Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(24)2022 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-36555462

RESUMO

During mouse pregnancy placental lactogens stimulate prolactin receptors on pancreatic islet beta cells to induce expression of the tryptophan hydroxylase Tph1, resulting in the synthesis and secretion of serotonin. Presently, the functional relevance of this phenomenon is unclear. One hypothesis is that serotonin-induced activation of 5-HT2B receptors on beta cells stimulates beta cell proliferation during pregnancy. We tested this hypothesis via three different mouse models: (i) total Tph1KO mice, (ii) 129P2/OlaHsd mice, which are incompetent to upregulate islet Tph1 during pregnancy, whereas Tph1 is normally expressed in the intestine, mammary glands, and placenta, and (iii) Htr2b-deficient mice. We observed normal pregnancy-induced levels of beta cell proliferation in total Tph1KO mice, 129P2/OlaHsd mice, and in Htr2b-/- mice. The three studied mouse models indicate that islet serotonin production and its signaling via 5-HT2B receptors are not required for the wave of beta cell proliferation that occurs during normal mouse pregnancy.


Assuntos
Células Secretoras de Insulina , Ilhotas Pancreáticas , Feminino , Animais , Gravidez , Camundongos , Serotonina/metabolismo , Placenta/metabolismo , Ilhotas Pancreáticas/metabolismo , Células Secretoras de Insulina/metabolismo , Proliferação de Células , Triptofano Hidroxilase/genética , Triptofano Hidroxilase/metabolismo
2.
Diabetologia ; 59(7): 1356-1363, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27056372

RESUMO

Pregnancy is a key mammalian reproductive event in which growth and differentiation of the fetus imposes extra metabolic and hormonal demands on the mother. Its successful outcome depends on major changes in maternal blood circulation, metabolism and endocrine function. One example is the endocrine pancreas, where beta cells undergo a number of changes in pregnancy that result in enhanced functional beta cell mass in order to compensate for the rising metabolic needs for maternal insulin. During the last 5 years, a series of studies have increased our understanding of the molecular events involved in this functional adaptation. In the mouse, a prominent functional change during pregnancy is the capacity of some beta cells to produce serotonin. In this review we will discuss the mechanism and potential effects of pregnancy-related serotonin production in beta cells, considering functional consequences at the local intra-islet and systemic level.


Assuntos
Ilhotas Pancreáticas/metabolismo , Serotonina/metabolismo , Animais , Feminino , Células Secretoras de Insulina/metabolismo , Camundongos , Gravidez , Receptores da Prolactina/genética , Receptores da Prolactina/metabolismo , Serotonina/genética , Triptofano Hidroxilase/metabolismo
3.
Genome Res ; 21(1): 95-105, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21088282

RESUMO

We report on a hitherto poorly characterized class of genes that are expressed in all tissues, except in one. Often, these genes have been classified as housekeeping genes, based on their nearly ubiquitous expression. However, the specific repression in one tissue defines a special class of "disallowed genes." In this paper, we used the intersection-union test to screen for such genes in a multi-tissue panel of genome-wide mRNA expression data. We propose that disallowed genes need to be repressed in the specific target tissue to ensure correct tissue function. We provide mechanistic data of repression with two metabolic examples, exercise-induced inappropriate insulin release and interference with ketogenesis in liver. Developmentally, this repression is established during tissue maturation in the early postnatal period involving epigenetic changes in histone methylation. In addition, tissue-specific expression of microRNAs can further diminish these repressed mRNAs. Together, we provide a systematic analysis of tissue-specific repression of housekeeping genes, a phenomenon that has not been studied so far on a genome-wide basis and, when perturbed, can lead to human disease.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Fígado/metabolismo , Pâncreas/metabolismo , Animais , Epigenômica , Feminino , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Lactato Desidrogenases/genética , Lactato Desidrogenases/metabolismo , Fígado/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Pâncreas/citologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Simportadores/genética , Simportadores/metabolismo
4.
Proc Natl Acad Sci U S A ; 107(11): 5208-13, 2010 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-20194741

RESUMO

Glucose homeostasis is critically dependent on insulin release from pancreatic beta-cells, which is strictly regulated by glucose-induced oscillations in membrane potential (V(m)) and the cytosolic calcium level ([Ca(2+)](cyt)). We propose that TRPM5, a Ca(2+)-activated monovalent cation channel, is a positive regulator of glucose-induced insulin release. Immunofluorescence revealed expression of TRPM5 in pancreatic islets. A Ca(2+)-activated nonselective cation current with TRPM5-like properties is significantly reduced in Trpm5(-/-) cells. Ca(2+)-imaging and electrophysiological analysis show that glucose-induced oscillations of V(m) and [Ca(2+)](cyt) have on average a reduced frequency in Trpm5(-/-) islets, specifically due to a lack of fast oscillations. As a consequence, glucose-induced insulin release from Trpm5(-/-) pancreatic islets is significantly reduced, resulting in an impaired glucose tolerance in Trpm5(-/-) mice.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Glucose/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Canais de Cátion TRPM/deficiência , Animais , Cátions , Regulação da Expressão Gênica/efeitos dos fármacos , Teste de Tolerância a Glucose , Insulina/metabolismo , Secreção de Insulina , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Ilhotas Pancreáticas/citologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Fenótipo , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo
5.
Am J Gastroenterol ; 106(4): 748-61, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21326222

RESUMO

OBJECTIVES: Inflammatory bowel disease (IBD) is characterized by a continuous influx of leukocytes into the gut wall. This migration is regulated by cell adhesion molecules (CAMs), and selective antimigration therapies have been developed. This study investigated the effect of infliximab therapy on the mucosal gene expression of CAMs in IBD. METHODS: Mucosal gene expression of 69 leukocyte/endothelial CAMs and E-cadherin was investigated in 61 IBD patients before and after first infliximab infusion and in 12 normal controls, using Affymetrix gene expression microarrays. Quantitative reverse transcriptase-PCR (qRT-PCR), immunohistochemistry, and western blotting were used to confirm the microarray data. RESULTS: When compared with control colons, the colonic mucosal gene expression of most leukocyte/endothelial adhesion molecules was upregulated and E-cadherin gene expression was downregulated in active colonic IBD (IBDc) before therapy, with no significant colonic gene expression differences between ulcerative colitis and colonic Crohn's disease. Infliximab therapy restored the upregulations of leukocyte CAMs in IBDc responders to infliximab that paralleled the disappearance of the inflammatory cells from the colonic lamina propria. Also, the colonic gene expression of endothelial CAMs and of most chemokines/chemokine receptors returned to normal after therapy in IBDc responders, and only CCL20 and CXCL1-2 expression remained increased after therapy in IBDc responders vs. control colons. When compared with control ileums, the ileal gene expression of MADCAM1, THY1, PECAM1, CCL28, CXCL1, -2, -5, -6, and -11, and IL8 was increased and CD58 expression was decreased in active ileal Crohn's disease (CDi) before therapy, and none of the genes remained dysregulated after therapy in CDi responders vs. control ileums. This microarray study identified a number of interesting targets for antiadhesion therapy including PECAM1, IL8, and CCL20, besides the currently studied α4ß7 integrin-MADCAM1 axis. CONCLUSIONS: Our data demonstrate that many leukocyte/endothelial CAMs and chemokines/chemokine receptors are upregulated in inflamed IBD mucosa. Controlling the inflammation with infliximab restores most of these dysregulations in IBD. These results show that at least part of the mechanism of anti-tumor necrosis factor-α therapy goes through downregulation of certain adhesion molecules.


Assuntos
Anti-Inflamatórios/uso terapêutico , Anticorpos Monoclonais/uso terapêutico , Moléculas de Adesão Celular/metabolismo , Quimiocinas/metabolismo , Doenças Inflamatórias Intestinais/metabolismo , Mucosa Intestinal/metabolismo , Receptores de Quimiocinas/metabolismo , Adulto , Western Blotting , Colite/metabolismo , Colite Ulcerativa/metabolismo , Colo/efeitos dos fármacos , Colo/metabolismo , Doença de Crohn/metabolismo , Regulação para Baixo , Feminino , Expressão Gênica/efeitos dos fármacos , Humanos , Ileíte/metabolismo , Íleo/efeitos dos fármacos , Íleo/metabolismo , Imuno-Histoquímica , Doenças Inflamatórias Intestinais/tratamento farmacológico , Infliximab , Mucosa Intestinal/efeitos dos fármacos , Masculino , Análise em Microsséries , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Regulação para Cima , Adulto Jovem
6.
PLoS One ; 12(8): e0181651, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28792951

RESUMO

The specific phenotype of mature differentiated beta cells not only depends on the specific presence of genes that allow beta cell function but also on the selective absence of housekeeping genes ("disallowed genes") that would interfere with this function. Recent studies have shown that both histone modifications and DNA methylation via the de novo methyltransferase DNMT3A are involved in repression of disallowed genes in neonatal beta cells when these cells acquire their mature phenotype. It is unknown, however, if the environmental influence of advanced age, pregnancy and the metabolic stress of high fat diet or diabetes could alter the repression of disallowed genes in beta cells. In the present study, we show that islet disallowed genes-which are also deeply repressed in FACS-purified beta cells-remain deeply repressed in animals of advanced age and in pregnant females. Moreover, the stability of this repression was correlated with strong and stable histone repression marks that persisted in islets isolated from 2 year old mice and with overall high expression of Dnmt3a in islets. Furthermore, repression of disallowed genes was unaffected by the metabolic stress of high fat diet. However, repression of about half of the disallowed genes was weakened in 16 week-old diabetic db/db mice. In conclusion, we show that the disallowed status of islet genes is stable under physiological challenging conditions (advanced age, pregnancy, high fat diet) but partially lost in islets from diabetic animals.


Assuntos
Envelhecimento/fisiologia , Metilação de DNA/genética , Diabetes Mellitus/metabolismo , Dieta Hiperlipídica , Código das Histonas/genética , Células Secretoras de Insulina/metabolismo , Estresse Fisiológico/fisiologia , Animais , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA Metiltransferase 3A , Feminino , Teste de Tolerância a Glucose , Insulina/metabolismo , Células Secretoras de Insulina/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Gravidez
7.
PLoS One ; 10(3): e0121868, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25816302

RESUMO

Pregnancy requires a higher functional beta cell mass and this is associated with profound changes in the gene expression profile of pancreatic islets. Taking Tph1 as a sensitive marker for pregnancy-related islet mRNA expression in female mice, we previously identified prolactin receptors and placental lactogen as key signalling molecules. Since beta cells from male mice also express prolactin receptors, the question arose whether male and female islets have the same phenotypic resilience at the mRNA level during pregnancy. We addressed this question in vitro, by stimulating cultured islets with placental lactogen and in vivo, by transplanting male or female islets into female acceptor mice. Additionally, the islet mRNA expression pattern of pregnant prolactin receptor deficient mice was compared with that of their pregnant wild-type littermates. When cultured with placental lactogen, or when transplanted in female recipients that became pregnant (day 12.5), male islets induced the 'islet pregnancy gene signature', which we defined as the 12 highest induced genes in non-transplanted female islets at day 12.5 of pregnancy. In addition, serotonin immunoreactivity and beta cell proliferation was also induced in these male transplanted islets at day 12.5 of pregnancy. In order to further investigate the importance of prolactin receptors in these mRNA changes we used a prolactin receptor deficient mouse model. For the 12 genes of the signature, which are highly induced in control pregnant mice, no significant induction of mRNA transcripts was found at day 9.5 of pregnancy. Together, our results support the key role of placental lactogen as a circulating factor that can trigger the pregnancy mRNA profile in both male and female beta cells.


Assuntos
Células Secretoras de Insulina/transplante , Lactogênio Placentário/sangue , Gravidez/genética , RNA Mensageiro/genética , Receptores da Prolactina/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Camundongos , Lactogênio Placentário/farmacologia , Gravidez/sangue , Receptores da Prolactina/genética
8.
PLoS One ; 10(8): e0135502, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26275221

RESUMO

The Nestin-Cre driver mouse line has mild hypopituitarism, reduced body weight, a metabolic phenotype and reduced anxiety. Although several causes have been suggested, a comprehensive explanation is still lacking. In this study we examined the molecular mechanisms leading to this compound phenotype. Upon generation of the Nestin-Cre mice, the human growth hormone (hGH) minigene was inserted downstream of the Cre recombinase to ensure efficient transgene expression. As a result, hGH is expressed in the hypothalamus. This results in the auto/paracrine activation of the GH receptor as demonstrated by the increased phosphorylation of signal transducer and activator of transcription 5 (STAT5) and reduced expression of growth hormone releasing hormone (Ghrh). Low Ghrh levels cause hypopituitarism consistent with the observed mouse growth hormone (mGH) deficiency. mGH deficiency caused reduced activation of the GH receptor and hence reduced phosphorylation of STAT5 in the liver. This led to decreased levels of hepatic Igf-1 mRNA and consequently postnatal growth retardation. Furthermore, genes involved in lipid uptake and synthesis, such as CD36 and very low-density lipoprotein receptor were upregulated, resulting in liver steatosis. In conclusion, this study demonstrates the unexpected expression of hGH in the hypothalamus of Nestin-Cre mice which is able to activate both the GH receptor and the prolactin receptor. Increased hypothalamic GH receptor signaling explains the observed hypopituitarism, reduced growth and metabolic phenotype of Nestin-Cre mice. Activation of either receptor is consistent with reduced anxiety.


Assuntos
Hormônio do Crescimento Humano/metabolismo , Hipotálamo/metabolismo , Animais , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Integrases/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Nestina/metabolismo , RNA Mensageiro/genética , Receptores de LDL/metabolismo , Fator de Transcrição STAT5/metabolismo
9.
Inflamm Bowel Dis ; 21(11): 2673-82, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26313692

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) is characterized by a chronic inflammation of the gut, partly driven by defects in the innate immune system. Considering the central role of inflammasome signaling in innate immunity, we studied inflammasome components in IBD mucosa. METHODS: Expression of genes encoding inflammasome sensor subunits was investigated in colonic mucosal biopsies from 2 cohorts of patients with IBD and controls. RESULTS: A significant upregulation (>2-fold change in expression, false discovery rate <0.05) of the PYHIN inflammasomes AIM2 and IFI16 in active IBD versus controls was found. Also IFI16 was significantly increased in inactive IBD versus controls. Moreover, responders to anti-tumor necrosis factor therapy showed decreased expression of these inflammasomes although IFI16 remained significantly increased in responders showing endoscopic healing versus controls. AIM2 was mainly expressed in epithelial cells, whereas IFI16 was expressed in both lymphocytes and epithelial cells. Functional activation of predominant AIM2/IFI16-mediated inflammasomes in active IBD colon was shown by the presence of the downstream effectors CASP1 and HMGB-1 in inflamed mucosa. CONCLUSIONS: Our results highlight the importance of PYHIN inflammasome signaling in IBD and also link anti-tumor necrosis factor responsiveness to inflammasome signaling. Together, this points to the potential value of the inflammasome pathway as a new therapeutic target for IBD treatment.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Inflamassomos/genética , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/patologia , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais , Adulto , Idoso , Biópsia , Estudos de Casos e Controles , Caspase 1/metabolismo , Estudos de Coortes , Proteínas de Ligação a DNA/genética , Células Epiteliais/metabolismo , Feminino , Proteína HMGB1/metabolismo , Humanos , Imunidade Inata , Masculino , Pessoa de Meia-Idade , Proteínas Nucleares/genética , Fosfoproteínas/genética , Ativação Transcricional , Regulação para Cima
10.
Cell Metab ; 20(6): 979-90, 2014 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-25470546

RESUMO

The human growth hormone (hGH) minigene is frequently used in the derivation of transgenic mouse lines to enhance transgene expression. Although this minigene is present in the transgenes as a secondcistron, and thus not thought to be expressed, we found that three commonly used lines, Pdx1-Cre(Late), RIP-Cre, and MIP-GFP, each expressed significant amounts of hGH in pancreatic islets. Locally secreted hGH binds to prolactin receptors on ß cells, activates STAT5 signaling, and induces pregnancy-like changes in gene expression, thereby augmenting pancreatic ß cell mass and insulin content. In addition, islets of Pdx1-Cre(Late) mice have lower GLUT2 expression and reduced glucose-induced insulin release and are protected against the ß cell toxin streptozotocin. These findings may be important when interpreting results obtained when these and other hGH minigene-containing transgenic mice are used.


Assuntos
Hormônio do Crescimento Humano/metabolismo , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Animais , Feminino , Hormônio do Crescimento Humano/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Receptores da Prolactina/genética , Receptores da Prolactina/metabolismo , Triptofano Hidroxilase/genética , Triptofano Hidroxilase/metabolismo
11.
PLoS One ; 8(2): e56719, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23460811

RESUMO

Oxidative phosphorylation in mitochondria is responsible for 90% of ATP synthesis in most cells. This essential housekeeping function is mediated by nuclear and mitochondrial genes encoding subunits of complex I to V of the respiratory chain. Although complex IV is the best studied of these complexes, the exact function of the striated muscle-specific subunit COX6A2 is still poorly understood. In this study, we show that Cox6a2-deficient mice are protected against high-fat diet-induced obesity, insulin resistance and glucose intolerance. This phenotype results from elevated energy expenditure and a skeletal muscle fiber type switch towards more oxidative fibers. At the molecular level we observe increased formation of reactive oxygen species, constitutive activation of AMP-activated protein kinase, and enhanced expression of uncoupling proteins. Our data indicate that COX6A2 is a regulator of respiratory uncoupling in muscle and we demonstrate that a novel and direct link exists between muscle respiratory chain activity and diet-induced obesity/insulin resistance.


Assuntos
Dieta Hiperlipídica , Complexo IV da Cadeia de Transporte de Elétrons/genética , Resistência à Insulina/genética , Proteínas Musculares/genética , Obesidade/genética , Obesidade/prevenção & controle , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Transporte de Elétrons/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Metabolismo Energético/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Teste de Tolerância a Glucose , Técnicas In Vitro , Insulina/farmacologia , Canais Iônicos/metabolismo , Camundongos , Proteínas Mitocondriais/metabolismo , Tamanho Mitocondrial/efeitos dos fármacos , Modelos Biológicos , Fadiga Muscular/efeitos dos fármacos , Proteínas Musculares/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Espécies Reativas de Oxigênio/metabolismo , Inanição/patologia , Termogênese/efeitos dos fármacos , Magreza/metabolismo , Proteína Desacopladora 1
12.
Cell Transplant ; 20(8): 1285-97, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21294959

RESUMO

Pleomorphic adenoma gene-like 1 (PLAGL1) has been linked to transient neonatal diabetes mellitus. Here, we investigated the role of the related pleomorphic adenoma gene 1 (PLAG1) in glucose homeostasis. PLAG1 transgenic mice in which expression of the PLAG1 transgene can be targeted to different organs by Cre-mediated modulation were crossed with Pdx1-Cre or Ngn3-Cre mice, resulting in double transgenic P1-Pdx1Cre or P1-Ngn3Cre mice, respectively. P1-Pdx1Cre and P1-Ngn3Cre mice developed hyperplasia of pancreatic islets due to increased ß- and δ- but not α-cell proliferation. In young P1-Pdx1Cre mice (less than 15 weeks) there was a balanced increase in the pancreatic content of insulin and somatostatin, which was associated with normoglycemia. In older P1-Pdx1Cre mice the pancreatic somatostatin content far exceeded that of insulin, leading to the progressive development of severe hypoglycemia beyond 30 weeks. In contrast, in older P1-Ngn3Cre mice the relative increase of the pancreatic insulin content exceeded that of somatostatin and these mice remained normoglycemic. In conclusion, forced expression of PLAG1 under the control of the Pdx1 or Ngn3 promoter in murine pancreas induces different degrees of endocrine hormone imbalances within the pancreas, which is associated with hypoglycemia in P1-Pdx1Cre mice but not P1-Ngn3Cre mice. These results suggest that once stem cell-derived islet transplantations become possible, the appropriate balance between different hormone-producing cells will need to be preserved to prevent deregulated glucose metabolism.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Glucose/metabolismo , Proteínas de Homeodomínio/metabolismo , Integrases/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Hormônios Pancreáticos/metabolismo , Transativadores/metabolismo , Animais , Proliferação de Células , Glucagon , Teste de Tolerância a Glucose , Homeostase , Hiperplasia , Hipoglicemia/patologia , Insulina , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/ultraestrutura , Transplante das Ilhotas Pancreáticas , Camundongos , Camundongos Transgênicos , Tamanho do Órgão , Somatostatina , Fatores de Tempo
13.
Inflamm Bowel Dis ; 16(12): 2090-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20848504

RESUMO

BACKGROUND: Infliximab (IFX) has become the mainstay of therapy of refractory Crohn's disease (CD). However, a subset of patients shows incomplete or no response to this agent. In this study we investigated whether we could identify a mucosal gene panel to predict (non)response to IFX in CD. METHODS: Mucosal biopsies were obtained during endoscopy from 37 patients with active CD (19 Crohn's colitis [CDc] and 18 Crohn's ileitis [CDi]) before and after first IFX treatment. Response was defined based on endoscopic and histologic findings. Total RNA was analyzed with Affymetrix Human Genome U133 Plus 2.0 Arrays. Quantitative real-time reverse-transcription polymerase chain reaction (RT-PCR) was used to confirm microarray data. RESULTS: At baseline, significant gene expression differences were found between CDc and CDi. For predicting response in CDc, comparative analysis of CDc pretreatment expression profiles identified 697 significant probe sets between CDc responders (n = 12) and CDc nonresponders (n = 7). Class prediction analysis of CDc top 20 and top 5 significant genes allowed complete separation between CDc responders and CDc nonresponders. The CDc top 5 genes were TNFAIP6, S100A8, IL11, G0S2, and S100A9. Only one patient with CDi completely healed the ileal mucosa. Even using less stringent response criteria, we could not identify a predictive gene panel for IFX responsiveness in CDi. CONCLUSIONS: This study identified a 100% accurate predictive gene signature for (non)response to IFX in CDc, whereas no such a predictive gene set could be identified for CDi.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Biomarcadores/metabolismo , Doença de Crohn/tratamento farmacológico , Doença de Crohn/genética , Fármacos Gastrointestinais/uso terapêutico , Perfilação da Expressão Gênica , Adulto , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/genética , Colite Ulcerativa/patologia , Doença de Crohn/patologia , Feminino , Humanos , Ileíte/tratamento farmacológico , Ileíte/genética , Ileíte/patologia , Infliximab , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Masculino , Pessoa de Meia-Idade , Mucosa/efeitos dos fármacos , Mucosa/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Terapia de Salvação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA