Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Proc Natl Acad Sci U S A ; 111(42): E4523-31, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25288742

RESUMO

The mucosa of the small intestine is renewed completely every 3-5 d throughout the entire lifetime by small populations of adult stem cells that are believed to reside in the bottom of the crypts and to migrate and differentiate into all the different populations of intestinal cells. When the cells reach the apex of the villi and are fully differentiated, they undergo cell death and are shed into the lumen. Reactive oxygen species (ROS) production is proportional to the electron transfer activity of the mitochondrial respiration chain. ROS homeostasis is maintained to control cell death and is finely tuned by an inducible antioxidant program. Here we show that peroxisome proliferator-activated receptor-γ coactivator-1ß (PGC-1ß) is highly expressed in the intestinal epithelium and possesses dual activity, stimulating mitochondrial biogenesis and oxygen consumption while inducing antioxidant enzymes. To study the role of PGC-1ß gain and loss of function in the gut, we generated both intestinal-specific PGC-1ß transgenic and PGC-1ß knockout mice. Mice overexpressing PGC-1ß present a peculiar intestinal morphology with very long villi resulting from increased enterocyte lifespan and also demonstrate greater tumor susceptibility, with increased tumor number and size when exposed to carcinogens. PGC-1ß knockout mice are protected from carcinogenesis. We show that PGC-1ß triggers mitochondrial respiration while protecting enterocytes from ROS-driven macromolecule damage and consequent apoptosis in both normal and dysplastic mucosa. Therefore, PGC-1ß in the gut acts as an adaptive self-point regulator, capable of providing a balance between enhanced mitochondrial activity and protection from increased ROS production.


Assuntos
Neoplasias do Colo/patologia , Enterócitos/citologia , Mucosa Intestinal/patologia , Intestino Delgado/patologia , Fatores de Transcrição/metabolismo , Animais , Antioxidantes/metabolismo , Apoptose , Carcinogênese , Transporte de Elétrons , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Homeostase , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Oxigênio/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição/genética
2.
J Biol Chem ; 289(41): 28421-32, 2014 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-25138215

RESUMO

Farnesoid X receptor (FXR, NR1H4) is a bile acid-activated transcription factor that belongs to the nuclear receptor superfamily. It is highly expressed in the enterohepatic system, where it senses bile acid levels to consequently reduce their synthesis while inducing their detoxification. Bile acids are intestinal tumor promoters and their concentrations have to be tightly regulated. Indeed, reduced expression of FXR in the intestine increases colorectal cancer susceptibility in mice, whereas its activation can promote apoptosis in genetically modified cells. Notably, despite the broad knowledge of the FXR enterohepatic transcriptional activity, the molecular mechanisms regulating FXR expression in the intestine are still unknown. Herein, by combining both gain and loss of function approaches and FXR promoter activity studies, we identified caudal-related homeobox 2 (CDX2) transcription factor as a positive regulator of FXR expression in the enterocytes. Our results provide a putative novel tool for modulating FXR expression against bile acid-related colorectal cancer progression.


Assuntos
Polipose Adenomatosa do Colo/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/genética , Transcrição Gênica , Polipose Adenomatosa do Colo/metabolismo , Polipose Adenomatosa do Colo/patologia , Proteína da Polipose Adenomatosa do Colo/genética , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Sítios de Ligação , Fator de Transcrição CDX2 , Linhagem Celular Tumoral , Proteínas de Homeodomínio/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Intestinos/patologia , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas , Ligação Proteica , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo
3.
Gastroenterology ; 144(7): 1518-1529.e3, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23462179

RESUMO

BACKGROUND & AIMS: Studies of the transcriptional networks that regulate nuclear receptor-mediated proliferation of quiescent hepatocytes could lead to new information about liver growth and hepatoprotective strategies. METHODS: We used quantitative real-time PCR to analyze expression of neuron-derived orphan receptor 1 (Nor-1) and its target genes during liver regeneration after hepatectomy in mice, and in hepatocellular carcinoma (HCC) samples from patients. We used adenoviral vectors to express Nor-1 in normal liver (Ad/CMV/V5-Nor-1), or reduce its level with small hairpin RNAs (Ad/BLOCK-iT/Nor-1(small hairpin RNA)) after partial hepatectomy. RESULTS: Levels of Nor-1 messenger RNA and protein, and transcription of Nor-1 target genes (Ccnd1 and Vcam-1), increased during the late priming and proliferative phases of liver regeneration after partial hepatectomy. Levels of NOR-1 messenger RNA and transcription of its target gene CCND1 and of the NOR-1 subfamily member NUR-77 also increased in human HCC samples compared with paired HCC-free tissue. Ad-Nor-1(small hairpin RNA) reduced the hepatocyte proliferation after hepatectomy. Overexpression of Nor-1 in normal livers of mice induced proliferation of quiescent hepatocytes independently of interleukin-6 and tumor necrosis factor-α signaling. In gene expression profile analysis, Nor-1 altered expression of genes involved in the cell cycle, proliferation, and tumorigenesis. CONCLUSIONS: In mice, the orphan nuclear receptor Nor-1 activates proliferation of quiescent hepatocytes and is required for hepatocyte proliferation after partial hepatectomy. Nor-1 and its gene targets are also up-regulated in human HCC samples. Nor-1 activates a transcriptional program that induces hepatocyte proliferation independently of inflammatory signaling pathways.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proliferação de Células , Proteínas de Ligação a DNA/fisiologia , Hepatócitos/citologia , Neoplasias Hepáticas/metabolismo , Regeneração Hepática/fisiologia , Proteínas de Membrana Transportadoras/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Receptores de Esteroides/fisiologia , Receptores dos Hormônios Tireóideos/fisiologia , Animais , Carcinoma Hepatocelular/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Proteínas de Ligação a DNA/genética , Hepatectomia , Humanos , Neoplasias Hepáticas/genética , Regeneração Hepática/genética , Masculino , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , RNA Mensageiro/análise , Receptores de Esteroides/genética , Receptores dos Hormônios Tireóideos/genética , Regulação para Cima , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
4.
Gastroenterology ; 144(7): 1497-507, 1507.e1-13, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23419360

RESUMO

BACKGROUND & AIMS: Liver X receptors (LXRs) are transcriptional regulators of cholesterol metabolism, controlling cholesterol flow into cells, catabolism, and efflux. Cholesterol controls cell proliferation; disruptions in cholesterol metabolism have been associated with the development of colon cancer. We investigated whether expression of activated LXR protects against intestinal tumorigenesis in mice. METHODS: We analyzed the development of colon cancer in mice that express a constitutive active form of LXRα only in the intestinal epithelium, under the control of villin promoter (iVP16LXRα). These mice were crossed with adenomatous polyposis coli (Apc)(min/+) mice, or given azoxymethane followed by dextran sodium sulfate, to assess intestinal tumor formation. We also assessed proliferation and apoptosis of a human colorectal cancer cell line (HT29) transfected with an adenoviral vector that expressed Ad VP16hLXRα, compared with cells expressing AdVP16 (control), and their ability to form xenograft tumors in mice. HT29 cells also were incubated with the LXR ligand GW3965. RESULTS: In human colorectal cancer cells, ligand-induced activation of LXR or transfection with Ad VP16hLXRα blocked the G1 phase, increased caspase-dependent apoptosis, and slowed growth of xenograft tumors in mice. iVP16LXRα mice formed fewer, smaller tumors than VP16 (control) mice after administration of azoxymethane and dextran sodium sulfate. APC(min/+)/iVP16LXRα mice also developed fewer, smaller intestinal tumors than APC(min/+)/iVP16 mice. Gene expression analysis indicated that activation of LXRα affected lipid metabolic networks and increased cholesterol efflux in the intestine. CONCLUSIONS: Expression of activated LXRα blocks proliferation of human colorectal cancer cells and slows the growth of xenograft tumors in mice. It also reduces intestinal tumor formation after administration of chemical carcinogens, and in Apc(min/+) mice. LXR agonists therefore might be developed as therapeutic treatments for colorectal cancer.


Assuntos
Adenocarcinoma/metabolismo , Adenoma/fisiopatologia , Transformação Celular Neoplásica , Neoplasias Colorretais/metabolismo , Neoplasias Intestinais/metabolismo , Receptores Nucleares Órfãos/fisiologia , Adenocarcinoma/patologia , Animais , Proliferação de Células , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Genes APC , Células HT29 , Humanos , Neoplasias Intestinais/patologia , Receptores X do Fígado , Camundongos , Camundongos Transgênicos , Receptores Nucleares Órfãos/metabolismo , Transdução de Sinais , Transplante Heterólogo
5.
Hepatology ; 57(4): 1343-56, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23299802

RESUMO

Development of hepatic steatosis and its progression to steatohepatitis may be the consequence of dysfunction of several metabolic pathways, such as triglyceride synthesis, very low-density lipoprotein (VLDL) secretion, and fatty acid ß-oxidation. Peroxisome proliferator-activated receptor γ coactivator-1ß (PGC-1ß) is a master regulator of mitochondrial biogenesis and oxidative metabolism, lipogenesis, and triglyceride (TG) secretion. Here we generated a novel mouse model with constitutive hepatic activation of PGC-1ß and studied the role of this transcriptional coactivator in dietary-induced steatosis and steatohepatitis. Selective activation of PGC-1ß within hepatocytes is able to protect the liver from lipid overload and from progression to fibrosis. The protective function exerted by PGC-1ß is due to its ability to induce mitochondrial oxidative phosphorylation, fatty acid ß-oxidation, and citrate cycle, as well as to decrease oxidative stress and promote TG secretion in the blood stream. These findings bolster the concept that a combined hepatic specific action of PGC-1ß on lipid synthesis and secretion, as well as on mitochondrial biogenesis and function, could protect against steatohepatitis.


Assuntos
Fígado Gorduroso/metabolismo , Fígado Gorduroso/prevenção & controle , Fígado/metabolismo , Transativadores/metabolismo , Animais , Apoptose/fisiologia , Deficiência de Colina/complicações , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Fígado Gorduroso/etiologia , Fibrose , Metabolismo dos Lipídeos/fisiologia , Fígado/patologia , Camundongos , Camundongos Transgênicos , Renovação Mitocondrial/fisiologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Transativadores/genética , Fatores de Transcrição , Triglicerídeos/sangue
6.
Proc Natl Acad Sci U S A ; 108(16): 6603-8, 2011 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-21467224

RESUMO

Peroxisome proliferator-activated receptor-γ coactivator 1-α (PGC1α) is a transcriptional coactivator able to up-regulate mitochondrial biogenesis, respiratory capacity, oxidative phosphorylation, and fatty acid ß-oxidation with the final aim of providing a more efficient pathway for aerobic energy production. In the continuously renewed intestinal epithelium, proliferative cells in the crypts migrate along the villus axis and differentiate into mature enterocytes, increasing their respiratory capacity and finally undergoing apoptosis. Here we show that in the intestinal epithelial surface, PGC1α drives mitochondrial biogenesis and respiration in the presence of reduced antioxidant enzyme activities, thus determining the accumulation of reactive oxygen species and fostering the fate of enterocytes toward apoptosis. Combining gain- and loss-of-function genetic approaches in human cells and mouse models of intestinal cancer, we present an intriguing scenario whereby PGC1α regulates enterocyte cell fate and protects against tumorigenesis.


Assuntos
Antioxidantes/metabolismo , Enterócitos/metabolismo , Proteínas de Choque Térmico/metabolismo , Neoplasias Intestinais/metabolismo , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição/metabolismo , Animais , Transformação Celular Neoplásica , Enterócitos/patologia , Proteínas de Choque Térmico/genética , Humanos , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Camundongos , Camundongos Nus , Proteínas de Neoplasias/genética , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Consumo de Oxigênio/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/genética
7.
Gastroenterology ; 142(2): 355-65.e1-4, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22057115

RESUMO

BACKGROUND & AIMS: Cholestasis is a liver disorder characterized by impaired bile flow, reduction of bile acids (BAs) in the intestine, and retention of BAs in the liver. The farnesoid X receptor (FXR) is the transcriptional regulator of BA homeostasis. Activation of FXR by BAs reduces circulating BA levels in a feedback mechanism, repressing hepatic cholesterol 7α-hydroxylase (Cyp7a1), the rate-limiting enzyme for the conversion of cholesterol to BAs. This mechanism involves the hepatic nuclear receptor small heterodimer partner and the intestinal fibroblast growth factor (FGF) 19 and 15. We investigated the role of activation of intestine-specific FXR in reducing hepatic levels of BAs and protecting the liver from cholestasis in mice. METHODS: We generated transgenic mice that express a constitutively active FXR in the intestine. Using FXR gain- and loss-of-function models, we studied the roles of intestinal FXR in mice with intrahepatic and extrahepatic cholestasis. RESULTS: Selective activation of intestinal FXR induced FGF15 and repressed hepatic Cyp7a1, reducing the pool size of BAs and changing the BA pool composition. Activation of intestinal FXR protected mice from obstructive extrahepatic cholestasis after bile duct ligation or administration of α-naphthylisothiocyanate. In Mdr2(-/-) mice, transgenic expression of activated FXR in the intestine protected against liver damage, whereas absence of FXR promoted progression of liver disease. CONCLUSIONS: Activation of FXR transcription in the intestine protects the liver from cholestasis in mice by inducing FGF15 expression and reducing the hepatic pool of BA; this approach might be developed to reverse cholestasis in patients.


Assuntos
Ácidos e Sais Biliares/metabolismo , Colestase/prevenção & controle , Mucosa Intestinal/metabolismo , Fígado/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Ativação Transcricional , Animais , Colestase/metabolismo , Colestase/patologia , Colesterol 7-alfa-Hidroxilase/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Transgênicos , Distribuição Aleatória , Receptores Citoplasmáticos e Nucleares/genética
8.
Biology (Basel) ; 12(8)2023 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-37626950

RESUMO

The precise arrangement and peculiar interaction of transverse tubule (T-tubule) and sarcoplasmic reticulum (SR) membranes efficiently guarantee adequate contractile properties of skeletal muscle fibers. Fast muscle fibers from mice lacking calsequestrin 1 (CASQ1) are characterized by the profound ultrastructural remodeling of T-tubule/SR junctions. This study investigates the role of CASQ1, an essential component of calcium release units (CRUs), in the postnatal development of muscle fibers. By using CASQ1-knockout mice, we examined the maturation of CRUs and the involvement of different junctional proteins in the juxtaposition of the membrane system. Our morphological investigation of both wild-type (WT) and CASQ1-null extensor digitorum longus (EDL) fibers, from 1 week to 4 months of age, yielded noteworthy findings. Firstly, we observed that the absence of CASQ1 hindered the full maturation of CRUs, despite the correct localization of key junctional components (ryanodine receptor, dihydropyridine receptor, and triadin) to the junctional SR in adult animals. Furthermore, analysis of protein expression profiles related to T-tubule biogenesis and organization (junctophilin 1, amphiphysin 2, caveolin 3, and mitsugumin 29) demonstrated delayed progression in their expression during postnatal development in the absence of CASQ1, suggesting the impaired maturation of CRUs. The absence of CASQ1 directly impacts the proper assembly of CRUs during development and influences the expression and coordination of other proteins involved in T-tubule biogenesis and organization.

9.
Life (Basel) ; 13(2)2023 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-36836748

RESUMO

Among the many factors inducing prostate inflammation, bacterial contribution is potentially underrated according to the scientific community. Bacterial prostatitis is characterized by modifications of the prostatic microenvironment, mainly driven by the immune system. Macrophages play a major role in bacterial prostatitis, secreting a plethora of proinflammatory and chemoattractive cytokines and proteolytic enzymes able to degrade the ECM, so facilitating the invasion of other immune cells. Consequently, macrophages represent a link between bacterial infection and prostate inflammation, as well as being the main target of prostate anti-inflammatory drugs and dietary supplements. This study aims to investigate the effect of a formulation composed of active principles and a probiotic strain with a particular focus on the anti-inflammatory effect in an in vitro bacterial prostatitis model. The results obtained showed that the formulation reduces the inflammatory response of prostatic epithelium induced by bacterial infection. This effect is mediated by the modulation of activated macrophages. Analysis of the cytokines released highlights that the tested formulation is able to reduce the expression of key proinflammatory cytokines involved in the pathogenesis of prostate diseases, in particular prostate cancer, and represents a valuable tool to prevent bacterial prostatitis and ensure favorable prostate health.

10.
Biochim Biophys Acta ; 1812(8): 851-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21540105

RESUMO

UNLABELLED: Hyperactivation of NF-κB is a key factor in the pathophysiology of inflammatory bowel disease (IBD). We previously showed that the bile salt nuclear Farnesoid X Receptor (FXR) counter-regulates intestinal inflammation, possibly via repression of NF-κB. Here, we examine whether mutual antagonism between NF-κB and FXR exists. FXR and its target genes IBABP and FGF15/19 expression were determined in HT29 colon carcinoma cells and ex vivo in intestinal specimens of wild type (WT) and Fxr-ko mice, treated with/without FXR ligands (GW4064/INT-747) and inflammatory stimuli (TNFα/IL-1ß). In addition, FXR activation was studied in vivo in WT and Fxr-ko mice with DSS-colitis. The involvement of NF-κB in decreasing FXR activity was investigated by reporter assays and Glutathione S-transferase pulldown assays. FXR target gene expression was highly reduced by inflammatory stimuli in all model systems, while FXR mRNA expression was unaffected. In line with these results, reporter assays showed reduced FXR transcriptional activity upon TNFα/IL-1ß stimulation. We show that this reduction in FXR activity is probably mediated by NF-κB, since overexpression of NF-κB subunits p50 and/or p65 also lead to inhibition of FXR activity. Finally, we report that p65 and p50 physically interact with FXR in vitro. CONCLUSIONS: Together, these results indicate that intestinal inflammation strongly reduces FXR activation, probably via NF-κB-dependent tethering of FXR. Therefore, FXR not only inhibits inflammation, but also is targeted by the inflammatory response itself. This could result in a vicious cycle where reduced FXR activity results in less repression of inflammation, contributing to development of chronic intestinal inflammation. This article is part of a Special Issue entitled: Translating nuclear receptors from health to disease.


Assuntos
Ácidos e Sais Biliares/metabolismo , Citocinas/fisiologia , Mediadores da Inflamação/fisiologia , Mucosa Intestinal/metabolismo , NF-kappa B/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais , Animais , Linhagem Celular , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Gut ; 60(4): 463-72, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21242261

RESUMO

BACKGROUND & AIMS: Inflammatory bowel disease (IBD) is characterised by chronic intestinal inflammation, resulting from dysregulation of the mucosal immune system and compromised intestinal epithelial barrier function. The bile salt, nuclear farnesoid X receptor (FXR), was recently implicated in intestinal antibacterial defence and barrier function. The aim of this study was to investigate the therapeutic potential of FXR agonists in the treatment of intestinal inflammation in complementary in vivo and in vitro models. METHODS: Colitis was induced in wild-type (WT) and Fxr-null mice using dextran sodium sulfate, and in WT mice using trinitrobenzenesulfonic acid. Mice were treated with vehicle or the FXR agonist INT-747, and colitis symptoms were assessed daily. Epithelial permeability assays and cytokine expression analysis were conducted in mouse colon and enterocyte-like cells (Caco-2/HT29) treated with medium or INT-747. Inflammatory cytokine secretion was determined by ELISA in various human immune cell types. RESULTS: INT-747-treated WT mice are protected from DSS- and TNBS-induced colitis, as shown by significant reduction of body weight loss, epithelial permeability, rectal bleeding, colonic shortening, ulceration, inflammatory cell infiltration and goblet cell loss. Furthermore, Fxr activation in intestines of WT mice and differentiated enterocyte-like cells downregulates expression of key proinflammatory cytokines and preserves epithelial barrier function. INT-747 significantly decreases tumour necrosis factor α secretion in activated human peripheral blood mononuclear cells, purified CD14 monocytes and dendritic cells, as well as in lamina propria mononuclear cells from patients with IBD. CONCLUSIONS: FXR activation prevents chemically induced intestinal inflammation, with improvement of colitis symptoms, inhibition of epithelial permeability, and reduced goblet cell loss. Furthermore, FXR activation inhibits proinflammatory cytokine production in vivo in the mouse colonic mucosa, and ex vivo in different immune cell populations. The findings provide a rationale to explore FXR agonists as a novel therapeutic strategy for IBD.


Assuntos
Ácido Quenodesoxicólico/análogos & derivados , Doenças Inflamatórias Intestinais/tratamento farmacológico , Absorção Intestinal/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/fisiologia , Animais , Células CACO-2 , Ácido Quenodesoxicólico/farmacologia , Ácido Quenodesoxicólico/uso terapêutico , Colo/metabolismo , Citocinas/metabolismo , Sulfato de Dextrana , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Íleo/metabolismo , Mediadores da Inflamação/metabolismo , Doenças Inflamatórias Intestinais/induzido quimicamente , Doenças Inflamatórias Intestinais/fisiopatologia , Absorção Intestinal/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores Citoplasmáticos e Nucleares/agonistas , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Ácido Trinitrobenzenossulfônico , Fator de Necrose Tumoral alfa/biossíntese
12.
Gastroenterology ; 138(2): 636-48, 648.e1-12, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19818784

RESUMO

BACKGROUND & AIMS: The WNT-adenomatous polyposis coli system controls cell fate in the intestinal epithelium, where compartment-specific genes tightly regulate proliferation, migration, and differentiation. Nuclear receptors are transcription factors functioning as sensors of hormones and nutrients that are known to contribute to colon cancer progression. Here we mapped the messenger RNA (mRNA) abundance and the epithelial localization of the entire nuclear receptor family in mouse and human intestine. METHODS: We used complementary high-resolution in situ hybridization and systematic real-time quantitative polymerase chain reaction in samples of normal distal ileum and proximal colon mucosa and tumors obtained from mouse and human adenomatous polyposis coli-initiated tumor models (ie, Apc(Min/+) mice and familial adenomatous polyposis patients) and in cellular models of human colon cancer. RESULTS: We first defined for each receptor an expression pattern based on its transcript localization in the distal ileum and the proximal colon. Then, we compared the mRNA levels between normal intestinal epithelium and neoplastic intestinal tissue. After analyzing the correspondence between mouse and human tumor samples plus genetically modified human colon cancer cells, we used complementary graphic and statistical approaches to present a comprehensive overview with several classification trees for the nuclear hormone receptor intestinal transcriptome. CONCLUSIONS: We defined the intestinal nuclear hormone receptor map, which indicates that the localization pattern of a receptor in normal intestine predicts the modulation of its expression in tumors. Our results are useful to select those nuclear receptors that could be used eventually as early diagnostic markers or targeted for clinical intervention in intestinal polyposis and cancer.


Assuntos
Adenoma/metabolismo , Colo/metabolismo , Neoplasias do Colo/metabolismo , Células Epiteliais/metabolismo , Íleo/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Adenoma/patologia , Polipose Adenomatosa do Colo/metabolismo , Polipose Adenomatosa do Colo/patologia , Adolescente , Adulto , Animais , Colo/patologia , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Células Epiteliais/patologia , Feminino , Perfilação da Expressão Gênica , Humanos , Íleo/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Adulto Jovem
13.
Hepatology ; 51(4): 1334-44, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20044803

RESUMO

Cholesterol homeostasis is critical for cellular proliferation. Liver X receptor (LXR) alpha and beta are the nuclear receptors responsible for regulation of cholesterol metabolism. In physiological conditions, high intracellular cholesterol levels cause increased synthesis of oxysterols, which activate LXR, thus triggering a transcriptional response for cholesterol secretion and catabolism. Here we employed a mouse model of partial hepatectomy (PH) to dissect the molecular pathways connecting cholesterol homeostasis, cellular proliferation, and LXR. First, we show that hepatic cholesterol content increases after PH, whereas the entire LXR transcriptome is down-regulated. Although LXR messenger RNA (mRNA) levels are unmodified, LXR target genes are significantly down-regulated on day 1 after PH and restored to control levels on day 7, when the liver reaches normal size. The inactivation of LXR following PH is related to the reduced oxysterol availability by way of decreased synthesis, and increased sulfation and secretion. On the contrary, cholesterol synthesis is up-regulated, and extracellular matrix remodeling is enhanced. Second, we show that reactivation of LXR by way of a synthetic ligand determines a negative modulation of hepatocyte proliferation. This effect is sustained by the reactivation of hepatic cholesterol catabolic and secretory pathways, coupled with a significant reduction of cholesterol biosynthesis. Our data unveil a previously unrecognized and apparently paradoxical scenario of LXR modulation. During liver regeneration LXR activity is abated in spite of increasing intracellular cholesterol levels. Turning off LXR-transcriptional pathways is crucial to guaranteeing the requisite intracellular cholesterol levels of regenerating hepatocytes. In line with this hypothesis, pharmacological LXR reactivation during PH significantly reduces liver regeneration capacity.


Assuntos
Colesterol/metabolismo , Perfilação da Expressão Gênica , Hepatócitos/metabolismo , Receptores Nucleares Órfãos/fisiologia , Animais , Proliferação de Células , Matriz Extracelular/metabolismo , Hepatectomia , Regeneração Hepática , Receptores X do Fígado , Masculino , Metaloproteinase 9 da Matriz/genética , Camundongos , Receptores Nucleares Órfãos/antagonistas & inibidores , Receptores Nucleares Órfãos/genética , Triglicerídeos/metabolismo
14.
Nutrients ; 13(12)2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34960072

RESUMO

Erectile dysfunction affects more than 50% of diabetic male patients, with a higher prevalence compared with the general population. Age, clinical factors, and lifestyle habits have been suggested to contribute to the pathophysiology and worsening of erectile dysfunction in diabetic patients. First- and second-line standard treatments are represented by phosphodiesterase type 5 (PDE5) inhibitors and alprostadil, respectively. However, natural compounds have been suggested to ameliorate this clinical condition. This study aims to preclinically characterize the potential synergism among plant-derived products for the improvement of erectile dysfunction in the diabetic condition. The effects of a nutritional supplement composed of Panax ginseng, Moringa oleifera and rutin, as single agents or as a mixture, were evaluated in a streptozotocin (STZ)-induced diabetic rat model with erectile dysfunction. The treatment efficacy was evaluated by measuring sexual-related parameters (i.e., mount and intromission latencies, the mount and intromission frequencies and the ejaculation latency). Results showed that only the mixture was able to significantly reduce the diabetes-related delay in mount latency (p < 0.01). Substantial similar effects were observed by measuring the intromission latency and the mean number of mounts was very similar between rats treated with the mixture and controls. Single agent treatments showed very low effects in terms of intromission frequency, whereas the mixture was able to increase this parameter. Additionally, a statistically significant reduced ejaculation latency was observed in rats treated with the mixture compared with the STZ control. These results are in agreement with the available literature and suggest that the study mixture may ameliorate sexual behavior compared with the administration of the study natural compounds as single agents in diabetic rats. Further preclinical and clinical studies are needed to perform a more comprehensive evaluation of the efficacy and safety of the study mixture.


Assuntos
Produtos Biológicos/farmacologia , Complicações do Diabetes/tratamento farmacológico , Diabetes Mellitus Experimental , Suplementos Nutricionais , Disfunção Erétil/etiologia , Extratos Vegetais/farmacologia , Animais , Produtos Biológicos/química , Humanos , Masculino , Moringa oleifera/química , Panax/química , Ereção Peniana/efeitos dos fármacos , Fitoterapia , Extratos Vegetais/química , Ratos , Rutina/química , Comportamento Sexual Animal
15.
J Neural Transm Suppl ; (73): 333-41, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20411791

RESUMO

There is extensive evidence that oxidative damage of dopamine (DA)-containing neurons in the substantia nigra pars compacta (SNc) may contribute to the pathogenesis of Parkinson's disease (PD). We evaluated the potential neuroprotective effect of diets enriched with wild-type Red Setter (RS) tomato or transgenic High Carotene (HC) tomato, rich in beta-carotene, obtained by the activation of lycopene beta-cyclase (tlcy-b), in an animal model of PD. Male Fischer 344 rats were fed for 14 days with standard Altromin diet, 5% RS- or 5% HC-enriched diet. Seven days after the beginning of this diet regimen, the rats were lesioned by 6-hydroxydopamine (6-OHDA) injected into the left SNc. After further 7 days, the rats were sacrificed, and DA and 3,4-dihydroxyphenylacetic acid (DOPAC) levels in both the left (ipsilateral) and the right (contralateral) striata were measured. Striatal DA levels were reduced by 86.5 +/- 5.0% in control, 86.2 +/- 5.0% in HC-, and 56.0 +/- 9.0% in RS-fed group. Striatal DOPAC was decreased by 85.6 +/- 5.0% in controls, 83.0 +/- 6.0% in HC-, and 58.9 +/- 10.0% in RS-fed group. Blood was obtained from the rats on day 14 and the plasma level of licopene and beta-carotene was measured by liquid chromatography-atmospheric pressure chemical ionization-mass spectrometry (LC-APCI-MS) for the determination of lycopene and beta-carotene levels. The plasma level of lycopene was 4.7 +/- 0.2 ng/ml in 5% RS-fed rats, while it was undetectable (< 2.5 ng ml(-1)) in control and HC-fed rats. The efficacy of RS diet to preserve striatal dopaminergic innervation can be attributed to the ability of lycopene to prevent the degeneration of DA-containing neurons in the SNc.


Assuntos
Dopamina/metabolismo , Degeneração Neural/patologia , Degeneração Neural/prevenção & controle , Solanum lycopersicum/química , Substância Negra/patologia , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Animais , Carotenoides/administração & dosagem , Carotenoides/biossíntese , Modelos Animais de Doenças , Lateralidade Funcional , Liases Intramoleculares/sangue , Liases Intramoleculares/genética , Solanum lycopersicum/genética , Masculino , Degeneração Neural/sangue , Degeneração Neural/etiologia , Oxidopamina/toxicidade , Doença de Parkinson/complicações , Doença de Parkinson/etiologia , Plantas Geneticamente Modificadas , Ratos , Ratos Endogâmicos F344
16.
J Agric Food Chem ; 55(9): 3398-407, 2007 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-17394337

RESUMO

The in vitro biochemical stability of caffeic acid phenethyl ester in rat and human plasma was investigated and compared with the stability of other caffeic acid esters (chlorogenic acid and rosmarinic acid). The incubation of the compounds in rat plasma for up to 6 h showed that caffeic acid phenethyl ester, but not the other compounds, was hydrolyzed, whereas human plasma did not affect the stability of all the assayed compounds. The products in rat plasma were caffeic acid and an unknown compound, which was identified by mass spectrometry as caffeic acid ethyl ester, produced by transesterification in the presence of ethanol used as vehicle for standard compounds. Specific inhibitors of different plasma esterases allowed the identification of a carboxylesterase as the enzyme involved in the metabolism of caffeic acid phenethyl ester. The oral administration in rats of caffeic acid phenethyl ester in the presence of both ethanol and 2-(2-ethoxyethoxy)ethanol gave rise to a dramatic increase of caffeic acid, as well as low levels of caffeic acid phenethyl ester, caffeic acid ethyl ester, and caffeic acid 2-(2-ethoxyethoxy)ethyl ester, in urine collected within 24 h after treatment. These results suggest that caffeic acid phenethyl ester is hydrolyzed also in vivo to caffeic acid as the major metabolite and that its biological activities should be more properly assayed and compared with those of caffeic acid, its bioactive hydrolysis product. Moreover, alcohols should be carefully used in vivo as solvents for caffeic acid phenethyl ester, since they can give rise to new bioactive caffeic acid esters.


Assuntos
Ácidos Cafeicos/administração & dosagem , Ácidos Cafeicos/sangue , Ácidos Cafeicos/farmacocinética , Álcool Feniletílico/análogos & derivados , Própole/química , Animais , Ácido Clorogênico/sangue , Cinamatos/sangue , Depsídeos/sangue , Estabilidade de Medicamentos , Humanos , Hidrólise , Álcool Feniletílico/administração & dosagem , Álcool Feniletílico/sangue , Álcool Feniletílico/farmacocinética , Ratos , Ácido Rosmarínico
17.
Artigo em Inglês | MEDLINE | ID: mdl-15358316

RESUMO

Caffeic acid phenethyl ester (CAPE) is one of the most bioactive compounds of propolis, a resinous substance collected and elaborated by honeybees. A new liquid chromatography-electrospray ionisation tandem mass spectrometric method was developed and validated for its determination in rat plasma and urine, using taxifolin as internal standard. After sample preparation by liquid/liquid extraction with ethyl acetate, chromatographic separations were carried out with an ODS-RP column using a binary mobile phase gradient of acetonitrile in water. Detection was performed using a turboionspray source operated in negative ion mode and by multiple reaction monitoring. The method was validated, showing good selectivity, sensitivity (LOD = 1 ng/ml), linearity (5-1000 ng/ml; r > or = 0.9968), intra- and inter-batch precision and accuracy (< or =14.5%), and recoveries (94-106%) in both plasma and urine. Stability assays have shown that CAPE is rapidly hydrolysed by plasmatic esterases, which are however inhibited by sodium fluoride. The method was applied to the determination of CAPE levels in rat plasma and urine after oral administration, showing that CAPE is rapidly absorbed and excreted in urine both as unmodified molecule and as glucuronide conjugate.


Assuntos
Ácidos Cafeicos/análise , Álcool Feniletílico/análogos & derivados , Álcool Feniletílico/análise , Animais , Ácidos Cafeicos/sangue , Ácidos Cafeicos/urina , Cromatografia Líquida de Alta Pressão , Glucuronidase/química , Glucuronídeos/sangue , Glucuronídeos/urina , Masculino , Álcool Feniletílico/sangue , Álcool Feniletílico/urina , Ratos , Padrões de Referência , Reprodutibilidade dos Testes , Espectrometria de Massas por Ionização por Electrospray
18.
Cell Rep ; 7(1): 12-8, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24656817

RESUMO

Gut microbiota influences host health status by providing trophic, protective, and metabolic functions, including bile acid (BA) biotransformation. Microbial imprinting on BA signature modifies pool size and hydrophobicity, thus contributing to BA enterohepatic circulation. Microbiota-targeted therapies are now emerging as effective strategies for preventing and/or treating gut-related diseases. Here, we show that gut microbiota modulation induced by VSL#3 probiotics enhances BA deconjugation and fecal excretion in mice. These events are associated with changes in ileal BA absorption, repression of the enterohepatic farnesoid X receptor-fibroblast growth factor 15 (FXR-FGF15) axis, and increased hepatic BA neosynthesis. Treatment with a FXR agonist normalized fecal BA levels in probiotic-administered mice, whereas probiotic-induced alterations in BA metabolism are abolished upon FXR and FGF15 deficiency. Our data provide clear in vivo evidence that VSL#3 probiotics promote ileal BA deconjugation with subsequent fecal BA excretion and induce hepatic BA neosynthesis via downregulation of the gut-liver FXR-FGF15 axis.


Assuntos
Ácidos e Sais Biliares/biossíntese , Fatores de Crescimento de Fibroblastos/metabolismo , Microbiota/efeitos dos fármacos , Probióticos/farmacologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Regulação para Baixo , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/microbiologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais
19.
Cancer Lett ; 344(1): 110-118, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24215867

RESUMO

Chemoresistance is a major obstacle to effective therapy against colorectal cancer (CRC) and may lead to deadly consequences. The metabolism of CRC cells depends highly on the p38 MAPK pathway, whose involvement in maintaining a chemoresistant behavior is currently being investigated. Our previous studies revealed that p38α is the main p38 isoform in CRC cells. Here we show that p38α pharmacological inhibition combined with cisplatin administration decreases colony formation and viability of cancer cells and strongly increases Bax-dependent apoptotic cell death by activating the tumor suppressor protein FoxO3A. Our results indicate that FoxO3A activation up-regulates transcription of its target genes (p21, PTEN, Bim and GADD45), which forces both chemosensitive and chemoresistant CRC cells to undergo apoptosis. Additionally, we found that FoxO3A is required for apoptotic cell death induction, as confirmed by RNA interference experiments. In animal models xenografted with chemoresistant HT29 cells, we further confirmed that the p38-targeted dual therapy strategy produced an increase in apoptosis in cancer tissue leading to tumor regression. Our study uncovers a major role for the p38-FoxO3A axis in chemoresistance, thereby suggesting a new therapeutic approach for CRC treatment; moreover, our results indicate that Bax status may be used as a predictive biomarker.


Assuntos
Neoplasias Colorretais/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Terapia de Alvo Molecular/métodos , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular , Cisplatino/farmacologia , Feminino , Citometria de Fluxo , Imunofluorescência , Proteína Forkhead Box O3 , Células HT29 , Humanos , Immunoblotting , Camundongos , Camundongos Nus , Reação em Cadeia da Polimerase em Tempo Real , Ensaios Antitumorais Modelo de Xenoenxerto
20.
PLoS One ; 9(8): e104449, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25116592

RESUMO

BACKGROUND & AIMS: Liver regeneration (LR) is a valuable model for studying mechanisms modulating hepatocyte proliferation. Nuclear receptors (NRs) are key players in the control of cellular functions, being ideal modulators of hepatic proliferation and carcinogenesis. METHODS & RESULTS: We used a previously validated RT-qPCR platform to profile modifications in the expression of all 49 members of the NR superfamily in mouse liver during LR. Twenty-nine NR transcripts were significantly modified in their expression during LR, including fatty acid (peroxisome proliferator-activated receptors, PPARs) and oxysterol (liver X receptors, Lxrs) sensors, circadian masters RevErbα and RevErbß, glucocorticoid receptor (Gr) and constitutive androxane receptor (Car). In order to detect the NRs that better characterize proliferative status vs. proliferating liver, we used the novel Random Forest (RF) analysis to selected a trio of down-regulated NRs (thyroid receptor alpha, Trα; farsenoid X receptor beta, Fxrß; Pparδ) as best discriminators of the proliferating status. To validate our approach, we further studied PPARδ role in modulating hepatic proliferation. We first confirmed the suppression of PPARδ both in LR and human hepatocellular carcinoma at protein level, and then demonstrated that PPARδ agonist GW501516 reduces the proliferative potential of hepatoma cells. CONCLUSIONS: Our data suggest that NR transcriptome is modulated in proliferating liver and is a source of biomarkers and bona fide pharmacological targets for the management of liver disease affecting hepatocyte proliferation.


Assuntos
Carcinoma Hepatocelular/genética , Hepatócitos/metabolismo , Neoplasias Hepáticas/genética , Regeneração Hepática/genética , Receptores Citoplasmáticos e Nucleares/genética , Animais , Carcinoma Hepatocelular/metabolismo , Linhagem Celular , Proliferação de Células , Análise por Conglomerados , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Fígado/metabolismo , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Receptores Citoplasmáticos e Nucleares/metabolismo , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA