Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nat Commun ; 14(1): 4342, 2023 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-37468468

RESUMO

Although the role of the Wnt pathway in colon carcinogenesis has been described previously, it has been recently demonstrated that Wnt signaling originates from highly dynamic nano-assemblies at the plasma membrane. However, little is known regarding the role of oncogenic APC in reshaping Wnt nanodomains. This is noteworthy, because oncogenic APC does not act autonomously and requires activation of Wnt effectors upstream of APC to drive aberrant Wnt signaling. Here, we demonstrate the role of oncogenic APC in increasing plasma membrane free cholesterol and rigidity, thereby modulating Wnt signaling hubs. This results in an overactivation of Wnt signaling in the colon. Finally, using the Drosophila sterol auxotroph model, we demonstrate the unique ability of exogenous free cholesterol to disrupt plasma membrane homeostasis and drive Wnt signaling in a wildtype APC background. Collectively, these findings provide a link between oncogenic APC, loss of plasma membrane homeostasis and CRC development.


Assuntos
Via de Sinalização Wnt , beta Catenina , Animais , beta Catenina/genética , beta Catenina/metabolismo , Carcinogênese/genética , Membrana Celular/metabolismo , Colo/metabolismo , Drosophila/metabolismo , Via de Sinalização Wnt/genética
2.
Am J Physiol Gastrointest Liver Physiol ; 321(1): G41-G51, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33949197

RESUMO

Assessing intestinal development and host-microbe interactions in healthy human infants requires noninvasive approaches. We have shown that the transcriptome of exfoliated epithelial cells in feces can differentiate breast-fed and formula-fed infants and term and preterm infants. However, it is not fully understood which regions of the intestine that the exfoliated cells represent. Herein, the transcriptional profiles of exfoliated cells with that of the ileal and colonic mucosa were compared. We hypothesized that exfoliated cells in the distal colon would reflect mucosal signatures of more proximal regions of the gut. Two-day-old piglets (n = 8) were fed formulas for 20 days. Luminal contents and mucosa were collected from ileum (IL), ascending colon (AC), and descending (DC) colon, and mRNA was extracted and sequenced. On average, ∼13,000 genes were mapped in mucosal tissues and ∼10,000 in luminal contents. The intersection of detected genes between three mucosa regions and DC exfoliome indicated an approximately 99% overlap. On average, 49% of the genes in IL, AC, and DC mucosa were present in the AC and DC exfoliome. Genes expressed predominantly in specific anatomic sites (stomach, pancreas, small intestine, colon) were detectable in exfoliated cells. In addition, gene markers for all intestinal epithelial cell types were expressed in the exfoliome representing a diverse array of cell types arising from both the small and large intestine. Genes were mapped to nutrient absorption and transport and immune function. Thus, the exfoliome represents a robust reservoir of information in which to assess intestinal development and responses to dietary interventions.NEW & NOTEWORTHY The transcriptome of exfoliated epithelial cells in stool contain gene signatures from both small and large intestinal mucosa affording a noninvasive approach to assess gut health and function.


Assuntos
Células Epiteliais/metabolismo , Intestino Grosso/metabolismo , Intestino Delgado/metabolismo , Transcriptoma/fisiologia , Animais , Colo/metabolismo , Perfilação da Expressão Gênica/métodos , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Mucosa Intestinal/metabolismo , Suínos
3.
Microorganisms ; 8(12)2020 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-33353204

RESUMO

Gut microbiota and the host exist in a mutualistic relationship, with the functional composition of the microbiota strongly influencing the health and well-being of the host. In addition to the standard differential expression analysis of host genes to assess the complex cross-talk between environment (diet), microbiome, and host intestinal physiology, data-driven integrative approaches are needed to identify potential biomarkers of both host genes and microbial communities that characterize these interactions. Our findings demonstrate that the complementary application of univariate differential gene expression analysis and multivariate approaches such as sparse Canonical Correlation Analysis (sCCA) and sparse Principal Components Analysis (sPCA) can be used to integrate data from both the healthy infant gut microbial community and host transcriptome (exfoliome) using stool derived exfoliated cells shed from the gut. These approaches reveal host genes and microbial functional categories related to the feeding phenotype of the infants. Our findings also confirm that combinatorial noninvasive -omic approaches provide an integrative genomics-based perspective of neonatal host-gut microbiome interactions.

4.
EMBO J ; 39(19): e104319, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32915464

RESUMO

The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that senses xenobiotics, diet, and gut microbial-derived metabolites, is increasingly recognized as a key regulator of intestinal biology. However, its effects on the function of colonic stem and progenitor cells remain largely unexplored. Here, we observed that inducible deletion of AhR in Lgr5+ stem cells increases the percentage of colonic stem cells and enhances organoid initiating capacity and growth of sorted stem and progenitor cells, while AhR activation has the opposite effect. Moreover, intestinal-specific AhR knockout increases basal stem cell and crypt injury-induced cell proliferation and promotes colon tumorigenesis in a preclinical colitis-associated tumor model by upregulating FoxM1 signaling. Mechanistically, AhR transcriptionally suppresses FoxM1 expression. Activation of AhR in human organoids recapitulates phenotypes observed in mice, such as reduction in the percentage of colonic stem cells, promotion of stem cell differentiation, and attenuation of FoxM1 signaling. These findings indicate that the AhR-FoxM1 axis, at least in part, mediates colonic stem/progenitor cell behavior.


Assuntos
Colo/metabolismo , Proteína Forkhead Box M1/metabolismo , Receptores de Hidrocarboneto Arílico/deficiência , Transdução de Sinais , Células-Tronco/metabolismo , Animais , Feminino , Proteína Forkhead Box M1/genética , Técnicas de Inativação de Genes , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Receptores de Hidrocarboneto Arílico/metabolismo
5.
Am J Clin Nutr ; 110(2): 377-390, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31175806

RESUMO

BACKGROUND: Microbial metabolism of lignans from high-fiber plant foods produces bioactive enterolignans, such as enterolactone (ENL) and enterodiol (END). Enterolignan exposure influences cellular pathways important to cancer risk and is associated with reduced colon tumorigenesis in animal models and lower colorectal cancer risk in humans. OBJECTIVES: The aim of this study was to test the effects of a flaxseed lignan supplement (50 mg secoisolariciresinol diglucoside/d) compared with placebo on host gene expression in colon biopsies and exfoliated colonocyte RNA in feces and fecal microbial community composition, and to compare responses in relation to ENL excretion. METHODS: We conducted a 2-period randomized, crossover intervention in 42 healthy men and women (20-45 y). We used RNA-seq to measure differentially expressed (DE) genes in colonic mucosa and fecal exfoliated cells through the use of edgeR and functional analysis with Ingenuity Pathway Analysis. We used 16S ribosomal RNA gene (V1-V3) analysis to characterize the fecal microbiome, and measured END and ENL in 24-h urine samples by gas chromatography-mass spectrometry. RESULTS: We detected 32 DE genes (false discovery rate <0.05) in the exfoliome, but none in the mucosal biopsies, in response to 60 d of lignan supplement compared with placebo. Statistically significant associations were detected between ENL excretion and fecal microbiome measured at baseline and at the end of the intervention periods. Further, we detected DE genes in colonic mucosa and exfoliome between low- and high-ENL excreters. Analysis of biopsy samples indicated that several anti-inflammatory upstream regulators, including transforming growth factor ß and interleukin 10 receptor, were suppressed in low-ENL excreters. Complementary analyses in exfoliated cells also suggested that low-ENL excreters may be predisposed to proinflammatory cellular events due to upregulation of nuclear transcription factor κB and NOS2, and an inhibition of the peroxisome proliferator-activated receptor γ network. CONCLUSIONS: These results suggest that ENL or other activities of the associated gut microbial consortia may modulate response to a dietary lignan intervention. This has important implications for dietary recommendations and chemoprevention strategies. This study was registered at clinicaltrials.gov as NCT01619020.


Assuntos
Fezes/microbiologia , Linho/química , Perfilação da Expressão Gênica , Mucosa Intestinal/efeitos dos fármacos , Lignanas/química , Extratos Vegetais/farmacologia , Adulto , Colo/efeitos dos fármacos , Estudos Cross-Over , Método Duplo-Cego , Feminino , Microbioma Gastrointestinal , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Mucosa Intestinal/metabolismo , Masculino , Extratos Vegetais/química
6.
Sci Rep ; 7(1): 14687, 2017 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-29089621

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most frequently used classes of medications in the world, yet they induce an enteropathy that is associated with high morbidity and mortality. A major limitation to better understanding the pathophysiology and diagnosis of this enteropathy is the difficulty of obtaining information about the primary site of injury, namely the distal small intestine. We investigated the utility of using mRNA from exfoliated cells in stool as a means to surveil the distal small intestine in a murine model of NSAID enteropathy. Specifically, we performed RNA-Seq on exfoliated cells found in feces and compared these data to RNA-Seq from both the small intestinal mucosa and colonic mucosa of healthy control mice or those exhibiting NSAID-induced enteropathy. Global gene expression analysis, data intersection, pathway analysis, and computational approaches including linear discriminant analysis (LDA) and sparse canonical correlation analysis (CCA) were used to assess the inter-relatedness of tissue (invasive) and stool (noninvasive) datasets. These analyses revealed that the exfoliated cell transcriptome closely mirrored the transcriptome of the small intestinal mucosa. Thus, the exfoliome may serve as a non-invasive means of detecting and monitoring NSAID enteropathy (and possibly other gastrointestinal mucosal inflammatory diseases).


Assuntos
Antirreumáticos/efeitos adversos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/genética , Fezes/citologia , Enteropatias/genética , Mucosa Intestinal/fisiologia , Intestino Delgado/fisiologia , Transcriptoma/genética , Animais , Antirreumáticos/uso terapêutico , Biologia Computacional , Modelos Animais de Doenças , Feminino , Humanos , Enteropatias/etiologia , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos
7.
Physiol Genomics ; 48(9): 651-9, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27401218

RESUMO

The strength of associations between various exposures (e.g., diet, tobacco, chemopreventive agents) and colorectal cancer risk may partially depend on the complex interaction between epithelium and stroma across anatomic subsites. Currently, baseline data describing genome-wide coding and long noncoding gene expression profiles in the healthy colon specific to tissue type and location are lacking. Therefore, colonic mucosal biopsies from 10 healthy participants who were enrolled in a clinical study to evaluate effects of lignan supplementation on gut resiliency were used to characterize the site-specific global gene expression signatures associated with stromal vs. epithelial cells in the sigmoid colon and rectum. Using RNA-seq, we demonstrate that tissue type and location patterns of gene expression and upstream regulatory pathways are distinct. For example, consistent with a key role of stroma in the crypt niche, mRNAs associated with immunoregulatory and inflammatory processes (i.e., CXCL14, ANTXR1), smooth muscle contraction (CALD1), proliferation and apoptosis (GLP2R, IGFBP3), and modulation of extracellular matrix (MMP2, COL3A1, MFAP4) were all highly expressed in the stroma. In comparison, HOX genes (HOXA3, HOXD9, HOXD10, HOXD11, and HOXD-AS2, a HOXD cluster antisense RNA 2), and WNT5B expression were also significantly higher in sigmoid colon compared with the rectum. These findings provide strong impetus for considering colorectal tissue subtypes and location in future observational studies and clinical trials designed to evaluate the effects of exposures on colonic health.


Assuntos
Colo Sigmoide/metabolismo , Colo/metabolismo , Células Epiteliais/metabolismo , Reto/metabolismo , Adulto , Biópsia , Colo/efeitos dos fármacos , Colo/patologia , Colo Sigmoide/efeitos dos fármacos , Colo Sigmoide/patologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Estudos Cross-Over , Método Duplo-Cego , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Epitélio/patologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Lignanas/administração & dosagem , Masculino , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Reto/efeitos dos fármacos , Reto/patologia , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética , Adulto Jovem
8.
J Nutr ; 146(6): 1189-96, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27146921

RESUMO

BACKGROUND: Dietary factors such as high-sodium or high-fat (HF) diets have been shown to induce a proinflammatory phenotype. However, there is limited information with respect to how microenvironments of distinct intra-abdominal adipose depots respond to the combination of a high-salt, HF diet. OBJECTIVE: We tested the hypothesis that HF feeding would cause changes in distinct adipose depots, which would be further amplified by the addition of high salt to the diet. METHODS: Twenty-seven male C57BL6 mice were fed an HF diet (60% of kcal from fat), an HF + high-salt diet (4% wt:wt), a control diet [low-fat (LF);10% of kcal from fat], or an LF + high-salt diet for 12 wk. The main sources of fat in the diets were corn oil and lard. Adipokines in serum and released from adipose tissue organ cultures were measured by immunoassays. QIAGEN's Ingenuity Pathway Analysis was used to perform functional analysis of the RNA-sequencing data from distinct adipose depots. RESULTS: Diet-induced obesity resulted in a classical inflammatory phenotype characterized by increased concentrations of circulating inflammatory mediators (38-56%) and reduced adiponectin concentrations (27%). However, high-salt feeding did not exacerbate the HF diet-induced changes in adipokines and cytokines. Leptin and interleukin-6 were differentially released from adipose depots and HF feeding impaired adiponectin and resistin secretion across all 3 depots (34-48% and 45-83%, respectively). The addition of high salt to the HF diet did not further modulate secretion in cultured adipose tissue experiments. Although gene expression data from RNA sequencing indicated a >4.3-fold upregulation of integrin αX (Itgax) with HF feeding in all 3 depots, markers of cellular function were differentially expressed in response to diet across depots. CONCLUSION: Collectively, these findings highlight the role of distinct adipose depots in mice in the development of obesity and emphasize the importance of selecting specific depots to study the effects of therapeutic interventions on adipose tissue function.


Assuntos
Tecido Adiposo/metabolismo , Dieta Hiperlipídica/efeitos adversos , Cloreto de Sódio na Dieta/efeitos adversos , Adiponectina/sangue , Adiponectina/metabolismo , Animais , Antígeno CD11c/genética , Antígeno CD11c/metabolismo , Interleucina-6/sangue , Leptina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Resistina/sangue , Resistina/metabolismo , Análise de Sequência de RNA , Cloreto de Sódio na Dieta/administração & dosagem , Regulação para Cima
9.
Gut Microbes ; 7(3): 246-61, 2016 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-27007819

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) are one of the most frequently used classes of medications in the world. Unfortunately, NSAIDs induce an enteropathy associated with high morbidity and mortality. Although the pathophysiology of this condition involves the interaction of the gut epithelium, microbiota, and NSAIDs, the precise mechanisms by which microbiota influence NSAID enteropathy are unclear. One possible mechanism is that the microbiota may attenuate the severity of disease by specific metabolite-mediated regulation of host inflammation and injury. The microbiota-derived tryptophan-metabolite indole is abundant in the healthy mammalian gut and positively influences intestinal health. We thus examined the effects of indole administration on NSAID enteropathy. Mice (n = 5 per group) were treated once daily for 7 days with an NSAID (indomethacin; 5 mg/kg), indole (20 mg/kg), indomethacin plus indole, or vehicle only (control). Outcomes compared among groups included: microscopic pathology; fecal calprotectin concentration; proportion of neutrophils in the spleen and mesenteric lymph nodes; fecal microbiota composition and diversity; small intestinal mucosal transcriptome; and, fecal tryptophan metabolites. Co-administration of indole with indomethacin: significantly reduced mucosal pathology scores, fecal calprotectin concentrations, and neutrophilic infiltration of the spleen and mesenteric lymph nodes induced by indomethacin; modulated NSAID-induced perturbation of the microbiota, fecal metabolites, and inferred metagenome; and, abrogated a pro-inflammatory gene expression profile in the small intestinal mucosa induced by indomethacin. The microbiota-derived metabolite indole attenuated multiple deleterious effects of NSAID enteropathy, including modulating inflammation mediated by innate immune responses and altering indomethacin-induced shift of the microbiota.


Assuntos
Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios/metabolismo , Enterite/tratamento farmacológico , Fármacos Gastrointestinais/farmacologia , Indóis/metabolismo , Indóis/farmacologia , Inflamação/patologia , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Bactérias/classificação , Bactérias/isolamento & purificação , Biota , Modelos Animais de Doenças , Enterite/induzido quimicamente , Fezes/química , Fezes/microbiologia , Fármacos Gastrointestinais/administração & dosagem , Histocitoquímica , Indóis/administração & dosagem , Complexo Antígeno L1 Leucocitário/análise , Linfonodos/patologia , Camundongos , Neutrófilos/imunologia , Baço/patologia , Resultado do Tratamento
10.
Data Brief ; 6: 398-404, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26862588

RESUMO

With the identification of Lgr5 as a definitive marker for intestinal stem cells, we used the highly novel, recently described, Lgr5-EGFP-IRES-cre ER (T2) knock in mouse model. Mice were injected with azoxymethane (AOM, a colon carcinogen) or saline (control) and fed a chemo-protective diet containing n-3 fatty acids and fermentable fiber (n-3 PUFA+pectin) or a control diet (n-6 PUFA + cellulose). Single cells were isolated from colonic mucosa crypts and three discrete populations of cells were collected via fluorescence activated cell sorting (FACS): Lgr5(high) (stem cells), Lgr5(low) (daughter cells) and Lgr5(negative) (differentiated cells). microRNA profiling and RNA sequencing were performed from the same sample and analyzed. These data refer to 'Comparative effects of diet and carcinogen on microRNA expression in the stem cell niche of the mouse colonic crypt' (Shah et al., 2016) [5].

11.
Biochim Biophys Acta ; 1862(1): 121-34, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26493444

RESUMO

There is mounting evidence that noncoding microRNAs (miRNA) are modulated by select chemoprotective dietary agents. For example, recently we demonstrated that the unique combination of dietary fish oil (containing n-3 fatty acids) plus pectin (fermented to butyrate in the colon) (FPA) up-regulates a subset of putative tumor suppressor miRNAs in intestinal mucosa, and down-regulates their predicted target genes following carcinogen exposure as compared to control (corn oil plus cellulose (CCA)) diet. To further elucidate the biological effects of diet and carcinogen modulated miR's in the colon, we verified that miR-26b and miR-203 directly target PDE4B and TCF4, respectively. Since perturbations in adult stem cell dynamics are generally believed to represent an early step in colon tumorigenesis and to better understand how the colonic stem cell population responds to environmental factors such as diet and carcinogen, we additionally determined the effects of the chemoprotective FPA diet on miRNAs and mRNAs in colonic stem cells obtained from Lgr5-EGFP-IRES-creER(T2) knock-in mice. Following global miRNA profiling, 26 miRNAs (P<0.05) were differentially expressed in Lgr5(high) stem cells as compared to Lgr5(negative) differentiated cells. FPA treatment up-regulated miR-19b, miR-26b and miR-203 expression as compared to CCA specifically in Lgr5(high) cells. In contrast, in Lgr5(negative) cells, only miR-19b and its indirect target PTK2B were modulated by the FPA diet. These data indicate for the first time that select dietary cues can impact stem cell regulatory networks, in part, by modulating the steady-state levels of miRNAs. To our knowledge, this is the first study to utilize Lgr5(+) reporter mice to determine the impact of diet and carcinogen on miRNA expression in colonic stem cells and their progeny.


Assuntos
Carcinógenos , Colo/patologia , Neoplasias do Colo/genética , Dieta , Ácidos Graxos Ômega-3/metabolismo , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Nicho de Células-Tronco , Animais , Carcinógenos/metabolismo , Carcinógenos/toxicidade , Colo/metabolismo , Neoplasias do Colo/etiologia , Neoplasias do Colo/patologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Quinase 2 de Adesão Focal/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos , Camundongos , Fatores de Proteção , Nicho de Células-Tronco/efeitos dos fármacos , Fator de Transcrição 4/genética
12.
Sci Rep ; 4: 5453, 2014 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-24965658

RESUMO

The state and development of the intestinal epithelium is vital for infant health, and increased understanding in this area has been limited by an inability to directly assess epithelial cell biology in the healthy newborn intestine. To that end, we have developed a novel, noninvasive, molecular approach that utilizes next generation RNA sequencing on stool samples containing intact epithelial cells for the purpose of quantifying intestinal gene expression. We then applied this technique to compare host gene expression in healthy term and extremely preterm infants. Bioinformatic analyses demonstrate repeatable detection of human mRNA expression, and network analysis shows immune cell function and inflammation pathways to be up-regulated in preterm infants. This study provides incontrovertible evidence that whole-genome sequencing of stool-derived RNA can be used to examine the neonatal host epithelial transcriptome in infants, which opens up opportunities for sequential monitoring of gut gene expression in response to dietary or therapeutic interventions.


Assuntos
Fezes/citologia , Perfilação da Expressão Gênica/métodos , Mucosa Intestinal/fisiologia , Proteoma/metabolismo , RNA/genética , Análise de Sequência de RNA/métodos , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Masculino , Proteoma/genética
13.
Carcinogenesis ; 35(3): 606-12, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24336194

RESUMO

Arachidonic acid (20:4(Δ5,8,11,14), AA)-derived prostaglandin E2 (PGE2) promotes colon cancer development. In contrast, chemoprotective n-3 polyunsaturated fatty acids supplant AA, thereby decreasing PGE2 biosynthesis in colonocytes, with eicosapentaenoic acid (20:5(Δ5,8,11,14,17), EPA) in particular being metabolized to a novel 3-series E-prostaglandin (PGE3), a putative anti-tumorigenic-cyclooxygenase metabolite. Because transformation of adult stem cells is an extremely important route toward initiating intestinal cancer, we utilized the leucine-rich-repeat-containing G-protein-coupled receptor 5 (Lgr5)-enhanced green fluorescent protein-internal ribosome entry site (IRES)-creER(T2) knock-in mouse model to isolate and culture colonic organoids, in order to document ex vivo responses to exogenous PGE2 and PGE3. Colonic crypts were isolated from transgenic mice and cultured in a Matrigel-based three-dimensional platform. Organoids were treated with exogenous PGE2, PGE3 or dimethyl sulfoxide (vehicle control) for 5 days and the number of viable organoids was recorded daily. Subsequently, samples were processed for immunohistochemistry, flow cytometry and real-time PCR analyses. PGE2 promoted optimal organoid growth and induced significantly higher levels of cell proliferation (P < 0.05) compared with PGE3 and control. In contrast, the Lgr5-green fluorescent protein-positive stem cell number was uniquely elevated by >2-fold in PGE2-treated cultures compared with PGE3 and control. This coincided with the upregulation of stem-cell-related Sox9, Axin2 and Cd44 messenger RNAs. Our results demonstrate that relative to AA-derived PGE2, a known promoter of colon tumorigenesis, EPA-derived PGE3 has diminished ability to support colonic stem cell expansion in mouse colonic organoids.


Assuntos
Divisão Celular/efeitos dos fármacos , Colo/efeitos dos fármacos , Prostaglandinas/farmacologia , Células-Tronco/efeitos dos fármacos , Colo/citologia , Proteínas de Fluorescência Verde/genética , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Células-Tronco/citologia
14.
Biochim Biophys Acta ; 1822(10): 1600-7, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22750333

RESUMO

Since aberrant wound healing and chronic inflammation can promote malignant transformation, we determined whether dietary bioactive fish oil (FO)-derived n-3 polyunsaturated fatty acids (n-3 PUFA) modulate stem cell kinetics in a colitis-wounding model. Lgr5-LacZ and Lgr5-EGFP-IRES-creER(T2) mice were fed diets enriched with n-3 PUFA vs n-6 PUFA (control) and exposed to dextran sodium sulfate (DSS) for 5days in order to induce crypt damage and colitis throughout the colon. Stem cell number, cell proliferation, apoptosis, expression of stem cell (Lgr5, Sox9, Bmi1, Hopx, mTert, Ascl2, and DCAMKL-1) and inflammation (STAT3) markers were quantified. DSS treatment resulted in the ablation of Lgr5(+) stem cells in the distal colon, concurrent with the loss of distal crypt structure and proliferating cells. Lgr5, Ascl2 and Hopx mRNA expression levels were decreased in damaged colonic mucosa. Lgr5(+) stem cells reappeared at day 5 of DSS recovery, with normal levels attained by day 6 of recovery. There was no effect of diet on the recovery of stem cells. FO fed animals exhibited higher levels of phospho-STAT3 at all time points, consistent with a higher wounding by DSS in FO feeding. n-3 PUFA-fed mice exhibited a reduction in stem cell associated factors, Ascl2, Axin2 and EphB3. These results indicate that rapidly cycling Lgr5(+) stem cells residing at position 1 in the colon epithelium are highly susceptible to DSS-induced damage and that dietary cues can impact stem cell regulatory networks.


Assuntos
Colo/fisiologia , Regeneração/fisiologia , Células-Tronco/fisiologia , Cicatrização/fisiologia , Animais , Apoptose/genética , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Colite/induzido quimicamente , Colite/genética , Colite/metabolismo , Colite/patologia , Colo/metabolismo , Colo/patologia , Sulfato de Dextrana , Dieta , Ácidos Graxos Ômega-3/metabolismo , Óleos de Peixe/metabolismo , Expressão Gênica/genética , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Regeneração/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Células-Tronco/metabolismo , Células-Tronco/patologia , Cicatrização/genética
15.
Genome Biol ; 13(4): r32, 2012 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-22546241

RESUMO

BACKGROUND: Gut microbiota and the host exist in a mutualistic relationship, with the functional composition of the microbiota strongly affecting the health and well-being of the host. Thus, it is important to develop a synthetic approach to study the host transcriptome and the microbiome simultaneously. Early microbial colonization in infants is critically important for directing neonatal intestinal and immune development, and is especially attractive for studying the development of human-commensal interactions. Here we report the results from a simultaneous study of the gut microbiome and host epithelial transcriptome of three-month-old exclusively breast- and formula-fed infants. RESULTS: Variation in both host mRNA expression and the microbiome phylogenetic and functional profiles was observed between breast- and formula-fed infants. To examine the interdependent relationship between host epithelial cell gene expression and bacterial metagenomic-based profiles, the host transcriptome and functionally profiled microbiome data were subjected to novel multivariate statistical analyses. Gut microbiota metagenome virulence characteristics concurrently varied with immunity-related gene expression in epithelial cells between the formula-fed and the breast-fed infants. CONCLUSIONS: Our data provide insight into the integrated responses of the host transcriptome and microbiome to dietary substrates in the early neonatal period. We demonstrate that differences in diet can affect, via gut colonization, host expression of genes associated with the innate immune system. Furthermore, the methodology presented in this study can be adapted to assess other host-commensal and host-pathogen interactions using genomic and transcriptomic data, providing a synthetic genomics-based picture of host-commensal relationships.


Assuntos
Aleitamento Materno , Fórmulas Infantis/administração & dosagem , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Metagenoma/imunologia , Metagenômica/métodos , Actinobacteria/classificação , Actinobacteria/genética , Actinobacteria/imunologia , Actinobacteria/isolamento & purificação , Bacteroidetes/classificação , Bacteroidetes/genética , Bacteroidetes/imunologia , Bacteroidetes/isolamento & purificação , Fezes/microbiologia , Perfilação da Expressão Gênica , Humanos , Lactente , Fórmulas Infantis/metabolismo , Mucosa Intestinal/citologia , Análise Multivariada , Filogenia , Proteobactérias/classificação , Proteobactérias/genética , Proteobactérias/imunologia , Proteobactérias/isolamento & purificação , RNA Bacteriano/análise , RNA Bacteriano/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , RNA Ribossômico 16S/análise , RNA Ribossômico 16S/genética , Análise de Sequência de RNA , Simbiose , Transcrição Gênica , Transcriptoma
16.
Br J Nutr ; 106(4): 519-29, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21401974

RESUMO

Both fish oil (FO) and curcumin have potential as anti-tumour and anti-inflammatory agents. To further explore their combined effects on dextran sodium sulphate (DSS)-induced colitis, C57BL/6 mice were randomised to four diets (2 × 2 design) differing in fatty acid content with or without curcumin supplementation (FO, FO+2 % curcumin, maize oil (control, MO) or MO+2 % curcumin). Mice were exposed to one or two cycles of DSS in the drinking-water to induce either acute or chronic intestinal inflammation, respectively. FO-fed mice exposed to the single-cycle DSS treatment exhibited the highest mortality (40 %, seventeen of forty-three) compared with MO with the lowest mortality (3 %, one of twenty-nine) (P = 0·0008). Addition of curcumin to MO increased (P = 0·003) mortality to 37 % compared with the control. Consistent with animal survival data, following the one- or two-cycle DSS treatment, both dietary FO and curcumin promoted mucosal injury/ulceration compared with MO. In contrast, compared with other diets, combined FO and curcumin feeding enhanced the resolution of chronic inflammation and suppressed (P < 0·05) a key inflammatory mediator, NF-κB, in the colon mucosa. Mucosal microarray analysis revealed that dietary FO, curcumin and FO plus curcumin combination differentially modulated the expression of genes induced by DSS treatment. These results suggest that dietary lipids and curcumin interact to regulate mucosal homeostasis and the resolution of chronic inflammation in the colon.


Assuntos
Colite/dietoterapia , Colo/metabolismo , Curcumina/uso terapêutico , Citocinas/metabolismo , Suplementos Nutricionais , Óleos de Peixe/uso terapêutico , Regulação da Expressão Gênica , Doença Aguda , Animais , Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios não Esteroides/uso terapêutico , Doença Crônica , Colite/imunologia , Colite/metabolismo , Colite/patologia , Colo/efeitos dos fármacos , Colo/imunologia , Colo/patologia , Curcumina/efeitos adversos , Citocinas/genética , Sulfato de Dextrana/administração & dosagem , Sulfato de Dextrana/toxicidade , Óleos de Peixe/efeitos adversos , Perfilação da Expressão Gênica , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Irritantes/administração & dosagem , Irritantes/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Distribuição Aleatória , Análise de Sobrevida
17.
Am J Physiol Gastrointest Liver Physiol ; 298(5): G582-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20203060

RESUMO

We have developed a novel molecular methodology that utilizes stool samples containing intact sloughed epithelial cells to quantify intestinal gene expression profiles in the developing human neonate. Since nutrition exerts a major role in regulating neonatal intestinal development and function, our goal was to identify gene sets (combinations) that are differentially regulated in response to infant feeding. For this purpose, fecal mRNA was isolated from exclusively breast-fed (n = 12) and formula-fed (n = 10) infants at 3 mo of age. Linear discriminant analysis was successfully used to identify the single genes and the two- to three-gene combinations that best distinguish the feeding groups. In addition, putative "master" regulatory genes were identified using coefficient of determination analysis. These results support our premise that mRNA isolated from stool has value in terms of characterizing the epigenetic mechanisms underlying the developmentally regulated transcriptional activation/repression of genes known to modulate gastrointestinal function. As larger data sets become available, this methodology can be extended to validation and, ultimately, identification of the main nutritional components that modulate intestinal maturation and function.


Assuntos
Células Epiteliais/metabolismo , Fezes/química , Trato Gastrointestinal/crescimento & desenvolvimento , Fenômenos Fisiológicos da Nutrição do Lactente , Adulto , Aleitamento Materno , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Fórmulas Infantis , Recém-Nascido , Masculino , Análise em Microsséries , Gravidez , RNA Mensageiro/metabolismo
18.
Hum Reprod ; 22(11): 2851-6, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17905746

RESUMO

BACKGROUND: Prostacyclin (PGI(2)) plays an important role in mouse embryo development and implantation. However, it is unclear whether its action is mediated via the I prostaglandin receptor (IP). METHODS: We compared the preimplantation development of IP deleted (IP-/-) embryos and wild-type (WT) embryos. We also evaluated the effect of iloprost, a stable PGI(2) analog, and L-165041, a peroxisome proliferator activated receptor delta (PPARdelta) ligand, on IP-/- versus WT embryos. Finally, we compared the development of heterozygous IP deficient embryos carrying a normal maternal IP allele versus paternal IP allele. RESULTS: Development of IP-/- embryos lagged behind WT embryos and was not enhanced by either the PGI(2) analog or the PPARdelta ligand. WT embryos had slightly higher, although statistically not significant, implantation rates than IP-/- embryos. Heterozygous IP deficient embryos carrying a normal maternal IP allele showed better development and responded to the PGI(2) analog, unlike those carrying the normal paternal IP allele. CONCLUSIONS: IP receptors play an important role in preimplantation embryo development and mediate the embryo's response to exogenous PGI(2). Early embryo development depends on the oocyte IP receptor.


Assuntos
Implantação do Embrião , Epoprostenol/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Receptores de Epoprostenol/genética , Receptores de Epoprostenol/metabolismo , Receptores de Epoprostenol/fisiologia , Transdução de Sinais , Animais , Blastocisto , Desenvolvimento Embrionário , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oócitos/metabolismo , Gravidez , Prenhez
19.
Hum Reprod ; 19(12): 2907-12, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15492023

RESUMO

BACKGROUND: Recently we discovered that human oviducts produce a significant amount of prostacyclin (prostaglandin I2, PGI2) and that PGI2 enhances the potentials of live birth of mouse embryos. However, the eicosanoid profile of mouse oviducts remains unknown. METHODS: The metabolites of [14C]arachidonic acid by mouse oviducts were analysed by high-performance liquid chromatography. The expression of cyclooxygenase (COX)-1, COX-2 and PGI2 synthase (PGIS) was analysed by western blot analysis and immunohistochemistry. The PGI2 synthetic capacities and the COX transcripts during the preimplantation period were compared. The effects of COX-2 inhibitor on PGI2 production were ascertained. RESULTS: Mouse oviducts produced, in order of abundance, PGI2, PGD2 and PGE2. Western blot analysis confirmed the expression of COX-1, -2 and PGIS which were expressed by luminal epithelia and smooth muscle cells. Day 2-3 post-coitus (p.c.) oviducts produced PGI2 10-fold higher than day 4 p.c. oviducts (P = 0.0087); day 1 p.c. oviducts expressed COX-2 transcript 5-fold higher than day 3 p.c. oviducts (P = 0.0004). The PGI2 production was markedly reduced by a selective COX-2 inhibitor. CONCLUSIONS: Mouse oviducts synthesized maximal PGI2 during day 2-3 p.c., coinciding with the transformation of 2-cell embryos to morulae. The results suggest that oviduct-derived PGI2 may enhance embryo development in a paracrine fashion.


Assuntos
Desenvolvimento Embrionário/fisiologia , Epoprostenol/metabolismo , Tubas Uterinas/metabolismo , Animais , Blastocisto , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Eicosanoides/metabolismo , Tubas Uterinas/efeitos dos fármacos , Feminino , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos , Nitrobenzenos/farmacologia , Gravidez , Prostaglandina-Endoperóxido Sintases/genética , Prostaglandina-Endoperóxido Sintases/metabolismo , Sulfonamidas/farmacologia
20.
Hum Reprod ; 19(12): 2900-6, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15489241

RESUMO

INTRODUCTION: The role of prostaglandins (PGs) in embryo hatching remains controversial. In addition, there is no direct evidence that mouse embryos synthesize PGs. METHODS: The effects of endogenous PG on mouse embryo hatching were evaluated by blocking endogenous PG synthesis with indomethacin. Specific cyclooxygenase (COX) inhibitors were used to identify the role of COX-1- and COX-2-derived PGs. An eicosanoid profile was generated by incubating blastocysts with [3H]arachidonic acid and analysing the metabolites by high performance liquid chromatography. The expression and the localization of COX-1, COX-2 and prostacyclin synthase (PGIS) were examined by western blot analysis and immunohistochemistry. RESULTS: The hatching of embryos cultured in 30 microl of protein-free medium was blocked by indomethacin (P = 0.007) or a selective COX-2 inhibitor (P = 0.004). Adding back iloprost, a prostacyclin analogue, abolished the effects of the COX-2 inhibitor. Prostacyclin was the most abundant PG produced by mouse blastocysts, which expressed COX-1, COX-2 and PGIS. COX-1, COX-2 and PGIS were expressed in 4-cell stage embryos and beyond; they were present in the inner cell mass and the trophectoderm of the blastocysts. CONCLUSION: Mouse embryos express COX-1, COX-2 and PGIS which catalyse the formation of PGI2; COX-2-derived PGI2 plays a critical role in embryo hatching.


Assuntos
Embrião de Mamíferos/fisiologia , Epoprostenol/fisiologia , Prostaglandina-Endoperóxido Sintases/metabolismo , Animais , Ácido Araquidônico/metabolismo , Ácido Araquidônico/farmacologia , Blastocisto/efeitos dos fármacos , Blastocisto/fisiologia , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Dinoprostona/farmacologia , Técnicas de Cultura Embrionária , Embrião de Mamíferos/efeitos dos fármacos , Feminino , Iloprosta/farmacologia , Indometacina/farmacologia , Oxirredutases Intramoleculares/metabolismo , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Gravidez , Prostaglandina-Endoperóxido Sintases/efeitos dos fármacos , Prostaglandinas F Sintéticas/farmacologia , Pirazóis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA