Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Allergy Clin Immunol ; 142(1): 86-95, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29452199

RESUMEN

BACKGROUND: Dynamic establishment of the nasal microbiota in early life influences local mucosal immune responses and susceptibility to childhood respiratory disorders. OBJECTIVE: The aim of this case-control study was to monitor, evaluate, and compare development of the nasal microbiota of infants with rhinitis and wheeze in the first 18 months of life with those of healthy control subjects. METHODS: Anterior nasal swabs of 122 subjects belonging to the Growing Up in Singapore Towards Healthy Outcomes (GUSTO) birth cohort were collected longitudinally over 7 time points in the first 18 months of life. Nasal microbiota signatures were analyzed by using 16S rRNA multiplexed pair-end sequencing from 3 clinical groups: (1) patients with rhinitis alone (n = 28), (2) patients with rhinitis with concomitant wheeze (n = 34), and (3) healthy control subjects (n = 60). RESULTS: Maturation of the nasal microbiome followed distinctive patterns in infants from both rhinitis groups compared with control subjects. Bacterial diversity increased over the period of 18 months of life in control infants, whereas infants with rhinitis showed a decreasing trend (P < .05). An increase in abundance of the Oxalobacteraceae family (Proteobacteria phylum) and Aerococcaceae family (Firmicutes phylum) was associated with rhinitis and concomitant wheeze (adjusted P < .01), whereas the Corynebacteriaceae family (Actinobacteria phylum) and early colonization with the Staphylococcaceae family (Firmicutes phylum; 3 weeks until 9 months) were associated with control subjects (adjusted P < .05). The only difference between the rhinitis and control groups was a reduced abundance of the Corynebacteriaceae family (adjusted P < .05). Determinants of nasal microbiota succession included sex, mode of delivery, presence of siblings, and infant care attendance. CONCLUSION: Our results support the hypothesis that the nasal microbiome is involved in development of early-onset rhinitis and wheeze in infants.


Asunto(s)
Microbiota , Mucosa Nasal/microbiología , Ruidos Respiratorios , Rinitis/microbiología , Estudios de Casos y Controles , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Mucosa Nasal/inmunología , Ruidos Respiratorios/inmunología , Rinitis/inmunología , Singapur
2.
Nat Methods ; 12(5): 458-64, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25799442

RESUMEN

Most disease associations detected by genome-wide association studies (GWAS) lie outside coding genes, but very few have been mapped to causal regulatory variants. Here, we present a method for detecting regulatory quantitative trait loci (QTLs) that does not require genotyping or whole-genome sequencing. The method combines deep, long-read chromatin immunoprecipitation-sequencing (ChIP-seq) with a statistical test that simultaneously scores peak height correlation and allelic imbalance: the genotype-independent signal correlation and imbalance (G-SCI) test. We performed histone acetylation ChIP-seq on 57 human lymphoblastoid cell lines and used the resulting reads to call 500,066 single-nucleotide polymorphisms de novo within regulatory elements. The G-SCI test annotated 8,764 of these as histone acetylation QTLs (haQTLs)­an order of magnitude larger than the set of candidates detected by expression QTL analysis. Lymphoblastoid haQTLs were highly predictive of autoimmune disease mechanisms. Thus, our method facilitates large-scale regulatory variant detection in any moderately sized cohort for which functional profiling data can be generated, thereby simplifying identification of causal variants within GWAS loci.


Asunto(s)
Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo , Acetilación , Alelos , Línea Celular , Regulación de la Expresión Génica , Marcadores Genéticos , Genotipo , Histonas/metabolismo , Humanos , Transcriptoma
3.
Gastroenterology ; 150(2): 355-7.e3, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26551551

RESUMEN

There have been increasing reports of food-borne zoonotic transmission of hepatitis E virus (HEV) genotype 3, which causes chronic infections in immunosuppressed patients. We performed phylogenetic analyses of the HEV sequence (partial and full-length) from 1 patient from the Middle East who underwent liver transplantation, and compared it with other orthohepevirus A sequences. We found the patient to be infected by camelid HEV. This patient regularly consumed camel meat and milk, therefore camelid HEV, which is genotype 7, might infect human beings. Our finding links consumption of camel-derived food products to post-transplantation hepatitis E, which, if detected at early stages, can be cured with antiviral therapy and reduced administration of immunosuppressive agents.


Asunto(s)
Camelus/virología , Contaminación de Alimentos , Virus de la Hepatitis E/patogenicidad , Hepatitis E/virología , Hepatitis Crónica/virología , Trasplante de Hígado/efectos adversos , Carne/virología , Leche/virología , Zoonosis , Animales , Antivirales/uso terapéutico , Genotipo , Hepatitis E/diagnóstico , Hepatitis E/tratamiento farmacológico , Hepatitis E/transmisión , Virus de la Hepatitis E/genética , Hepatitis Crónica/diagnóstico , Hepatitis Crónica/tratamiento farmacológico , Humanos , Huésped Inmunocomprometido , Inmunosupresores/efectos adversos , Masculino , Persona de Mediana Edad , Filogenia , Factores de Tiempo , Resultado del Tratamiento
4.
Infect Immun ; 84(9): 2505-23, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27324481

RESUMEN

Increasing experimental evidence supports the idea that Mycobacterium tuberculosis has evolved strategies to survive within lysosomes of activated macrophages. To further our knowledge of M. tuberculosis response to the hostile lysosomal environment, we profiled the global transcriptional activity of M. tuberculosis when exposed to the lysosomal soluble fraction (SF) prepared from activated macrophages. Transcriptome sequencing (RNA-seq) analysis was performed using various incubation conditions, ranging from noninhibitory to cidal based on the mycobacterial replication or killing profile. Under inhibitory conditions that led to the absence of apparent mycobacterial replication, M. tuberculosis expressed a unique transcriptome with modulation of genes involved in general stress response, metabolic reprogramming, respiration, oxidative stress, dormancy response, and virulence. The transcription pattern also indicates characteristic cell wall remodeling with the possible outcomes of increased infectivity, intrinsic resistance to antibiotics, and subversion of the host immune system. Among the lysosome-specific responses, we identified the glgE-mediated 1,4 α-glucan synthesis pathway and a defined group of VapBC toxin/anti-toxin systems, both of which represent toxicity mechanisms that potentially can be exploited for killing intracellular mycobacteria. A meta-analysis including previously reported transcriptomic studies in macrophage infection and in vitro stress models was conducted to identify overlapping and nonoverlapping pathways. Finally, the Tap efflux pump-encoding gene Rv1258c was selected for validation. An M. tuberculosis ΔRv1258c mutant was constructed and displayed increased susceptibility to killing by lysosomal SF and the antimicrobial peptide LL-37, as well as attenuated survival in primary murine macrophages and human macrophage cell line THP-1.


Asunto(s)
Regulación Bacteriana de la Expresión Génica/genética , Lisosomas/genética , Mycobacterium tuberculosis/genética , Estrés Oxidativo/genética , Transcripción Genética/genética , Animales , Péptidos Catiónicos Antimicrobianos , Catelicidinas/genética , Línea Celular , Interacciones Huésped-Patógeno/genética , Humanos , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Transcriptoma/genética , Tuberculosis/microbiología , Virulencia/genética
5.
Am J Hum Genet ; 93(2): 197-210, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-23810381

RESUMEN

White matter hyperintensities (WMHs) of the brain are important markers of aging and small-vessel disease. WMHs are rare in healthy children and, when observed, often occur with comorbid neuroinflammatory or vasculitic processes. Here, we describe a complex 4 kb deletion in 2q36.3 that segregates with early childhood communication disorders and WMH in 15 unrelated families predominantly from Southeast Asia. The premature brain aging phenotype with punctate and multifocal WMHs was observed in ~70% of young carrier parents who underwent brain MRI. The complex deletion removes the penultimate exon 3 of TM4SF20, a gene encoding a transmembrane protein of unknown function. Minigene analysis showed that the resultant net loss of an exon introduces a premature stop codon, which, in turn, leads to the generation of a stable protein that fails to target to the plasma membrane and accumulates in the cytoplasm. Finally, we report this deletion to be enriched in individuals of Vietnamese Kinh descent, with an allele frequency of about 1%, embedded in an ancestral haplotype. Our data point to a constellation of early language delay and WMH phenotypes, driven by a likely toxic mechanism of TM4SF20 truncation, and highlight the importance of understanding and managing population-specific low-frequency pathogenic alleles.


Asunto(s)
Envejecimiento Prematuro/genética , Secuencia de Bases , Predisposición Genética a la Enfermedad , Trastornos del Desarrollo del Lenguaje/genética , Leucoencefalopatías/genética , Eliminación de Secuencia , Tetraspaninas/genética , Edad de Inicio , Envejecimiento Prematuro/complicaciones , Envejecimiento Prematuro/etnología , Envejecimiento Prematuro/patología , Pueblo Asiatico , Encéfalo/metabolismo , Encéfalo/patología , Niño , Preescolar , Cromosomas Humanos Par 2 , Exones , Femenino , Humanos , Trastornos del Desarrollo del Lenguaje/complicaciones , Trastornos del Desarrollo del Lenguaje/etnología , Trastornos del Desarrollo del Lenguaje/patología , Leucoencefalopatías/complicaciones , Leucoencefalopatías/etnología , Leucoencefalopatías/patología , Imagen por Resonancia Magnética , Masculino , Datos de Secuencia Molecular , Linaje , Análisis de Secuencia de ADN
6.
J Hepatol ; 60(1): 54-61, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23994382

RESUMEN

BACKGROUND & AIMS: The interaction between HBV replication and immune modulatory effects mediated by IFNα therapy is not well understood. We characterized the impact of HBV DNA replication on the early IFNα-induced immunomodulatory mechanisms. METHODS: We interrogated the transcriptional, serum cytokine/chemokine and cellular immune profiles of 28 patients with HBeAg+ chronic HBV infection (CHB) randomly assigned to one of 4 treatment cohorts (untreated n=5, weekly dosing of 360 µg Pegasys [PegIFNα] n=11, daily dose of 300 mg Viread [tenofovir disoproxil fumarate, TDF] n=6, or a combination of both n=6). Samples were characterized at multiple early time points through day 14 of therapy, after which all patients were given standard of care (180 µg Pegasys injected subcutaneously, weekly). RESULTS: PegIFNα induced a distinct and rapid up-regulation of IFN signaling pathway that coincided with increase detection of distinct serum cytokines/chemokines (IL-15, IL-6, and CXCL-10) and the up-regulation of the frequency of proliferating NK and activated total CD8+ T cells. IFNα treatment alone did not result in rapid decay of HBV replication and was not able to restore the defective HBV-specific T cell response present in CHB patients. In addition, the IFNα immune-stimulatory effects diminished after the first dose, but this refractory effect was reduced in patients where HBV replication was simultaneously inhibited with TDF. CONCLUSIONS: We present here the first comprehensive description of the early effects of IFNα treatment on immune and viral biomarkers in HBeAg+ CHB patients. Our results show that PegIFNα-induced innate immune activation directly benefits from the suppression of HBV replication.


Asunto(s)
Antivirales/uso terapéutico , Virus de la Hepatitis B/fisiología , Hepatitis B Crónica/tratamiento farmacológico , Interferón-alfa/uso terapéutico , Polietilenglicoles/uso terapéutico , Replicación Viral/efectos de los fármacos , Adolescente , Adulto , Estudios de Cohortes , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/virología , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Recombinantes/uso terapéutico , Linfocitos T/inmunología
7.
Nucleic Acids Res ; 40(22): 11189-201, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23066108

RESUMEN

The study of cell-population heterogeneity in a range of biological systems, from viruses to bacterial isolates to tumor samples, has been transformed by recent advances in sequencing throughput. While the high-coverage afforded can be used, in principle, to identify very rare variants in a population, existing ad hoc approaches frequently fail to distinguish true variants from sequencing errors. We report a method (LoFreq) that models sequencing run-specific error rates to accurately call variants occurring in <0.05% of a population. Using simulated and real datasets (viral, bacterial and human), we show that LoFreq has near-perfect specificity, with significantly improved sensitivity compared with existing methods and can efficiently analyze deep Illumina sequencing datasets without resorting to approximations or heuristics. We also present experimental validation for LoFreq on two different platforms (Fluidigm and Sequenom) and its application to call rare somatic variants from exome sequencing datasets for gastric cancer. Source code and executables for LoFreq are freely available at http://sourceforge.net/projects/lofreq/.


Asunto(s)
Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Simulación por Computador , Virus del Dengue/genética , Escherichia coli/genética , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/normas , Humanos , Mutación , Sensibilidad y Especificidad , Neoplasias Gástricas/genética , Proteínas Virales/química , Proteínas Virales/genética
8.
Sci Immunol ; 4(32)2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30737354

RESUMEN

Associations between chronic antigen stimulation, T cell dysfunction, and the expression of various inhibitory receptors are well characterized in several mouse and human systems. During chronic hepatitis B virus (HBV) infection (CHB), T cell responses are blunted with low frequencies of virus-specific T cells observed, making these parameters difficult to study. Here, using mass cytometry and a highly multiplexed combinatorial peptide-major histocompatibility complex (pMHC) tetramer strategy that allows for the detection of rare antigen-specific T cells, we simultaneously probed 484 unique HLA-A*1101-restricted epitopes spanning the entire HBV genome on T cells from patients at various stages of CHB. Numerous HBV-specific T cell populations were detected, validated, and profiled. T cells specific for two epitopes (HBVpol387 and HBVcore169) displayed differing and complex heterogeneities that were associated with the disease progression, and the expression of inhibitory receptors on these cells was not linearly related with their extent of T cell dysfunction. For HBVcore169-specific CD8+ T cells, we found cellular markers associated with long-term memory, polyfunctionality, and the presence of several previously unidentified public TCR clones that correlated with viral control. Using high-dimensional trajectory analysis of these cellular phenotypes, a pseudo-time metric was constructed that fit with the status of viral infection in corresponding patients. This was validated in a longitudinal cohort of patients undergoing antiviral therapy. Our study uncovers complex relationships of inhibitory receptors between the profiles of antigen-specific T cells and the status of CHB with implications for new strategies of therapeutic intervention.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Progresión de la Enfermedad , Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/patología , Adulto , Anciano , Antivirales/uso terapéutico , Niño , ADN Viral/genética , ADN Viral/aislamiento & purificación , Epítopos de Linfocito T/inmunología , Femenino , Antígenos HLA-A/genética , Antígenos HLA-A/inmunología , Virus de la Hepatitis B/genética , Hepatitis B Crónica/tratamiento farmacológico , Hepatitis B Crónica/virología , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Fenotipo , Receptores de Antígenos de Linfocitos T/genética , Adulto Joven
9.
Sci Rep ; 7: 42170, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28176844

RESUMEN

Previously genome-wide association methods in patients with classic bladder exstrophy (CBE) found association with ISL1, a master control gene expressed in pericloacal mesenchyme. This study sought to further explore the genetics in a larger set of patients following-up on the most promising genomic regions previously reported. Genotypes of 12 markers obtained from 268 CBE patients of Australian, British, German Italian, Spanish and Swedish origin and 1,354 ethnically matched controls and from 92 CBE case-parent trios from North America were analysed. Only marker rs6874700 at the ISL1 locus showed association (p = 2.22 × 10-08). A meta-analysis of rs6874700 of our previous and present study showed a p value of 9.2 × 10-19. Developmental biology models were used to clarify the location of ISL1 activity in the forming urinary tract. Genetic lineage analysis of Isl1-expressing cells by the lineage tracer mouse model showed Isl1-expressing cells in the urinary tract of mouse embryos at E10.5 and distributed in the bladder at E15.5. Expression of isl1 in zebrafish larvae staged 48 hpf was detected in a small region of the developing pronephros. Our study supports ISL1 as a major susceptibility gene for CBE and as a regulator of urinary tract development.


Asunto(s)
Extrofia de la Vejiga/genética , Predisposición Genética a la Enfermedad , Proteínas con Homeodominio LIM/genética , Mesodermo/metabolismo , Organogénesis/genética , Factores de Transcripción/genética , Sistema Urinario/metabolismo , Animales , Extrofia de la Vejiga/metabolismo , Extrofia de la Vejiga/patología , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas con Homeodominio LIM/metabolismo , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Mesodermo/anomalías , Mesodermo/crecimiento & desarrollo , Ratones , Polimorfismo de Nucleótido Simple , Pronefro/crecimiento & desarrollo , Pronefro/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Factores de Transcripción/metabolismo , Sistema Urinario/anomalías , Sistema Urinario/crecimiento & desarrollo , Pez Cebra
10.
PLoS One ; 9(5): e97523, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24830946

RESUMEN

Colonizing commensal bacteria after birth are required for the proper development of the gastrointestinal tract. It is believed that bacterial colonization pattern in neonatal gut affects gut barrier function and immune system maturation. Studies on the development of faecal microbiota in infants showed that the neonatal gut was first colonized with enterococci followed by other microbiota such as Bifidobacterium. Other studies showed that babies who developed allergy were less often colonized with Enterococcus during the first month of life as compared to healthy infants. Many studies have been conducted to elucidate how bifidobacteria or lactobacilli, some of which are considered probiotic, regulate infant gut immunity. However, fewer studies have been focused on enterococi. In our study, we demonstrate that E. faecalis, isolated from healthy newborns, suppress inflammatory responses activated in vivo and in vitro. We found E. faecalis attenuates proinflammatory cytokine secretions, especially IL-8, through JNK and p38 signaling pathways. This finding shed light on how the first colonizer, E.faecalis, regulates inflammatory responses in the host.


Asunto(s)
Enterococcus faecalis/metabolismo , Inflamación/metabolismo , Inflamación/microbiología , Sistema de Señalización de MAP Quinasas , Animales , Células CACO-2 , Medios de Cultivo Condicionados/química , Enterococcus faecalis/genética , Células HCT116 , Células HT29 , Humanos , Indonesia , Lactante , Recién Nacido , Interleucina-1beta/metabolismo , Interleucina-8/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Lactobacillus/genética , Lactobacillus/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Filogenia , Reacción en Cadena de la Polimerasa , ARN Ribosómico 16S/genética , Salmonella typhimurium/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
11.
Methods Mol Biol ; 1138: 175-95, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24696338

RESUMEN

RNA viruses are notorious for their ability to quickly adapt to selective pressure from the host immune system and/or antivirals. This adaptability is likely due to the error-prone characteristics of their RNA-dependent, RNA polymerase [1, 2]. Dengue virus, a member of the Flaviviridae family of positive-strand RNA viruses, is also known to share these error-prone characteristics [3]. Utilizing high-throughput, massively parallel sequencing methodologies, or next-generation sequencing (NGS), we can now accurately quantify these populations of viruses and track the changes to these populations over the course of a single infection. The aim of this chapter is twofold: to describe the methodologies required for sample preparation prior to sequencing and to describe the bioinformatics analyses required for the resulting data.


Asunto(s)
Virus del Dengue/genética , Genoma Viral/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Emparejamiento Base/genética , Biología Computacional , ADN Complementario/biosíntesis , ADN Viral/genética , ADN Viral/aislamiento & purificación , Reacción en Cadena de la Polimerasa , ARN/metabolismo , ARN Viral/genética , ARN Viral/aislamiento & purificación , Programas Informáticos
12.
PLoS One ; 9(11): e111640, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25365328

RESUMEN

BACKGROUND: The influenza A virus is an RNA virus that is responsible for seasonal epidemics worldwide with up to five million cases of severe illness and 500,000 deaths annually according to the World Health Organization estimates. The factors associated with severe diseases are not well defined, but more severe disease is more often seen among persons aged >65 years, infants, pregnant women, and individuals of any age with underlying health conditions. METHODOLOGY/PRINCIPAL FINDINGS: Using gene expression microarrays, the transcriptomic profiles of influenza-infected patients with severe (N = 11), moderate (N = 40) and mild (N = 83) symptoms were compared with the febrile patients of unknown etiology (N = 73). We found that influenza-infected patients, regardless of their clinical outcomes, had a stronger induction of antiviral and cytokine responses and a stronger attenuation of NK and T cell responses in comparison with those with unknown etiology. More importantly, we found that both interferon and ubiquitination signaling were strongly attenuated in patients with the most severe outcomes in comparison with those with moderate and mild outcomes, suggesting the protective roles of these pathways in disease pathogenesis. CONCLUSION/SIGNIFICANCES: The attenuation of interferon and ubiquitination pathways may associate with the clinical outcomes of influenza patients.


Asunto(s)
Virus de la Influenza A , Gripe Humana/genética , Gripe Humana/metabolismo , Interferones/genética , Interferones/metabolismo , Transducción de Señal , Transcriptoma , Adolescente , Adulto , Anciano , Biología Computacional , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Virus de la Influenza A/inmunología , Gripe Humana/diagnóstico , Gripe Humana/inmunología , Masculino , Persona de Mediana Edad , Pronóstico , Índice de Severidad de la Enfermedad , Ubiquitinación , Adulto Joven
13.
PLoS One ; 8(4): e60811, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23579286

RESUMEN

The high throughput and cost-effectiveness afforded by short-read sequencing technologies, in principle, enable researchers to perform 16S rRNA profiling of complex microbial communities at unprecedented depth and resolution. Existing Illumina sequencing protocols are, however, limited by the fraction of the 16S rRNA gene that is interrogated and therefore limit the resolution and quality of the profiling. To address this, we present the design of a novel protocol for shotgun Illumina sequencing of the bacterial 16S rRNA gene, optimized to amplify more than 90% of sequences in the Greengenes database and with the ability to distinguish nearly twice as many species-level OTUs compared to existing protocols. Using several in silico and experimental datasets, we demonstrate that despite the presence of multiple variable and conserved regions, the resulting shotgun sequences can be used to accurately quantify the constituents of complex microbial communities. The reconstruction of a significant fraction of the 16S rRNA gene also enabled high precision (>90%) in species-level identification thereby opening up potential application of this approach for clinical microbial characterization.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Metagenoma , ARN Ribosómico 16S , Adulto , Bacterias/clasificación , Bacterias/genética , Preescolar , Biología Computacional/métodos , Tracto Gastrointestinal/microbiología , Humanos , Filogenia
14.
BMC Med Genomics ; 6: 41, 2013 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-24119614

RESUMEN

BACKGROUND: The majority of gastric cancer cases are believed to be caused by chronic infection with the bacterium Helicobacter pylori, and atrophic corpus gastritis is a predisposing condition to gastric cancer development. We aimed to increase understanding of the molecular details of atrophy by performing a global transcriptome analysis of stomach tissue. METHODS: Biopsies from patients with different stages of H. pylori infection were taken from both the antrum and corpus mucosa and analyzed on microarrays. The stages included patients without current H. pylori infection, H. pylori-infected without corpus atrophy and patients with current or past H. pylori-infection with corpus-predominant atrophic gastritis. RESULTS: Using clustering and integrated analysis, we found firm evidence for antralization of the corpus mucosa of atrophy patients. This antralization harbored gain of gastrin expression, as well as loss of expression of corpus-related genes, such as genes associated with acid production, energy metabolism and blood clotting. The analyses provided detailed molecular evidence for simultaneous intestinal metaplasia (IM) and spasmolytic polypeptide expressing metaplasia (SPEM) in atrophic corpus tissue. Finally, acidic mammalian chitinase, a chitin-degrading enzyme produced by chief cells, was shown to be strongly down-regulated in corpus atrophy. CONCLUSIONS: Transcriptome analysis revealed several gene groups which are related to development of corpus atrophy, some of which were increased also in H. pylori-infected non-atrophic patients. Furthermore, loss of acidic chitinase expression is a promising marker for corpus atrophy.


Asunto(s)
Quitinasas/genética , Mucosa Gástrica/microbiología , Gastritis Atrófica/enzimología , Gastritis Atrófica/genética , Helicobacter pylori/fisiología , Transcriptoma , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/metabolismo , Vasos Sanguíneos/fisiopatología , Quitinasas/deficiencia , Metabolismo Energético/genética , Femenino , Mucosa Gástrica/irrigación sanguínea , Mucosa Gástrica/metabolismo , Gastritis Atrófica/metabolismo , Gastritis Atrófica/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Transcripción Genética
15.
PLoS One ; 4(9): e6958, 2009 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-19742318

RESUMEN

BACKGROUND: The liver is the central organ for xenobiotic metabolism (XM) and is regulated by nuclear receptors such as CAR and PXR, which control the metabolism of drugs. Here we report that gut microbiota influences liver gene expression and alters xenobiotic metabolism in animals exposed to barbiturates. PRINCIPAL FINDINGS: By comparing hepatic gene expression on microarrays from germfree (GF) and conventionally-raised mice (SPF), we identified a cluster of 112 differentially expressed target genes predominantly connected to xenobiotic metabolism and pathways inhibiting RXR function. These findings were functionally validated by exposing GF and SPF mice to pentobarbital which confirmed that xenobiotic metabolism in GF mice is significantly more efficient (shorter time of anesthesia) when compared to the SPF group. CONCLUSION: Our data demonstrate that gut microbiota modulates hepatic gene expression and function by altering its xenobiotic response to drugs without direct contact with the liver.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Intestinos/microbiología , Hígado/microbiología , Xenobióticos/metabolismo , Animales , Barbitúricos/farmacología , Núcleo Celular/metabolismo , Hígado/metabolismo , Masculino , Ratones , Modelos Biológicos , Familia de Multigenes , Análisis de Secuencia por Matrices de Oligonucleótidos , Pentobarbital/farmacología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda