numerous natural products have demonstrated anti-H. pylori activity, mostly in vitro, but further clinical studies are needed to fully disclose their role in H. pylori eradication. Finally, phage therapy has the potential to emerge as a valid alternative, but major challenges remain, namely the isolation of more H. pylori strictly virulent bacterio(phages).


Asunto(s)
Productos Biológicos , Infecciones por Helicobacter , Helicobacter pylori , Probióticos , Antibacterianos/farmacología , Productos Biológicos/farmacología , Infecciones por Helicobacter/tratamiento farmacológico , Humanos , Probióticos/uso terapéutico
6.
BMC Cancer ; 22(1): 646, 2022 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-35692051

RESUMEN

BACKGROUND: Gastric Cancer (GC) is the fourth most deadly cancer worldwide. Enhanced understanding of its key epidemiological and molecular drivers is urgently needed to lower the incidence and improve outcomes. Furthermore, tumor biology in European (EU) and Latin American (LATAM) countries is understudied. The LEGACy study is a Horizon 2020 funded multi-institutional research approach to 1) detail the epidemiological features including risk factors of GC in current time and 2) develop cost-effective methods to identify and integrate biological biomarkers needed to guide diagnostic and therapeutic approaches with the aim of filling the knowledge gap on GC in these areas. METHODS: This observational study has three parts that are conducted in parallel during 2019-2023 across recruiting centers from four EU and four LATAM countries: Part 1) A case-control study (800 cases and 800 controls) using questionnaires on candidate risk factors for GC, which will be correlated with clinical, demographic and epidemiological parameters. Part 2) A case-control tissue sampling study (400 cases and 400 controls) using proteome, genome, microbiome and immune analyses to characterize advanced (stage III and IV) GC. Patients in this part of the study will be followed over time to observe clinical outcomes. The first half of samples will be used as training cohort to identify the most relevant risk factors and biomarkers, which will be selected to propose cost-effective diagnostic and predictive methods that will be validated with the second half of samples. Part 3) An educational study, as part of our prevention strategy (subjects recruited from the general public) to test and disseminate knowledge on GC risk factors and symptoms by a questionnaire and informative video. Patients could be recruited for more than one of the three LEGACy studies. DISCUSSION: The LEGACy study aims to generate novel, in-depth knowledge on the tumor biological characteristics through integrating epidemiological, multi-omics and clinical data from GC patients at an EU-LATAM partnership. During the study, cost-effective panels with potential use in clinical decision making will be developed and validated. TRIAL REGISTRATION: ClinicalTrials.gov Identifiers: Part 1: NCT03957031 . Part 2: NCT04015466 . Part 3: NCT04019808 .


Asunto(s)
Neoplasias Gástricas , Estudios de Casos y Controles , Toma de Decisiones Clínicas , Humanos , América Latina/epidemiología , Fenotipo , Factores de Riesgo , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/epidemiología , Neoplasias Gástricas/genética
7.
Gastric Cancer ; 25(1): 124-137, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486077

RESUMEN

BACKGROUND: Tumour progression relies on the ability of cancer cells to penetrate and invade neighbouring tissues. E-cadherin loss is associated with increased cell invasion in gastric carcinoma, and germline mutations of the E-cadherin gene are causative of hereditary diffuse gastric cancer. Although E-cadherin dysfunction impacts cell-cell adhesion, cell dissemination also requires an imbalance of adhesion to the extracellular matrix (ECM). METHODS: To identify ECM components and receptors relevant for adhesion of E-cadherin dysfunctional cells, we implemented a novel ECM microarray platform coupled with molecular interaction networks. The functional role of putative candidates was determined by combining micropattern traction microscopy, protein modulation and in vivo approaches, as well as transcriptomic data of 262 gastric carcinoma samples, retrieved from the cancer genome atlas (TCGA). RESULTS: Here, we show that E-cadherin mutations induce an abnormal interplay of cells with specific components of the ECM, which encompasses increased traction forces and Integrin ß1 activation. Integrin ß1 synergizes with E-cadherin dysfunction, promoting cell scattering and invasion. The significance of the E-cadherin-Integrin ß1 crosstalk was validated in Drosophila models and found to be consistent with evidence from human gastric carcinomas, where increased tumour grade and poor survival are associated with low E-cadherin and high Integrin ß1 levels. CONCLUSIONS: Integrin ß1 is a key mediator of invasion in carcinomas with E-cadherin impairment and should be regarded as a biomarker of poor prognosis in gastric cancer.


Asunto(s)
Integrina beta1 , Neoplasias Gástricas , Animales , Cadherinas/genética , Cadherinas/metabolismo , Adhesión Celular/fisiología , Drosophila melanogaster , Matriz Extracelular/metabolismo , Humanos , Integrina beta1/genética , Integrina beta1/metabolismo , Invasividad Neoplásica , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo
8.
Int J Mol Sci ; 23(14)2022 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-35887231

RESUMEN

Helicobacter pylori, a significant human gastric pathogen, has been demonstrating increased antibiotic resistance, causing difficulties in infection treatment. It is therefore important to develop alternatives or complementary approaches to antibiotics to tackle H. pylori infections, and (bacterio)phages have proven to be effective antibacterial agents. In this work, prophage isolation was attempted using H. pylori strains and UV radiation. One phage was isolated and further characterized to assess potential phage-inspired therapeutic alternatives to H. pylori infections. HPy1R is a new podovirus prophage with a genome length of 31,162 bp, 37.1% GC, encoding 36 predicted proteins, of which 17 were identified as structural. Phage particles remained stable at 37 °C, from pH 3 to 11, for 24 h in standard assays. Moreover, when submitted to an in vitro gastric digestion model, only a small decrease was observed in the gastric phase, suggesting that it is adapted to the gastric tract environment. Together with its other characteristics, its capability to suppress H. pylori population levels for up to 24 h post-infection at multiplicities of infection of 0.01, 0.1, and 1 suggests that this newly isolated phage is a potential candidate for phage therapy in the absence of strictly lytic phages.


Asunto(s)
Bacteriófagos , Infecciones por Helicobacter , Helicobacter pylori , Antibacterianos , Bacteriófagos/genética , Genómica , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/terapia , Humanos , Profagos/genética
9.
Int J Mol Sci ; 21(3)2020 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-32046034

RESUMEN

Large numbers of well-characterized clinical samples are fundamental to establish relevant associations between the microbiota and disease. Formalin-fixed and paraffin-embedded (FFPE) tissues are routinely used and are widely available clinical materials. Since current approaches to study the microbiota are based on next-generation sequencing (NGS) targeting the bacterial 16S rRNA gene, our aim was to evaluate the feasibility of FFPE gastric tissues for NGS-based microbiota characterization. Analysis of sequencing data revealed the presence of bacteria in the paraffin control. After the subtraction of the operational taxonomic units (OTUs) present in the paraffin control to the FFPE tissue sample dataset, we evaluated the microbiota profiles between paired FFPE and frozen gastric tissues, and between different times of archiving. Compared with frozen gastric tissues, we detected a lower number of OTUs in the microbiota of paired FFPE tissues, regardless of the time of archiving. No major differences in microbial diversity were identified, but taxonomic variation in the relative abundance of phyla and orders was observed between the two preservation methods. This variation was also evident in each case and throughout the times of FFPE archiving. The use of FFPE tissues for NGS-based microbiota characterization should be considered carefully, as biases can be introduced by the embedding process and the time of tissue archiving.


Asunto(s)
Código de Barras del ADN Taxonómico/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Microbiota , Adhesión en Parafina/métodos , Estómago/microbiología , Fijación del Tejido/métodos , Fijadores/efectos adversos , Formaldehído/efectos adversos , Genoma Bacteriano , Humanos , ARN Ribosómico 16S/genética , Estómago/citología
10.
Helicobacter ; 24 Suppl 1: e12642, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31486241

RESUMEN

Gastric cancer is the third deadliest cancer in the world, and the absolute number of cases is increasing every year due to aging and growing of high-risk populations. This disease is a consequence of the complex interaction of microbial agents, with environmental and host factors, resulting in the dysregulation of multiple oncogenic and tumor-suppressing signaling pathways. Despite the advances in our understanding of carcinogenesis, there are still reduced therapeutic options for patients with gastric cancer. In recent years, genomic analyses of gastric tumors have emphasized their molecular heterogeneity. The distinction of gastric cancer molecular subtypes may be a key to identify novel therapeutic targets, to predict patient outcome and response to therapy, and to guide early diagnosis strategies. In this review, we summarize the most recent updates on the relationship between microbial agents and gastric cancer, in particular, Helicobacter pylori, the non-H pylori microbiome, and Epstein-Barr virus. We also highlight the main advances made in the past year regarding the molecular characterization of gastric cancer, especially the signatures with potential clinical utility.


Asunto(s)
Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Helicobacter/complicaciones , Interacciones Huésped-Patógeno , Neoplasias Gástricas/epidemiología , Neoplasias Gástricas/patología , Investigación Biomédica/tendencias , Humanos , Neoplasias Gástricas/etiología , Neoplasias Gástricas/fisiopatología
11.
Adv Exp Med Biol ; 1149: 195-210, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31016631

RESUMEN

After a long period during which the stomach was considered as an organ where microorganisms could not thrive, Helicobacter pylori was isolated in vitro from gastric biopsies, revolutionising the fields of Microbiology and Gastroenterology. Since then, and with the introduction of high-throughput sequencing technologies that allowed deep characterization of microbial communities, a growing body of knowledge has shown that the stomach contains a diverse microbial community, which is different from that of the oral cavity and of the intestine. Gastric cancer is a heterogeneous disease that is the end result of a cascade of events arising in a small fraction of patients colonized with H. pylori. In addition to H. pylori infection and to multiple host and environmental factors that influence disease development, alterations to the composition and function of the normal gastric microbiome, also known as dysbiosis, may also contribute to malignancy. Chronic inflammation of the mucosa in response to H. pylori may alter the gastric environment, paving the way to the growth of a dysbiotic gastric bacterial community. This dysbiotic microbiome may promote the development of gastric cancer by sustaining inflammation and/or inducing genotoxicity. This chapter summarizes what is known about the gastric microbiome in the context of H. pylori-associated gastric cancer, introducing the emerging dimension of the microbiome into the pathogenesis of this highly incident and deadly disease.


Asunto(s)
Microbioma Gastrointestinal , Infecciones por Helicobacter , Helicobacter pylori , Disbiosis , Mucosa Gástrica/microbiología , Microbioma Gastrointestinal/fisiología , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/microbiología , Humanos
12.
Gut ; 67(2): 226-236, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29102920

RESUMEN

OBJECTIVE: Gastric carcinoma development is triggered by Helicobacter pylori. Chronic H. pylori infection leads to reduced acid secretion, which may allow the growth of a different gastric bacterial community. This change in the microbiome may increase aggression to the gastric mucosa and contribute to malignancy. Our aim was to evaluate the composition of the gastric microbiota in chronic gastritis and in gastric carcinoma. DESIGN: The gastric microbiota was retrospectively investigated in 54 patients with gastric carcinoma and 81 patients with chronic gastritis by 16S rRNA gene profiling, using next-generation sequencing. Differences in microbial composition of the two patient groups were assessed using linear discriminant analysis effect size. Associations between the most relevant taxa and clinical diagnosis were validated by real-time quantitative PCR. Predictive functional profiling of microbial communities was obtained with PICRUSt. RESULTS: The gastric carcinoma microbiota was characterised by reduced microbial diversity, by decreased abundance of Helicobacter and by the enrichment of other bacterial genera, mostly represented by intestinal commensals. The combination of these taxa into a microbial dysbiosis index revealed that dysbiosis has excellent capacity to discriminate between gastritis and gastric carcinoma. Analysis of the functional features of the microbiota was compatible with the presence of a nitrosating microbial community in carcinoma. The major observations were confirmed in validation cohorts from different geographic origins. CONCLUSIONS: Detailed analysis of the gastric microbiota revealed for the first time that patients with gastric carcinoma exhibit a dysbiotic microbial community with genotoxic potential, which is distinct from that of patients with chronic gastritis.


Asunto(s)
Bacterias , Carcinoma/microbiología , Disbiosis/microbiología , Gastritis/microbiología , Microbioma Gastrointestinal , Infecciones por Helicobacter/microbiología , Neoplasias Gástricas/microbiología , Estómago/microbiología , Adulto , Anciano , Bacterias/genética , Bacterias/metabolismo , Enfermedad Crónica , Femenino , Mucosa Gástrica/microbiología , Helicobacter pylori , Humanos , Masculino , Persona de Mediana Edad , Nitrosación , ARN Ribosómico 16S/análisis , Estudios Retrospectivos
13.
BMC Mol Biol ; 19(1): 6, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29879907

RESUMEN

BACKGROUND: Gastric cancer is the third leading cause of cancer-related mortality worldwide. Recently, it has been demonstrated that gastric cancer cells display a specific miRNA expression profile, with increasing evidence of the role of miRNA-9 in this disease. miRNA-9 upregulation has been shown to influence the expression of E-cadherin-encoding gene, triggering cell motility and invasiveness. RESULTS: In this study, we designed LNA anti-miRNA oligonucleotides with a complementary sequence to miRNA-9 and tested their properties to both detect and silence the target miRNA. We could identify and visualize the in vitro uptake of low-dosing LNA-based anti-miRNA oligonucleotides without any carrier or transfection agent, as early as 2 h after the addition of the oligonucleotide sequence to the culture medium. Furthermore, we were able to assess the silencing potential of miRNA-9, using different LNA anti-miRNA oligonucleotide designs, and to observe its subsequent effect on E-cadherin expression. CONCLUSIONS: The administration of anti-miRNA sequences even at low-doses, rapidly repressed the target miRNA, and influenced the expression of E-cadherin by significantly increasing its levels.


Asunto(s)
Cadherinas/genética , MicroARNs/antagonistas & inhibidores , Oligonucleótidos/farmacología , Neoplasias Gástricas/genética , Antígenos CD , Cadherinas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo
14.
Curr Top Microbiol Immunol ; 400: 277-304, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28124158

RESUMEN

Gastric cancer is the fifth most incident and the third most common cause of cancer-related death in the world. Infection with Helicobacter pylori is the major risk factor for this disease. Gastric cancer is the final outcome of a cascade of events that takes decades to occur and results from the accumulation of multiple genetic and epigenetic alterations. These changes are crucial for tumor cells to expedite and sustain the array of pathways involved in the cancer development, such as cell cycle, DNA repair, metabolism, cell-to-cell and cell-to-matrix interactions, apoptosis, angiogenesis, and immune surveillance. Comprehensive molecular analyses of gastric cancer have disclosed the complex heterogeneity of this disease. In particular, these analyses have confirmed that Epstein-Barr virus (EBV)-positive gastric cancer is a distinct entity. The identification of gastric cancer subtypes characterized by recognizable molecular profiles may pave the way for a more personalized clinical management and to the identification of novel therapeutic targets and biomarkers for screening, prognosis, prediction of response to treatment, and monitoring of gastric cancer progression.


Asunto(s)
Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/patología , Helicobacter pylori/fisiología , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Progresión de la Enfermedad , Infecciones por Virus de Epstein-Barr/microbiología , Infecciones por Virus de Epstein-Barr/virología , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/virología , Helicobacter pylori/genética , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/aislamiento & purificación , Herpesvirus Humano 4/fisiología , Humanos , Estómago/microbiología , Estómago/patología , Estómago/virología , Neoplasias Gástricas/microbiología , Neoplasias Gástricas/virología
15.
Gastrointest Endosc ; 87(6): 1566-1575, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29454568

RESUMEN

BACKGROUND AND AIMS: The time course for the development of clinically significant hereditary diffuse gastric cancer (HDGC) is unpredictable. Little is known about the progression from preclinical, indolent lesions to widely invasive, aggressive phenotypes. Gastroendoscopy often fails to detect early lesions, and risk-reducing/prophylactic total gastrectomy (PTG) is the only curative approach. We present an HDGC family with early-onset disease in which clinical and histologic findings provided insight into the understanding of different HDGC phenotypes. METHODS: The proband was diagnosed at age 18 years with widely invasive, metastatic DGC. CDH1 genetic testing identified a pathogenic, germline CDH1 variant (c.1901C>T, p.Ala634Val). Thirty family members were tested, and 15 CDH1 carriers were identified. RESULTS: Six family members had PTG, with negative preoperative workup. The proband's 14-year-old sister is the youngest patient, reported to date, to have PTG after negative preoperative biopsy sampling. Intramucosal HDGC foci were detected in all PTG specimens (1-33). In contrast to the "indolent" phenotype of these foci, the aggressive DGC from the proband showed pleomorphic cells, absent E-cadherin expression, increased proliferation (Ki-67 index), and activation of oncogenic events (p53, pSrc and pStat3 overexpression). All family members had Helicobacter pylori gastritis. Cag-A-positive strains were detected in all specimens, except in the proband's sister. CONCLUSIONS: HDGC is a heterogeneous disease regarding clinical behavior, endoscopic findings, histopathologic features, and immunophenotypic/molecular profile. The presence of bizarre, pleomorphic cells in endoscopic biopsy specimens is suggestive of advanced disease and should prompt clinical intervention. The involvement of a full multidisciplinary team is essential for the management of these patients.


Asunto(s)
Neoplasias de la Mama/patología , Carcinoma Lobular/patología , Síndromes Neoplásicos Hereditarios/patología , Neoplasias Gástricas/patología , Adolescente , Adulto , Edad de Inicio , Anciano de 80 o más Años , Antígenos CD , Neoplasias de la Mama/genética , Cadherinas/genética , Carcinoma Lobular/genética , Familia , Femenino , Gastrectomía , Gastritis/complicaciones , Gastritis/microbiología , Gastroscopía , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Infecciones por Helicobacter/complicaciones , Helicobacter pylori , Humanos , Antígeno Ki-67/metabolismo , Masculino , Persona de Mediana Edad , Mutación Missense , Síndromes Neoplásicos Hereditarios/complicaciones , Síndromes Neoplásicos Hereditarios/genética , Síndromes Neoplásicos Hereditarios/prevención & control , Linaje , Fenotipo , Procedimientos Quirúrgicos Profilácticos , Neoplasias Gástricas/complicaciones , Neoplasias Gástricas/genética , Neoplasias Gástricas/prevención & control , Proteína p53 Supresora de Tumor/metabolismo , Adulto Joven
16.
RNA Biol ; 15(3): 338-352, 2018 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-29570036

RESUMEN

MicroRNAs (miRNAs) are small, non-coding RNA molecules that regulate gene expression post-transcriptionally. As a consequence of their function towards mRNA, miRNAs are widely associated with the pathogenesis of several human diseases, making miRNAs a target for new therapeutic strategies based on the control of their expression. Indeed, numerous works were published in the past decades showing the potential use of antisense oligonucleotides to target aberrant miRNAs (AMOs) involved in several human pathologies. New classes of chemical-modified-AMOs, including locked nucleic acid oligonucleotides, have recently proved their worth in silencing miRNAs. A correct design of a specific AMOs can help to improve their performance and potency towards the target miRNA by increasing for instance nuclease resistance and target affinity. This review outlines the technologies involved to suppress aberrant miRNAs. From the design strategies used in AMOs to its application in novel miRNA-based therapeutics and detection methodologies.


Asunto(s)
Antineoplásicos/química , MicroARNs/antagonistas & inhibidores , Neoplasias/genética , Oligonucleótidos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ensayos Clínicos como Asunto , Diseño de Fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Oligonucleótidos/farmacología , Oligonucleótidos/uso terapéutico
17.
Helicobacter ; 22 Suppl 12017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28891129

RESUMEN

Gastric cancer is one of the most incident and deadliest malignancies in the world. Gastric cancer is a heterogeneous disease and the end point of a long and multistep process, which results from the stepwise accumulation of numerous (epi)genetic alterations, leading to dysregulation of oncogenic and tumor suppressor pathways. Gastric cancer stem cells have emerged as fundamental players in cancer development and as contributors to gastric cancer heterogeneity. For this special issue, we will report last year's update on the gastric cancer molecular classification, and in particular address the gastric cancer groups who could benefit from immune checkpoint therapy. We will also review the latest advances on gastric cancer stem cells, their properties as gastric cancer markers and therapeutic targets, and associated signaling pathways. The understanding of the molecular basis underlying gastric cancer heterogeneity and of the role played by gastric cancer stem cells in cancer development and heterogeneity is of major significance, not only for identifying novel targets for cancer prevention and treatment, but also for clinical management and patient stratification for targeted therapies.


Asunto(s)
Carcinogénesis , Células Madre Neoplásicas/fisiología , Neoplasias Gástricas/patología , Neoplasias Gástricas/fisiopatología , Proliferación Celular , Humanos , Transducción de Señal
18.
J Infect Dis ; 213(4): 669-73, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26401027

RESUMEN

Heterogeneity at the Helicobacter pylori cagA gene promoter region has been linked to variation in CagA expression and gastric histopathology. Here, we characterized the cagA promoter and expression in 46 H. pylori strains from Portugal. Our results confirm the relationship between cagA promoter region variation and protein expression originally observed in strains from Colombia. We observed that individuals with intestinal metaplasia were all infected with H. pylori strains containing a specific cagA motif. Additionally, we provided novel functional evidence that strain-specific sequences in the cagA promoter region and CagA expression levels influence interleukin 8 secretion by the host gastric epithelial cells.


Asunto(s)
Antígenos Bacterianos/genética , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Expresión Génica , Helicobacter pylori/genética , Helicobacter pylori/inmunología , Interleucina-8/metabolismo , Regiones Promotoras Genéticas , Adulto , Anciano , Estudios de Casos y Controles , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Femenino , Variación Genética , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/aislamiento & purificación , Humanos , Masculino , Persona de Mediana Edad , Portugal , Adulto Joven
19.
J Infect Dis ; 213(11): 1767-76, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-26802142

RESUMEN

Helicobacter pylori colonizes the human stomach and increases the risk for peptic ulcer disease and gastric carcinoma. H. pylori upregulates the expression and activity of several matrix metalloproteinases (MMPs) in cell lines and in the gastric mucosa. The aim of this study was to explore the mechanisms leading to upregulation of MMP10 in gastric epithelial cells induced by H. pylori Infection of gastric cells with H. pylori led to an increase in levels of MMP-10 messenger RNA, protein secretion, and activity. cagA knockout mutants or CagA phosphorylation-defective mutants failed to increase MMP10 expression. These results were confirmed in infection experiments with clinical isolates with known cagA status and in human gastric biopsy specimens. Treatment of cells with chemical inhibitors of the receptor tyrosine kinase EGFR and the kinase Src abrogated H. pylori-induced MMP10 expression. Inhibitors of ERK1/2 and JNK kinases abolished and significantly decreased H. pylori-induced MMP10 expression, respectively, whereas inhibition of the kinase p38 had no effect. Finally, inhibition of MMP10 expression by small interfering RNA led to a decrease in the gastric cell-invasive phenotype mediated by the infection. In conclusion, CagA-positive H. pylori strains stimulate MMP10 expression. MMP-10 modulation occurs via EGFR activation in a process that involves Src, ERK, and JNK pathways. MMP-10 may be implicated in H. pylori-mediated extracellular matrix remodeling.


Asunto(s)
Receptores ErbB/metabolismo , Mucosa Gástrica/enzimología , Helicobacter pylori/patogenicidad , Sistema de Señalización de MAP Quinasas , Metaloproteinasa 10 de la Matriz/metabolismo , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Línea Celular , Línea Celular Tumoral , Activación Enzimática , Femenino , Mucosa Gástrica/microbiología , Humanos , MAP Quinasa Quinasa 4/metabolismo , Masculino , Persona de Mediana Edad , Factores de Virulencia/metabolismo
20.
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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA