Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell Mol Gastroenterol Hepatol ; 13(4): 1095-1120, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35017061

RESUMO

BACKGROUND AND AIMS: Phagocytosis (efferocytosis) of apoptotic neutrophils by macrophages anchors the resolution of intestinal inflammation. Efferocytosis prevents secondary necrosis and inhibits further inflammation, and also reprograms macrophages to facilitate tissue repair and promote resolution function. Macrophage efferocytosis and efferocytosis-dependent reprogramming are implicated in the pathogenesis of inflammatory bowel disease. We previously reported that absence of macrophage cyclooxygenase 2 (COX2) exacerbates inflammatory bowel disease-like intestinal inflammation. To elucidate the underlying pathogenic mechanism, we investigated here whether COX2 mediates macrophage efferocytosis and efferocytosis-dependent reprogramming, including intestinal epithelial repair capacity. METHODS: Using apoptotic neutrophils and synthetic apoptotic targets, we determined the effects of macrophage specific Cox2 knockout and pharmacological COX2 inhibition on the efferocytosis capacity of mouse primary macrophages. COX2-mediated efferocytosis-dependent eicosanoid lipidomics was determined by liquid chromatography tandem mass spectrometry. Small intestinal epithelial organoids were employed to assay the effects of COX2 on efferocytosis-dependent intestinal epithelial repair. RESULTS: Loss of COX2 impaired efferocytosis in mouse primary macrophages, in part, by affecting the binding capacity of macrophages for apoptotic cells. This effect was comparable to that of high-dose lipopolysaccharide and was accompanied by both dysregulation of macrophage polarization and the inhibited expression of genes involved in apoptotic cell binding. COX2 modulated the production of efferocytosis-dependent lipid inflammatory mediators that include the eicosanoids prostaglandin I2, prostaglandin E2, lipoxin A4, and 15d-PGJ2; and further affected secondary efferocytosis. Finally, macrophage efferocytosis induced, in a macrophage COX2-dependent manner, a tissue restitution and repair phenotype in intestinal epithelial organoids. CONCLUSIONS: Macrophage COX2 potentiates efferocytosis capacity and efferocytosis-dependent reprogramming, facilitating macrophage intestinal epithelial repair capacity.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Doenças Inflamatórias Intestinais , Fagocitose , Animais , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/farmacologia , Inflamação/patologia , Doenças Inflamatórias Intestinais/patologia , Macrófagos/metabolismo , Camundongos , Fagocitose/genética
2.
JCI Insight ; 5(1)2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31805014

RESUMO

Biallelic mutations of the gene encoding the transcription factor NEUROG3 are associated with a rare disorder that presents in neonates as generalized malabsorption - due to a complete absence of enteroendocrine cells - followed, in early childhood or beyond, by insulin-dependent diabetes mellitus (IDDM). The commonly delayed onset of IDDM suggests a differential requirement for NEUROG3 in endocrine cell generation in the human pancreas versus the intestine. However, previously identified human mutations were hypomorphic and, hence, may have had residual function in pancreas. We report 2 patients with biallelic functionally null variants of the NEUROG3 gene who nonetheless did not present with IDDM during infancy but instead developed permanent IDDM during middle childhood ages. The variants showed no evidence of function in traditional promoter-based assays of NEUROG3 function and also failed to exhibit function in a variety of potentially novel in vitro and in vivo molecular assays designed to discern residual NEUROG3 function. These findings imply that, unlike in mice, pancreatic endocrine cell generation in humans is not entirely dependent on NEUROG3 expression and, hence, suggest the presence of unidentified redundant in vivo pathways in human pancreas capable of yielding ß cell mass sufficient to maintain euglycemia until early childhood.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diabetes Mellitus/genética , Predisposição Genética para Doença , Mutação com Perda de Função , Proteínas do Tecido Nervoso/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proliferação de Células , Criança , Diabetes Mellitus Tipo 1 , Células Enteroendócrinas/metabolismo , Feminino , Regulação da Expressão Gênica , Sequências Hélice-Alça-Hélice/genética , Humanos , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas , Síndromes de Malabsorção , Masculino , Proteínas do Tecido Nervoso/metabolismo , Pâncreas , Regiões Promotoras Genéticas
3.
J Biol Chem ; 294(41): 15182-15192, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31341016

RESUMO

Neurogenin-3 (NEUROG3) is a helix-loop-helix (HLH) transcription factor involved in the production of endocrine cells in the intestine and pancreas of humans and mice. However, the human NEUROG3 loss-of-function phenotype differs subtly from that in mice, but the reason for this difference remains poorly understood. Because NEUROG3 expression precedes exit of the cell cycle and the expression of endocrine cell markers during differentiation, we investigated the effect of lentivirus-mediated overexpression of the human NEUROG3 gene on the cell cycle of BON4 cells and various human nonendocrine cell lines. NEUROG3 overexpression induced a reversible cell cycle exit, whereas expression of a neuronal lineage homolog, NEUROG1, had no such effect. In endocrine lineage cells, the cellular quiescence induced by short-term NEUROG3 expression required cyclin-dependent kinase inhibitor 1A (CDKN1A)/p21CIP1 expression. Expression of endocrine differentiation markers required sustained NEUROG3 expression in the quiescent, but not in the senescent, state. Inhibition of the phosphatase and tensin homolog (PTEN) pathway reversed quiescence by inducing cyclin-dependent kinase 2 (CDK2) and reducing p21CIP1 and NEUROG3 protein levels in BON4 cells and human enteroids. We discovered that NEUROG3 expression stimulates expression of CDKN2a/p16INK4a and BMI1 proto-oncogene polycomb ring finger (BMI1), with the latter limiting expression of the former, delaying the onset of CDKN2a/p16INK4a -driven cellular senescence. Furthermore, NEUROG3 bound to the promoters of both CDKN1a/p21CIP1 and BMI1 genes, and BMI1 attenuated NEUROG3 binding to the CDKN1a/p21CIP1 promoter. Our findings reveal how human NEUROG3 integrates inputs from multiple signaling pathways and thereby mediates cell cycle exit at the onset of differentiation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Pontos de Checagem do Ciclo Celular , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Proteínas do Tecido Nervoso/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Linhagem Celular , Senescência Celular , Regulação da Expressão Gênica , Genes p16 , Humanos , Proto-Oncogene Mas
4.
J Clin Invest ; 129(9): 3670-3685, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31184596

RESUMO

Cyclooxygenase 2 (Cox2) total knockout and myeloid knockout (MKO) mice develop Crohn's-like intestinal inflammation when fed cholate-containing high fat diet (CCHF). We demonstrated that CCHF impaired intestinal barrier function and increased translocation of endotoxin, initiating TLR/MyD88-dependent inflammation in Cox2 KO but not WT mice. Cox2 MKO increased pro-inflammatory mediators in LPS-activated macrophages, and in the intestinal tissue and plasma upon CCHF challenge. Cox2 MKO also reduced inflammation resolving lipoxin A4 (LXA4) in intestinal tissue, while administration of an LXA4 analog rescued disease in Cox2 MKO mice fed CCHF. The apolipoprotein A-I (APOA1) mimetic 4F mitigated disease in both the Cox2 MKO/CCHF and piroxicam-accelerated Il10-/- models of inflammatory bowel disease (IBD) and reduced elevated levels of pro-inflammatory mediators in tissue and plasma. APOA1 mimetic Tg6F therapy was also effective in reducing intestinal inflammation in the Cox2 MKO/CCHF model. We further demonstrated that APOA1 mimetic peptides: i) inhibited LPS and oxidized 1-palmitoyl-2-arachidonoyl-sn-phosphatidylcholine (oxPAPC) dependent pro-inflammatory responses in human macrophages and intestinal epithelium; and ii) directly cleared pro-inflammatory lipids from mouse intestinal tissue and plasma. Our results support a causal role for pro-inflammatory and inflammation resolving lipids in IBD pathology and a translational potential for APOA1 mimetic peptides for the treatment of IBD.


Assuntos
Apolipoproteína A-I/farmacologia , Ciclo-Oxigenase 2/genética , Doenças Inflamatórias Intestinais/tratamento farmacológico , Intestinos/patologia , Animais , Modelos Animais de Doenças , Endotoxinas/metabolismo , Feminino , Humanos , Doenças Inflamatórias Intestinais/patologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxigênio/metabolismo , Peptídeos/química , Permeabilidade , Piroxicam/farmacologia , Receptores de Formil Peptídeo/metabolismo , Transdução de Sinais
5.
PLoS One ; 13(5): e0195315, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29718926

RESUMO

Although critical for studies of gut motility and intestinal regeneration, the in vitro culture of intestinal muscularis with peristaltic function remains a significant challenge. Periodic contractions of intestinal muscularis result from the coordinated activity of smooth muscle cells (SMC), the enteric nervous system (ENS), and interstitial cells of Cajal (ICC). Reproducing this activity requires the preservation of all these cells in one system. Here we report the first serum-free culture methodology that consistently maintains spontaneous and periodic contractions of murine and human intestinal muscularis cells for months. In this system, SMC expressed the mature marker myosin heavy chain, and multipolar/dipolar ICC, uniaxonal/multipolar neurons and glial cells were present. Furthermore, drugs affecting neural signals, ICC or SMC altered the contractions. Combining this method with scaffolds, contracting cell sheets were formed with organized architecture. With the addition of intestinal epithelial cells, this platform enabled up to 11 types of cells from mucosa, muscularis and serosa to coexist and epithelial cells were stretched by the contracting muscularis cells. The method constitutes a powerful tool for mechanistic studies of gut motility disorders and the functional regeneration of the engineered intestine.


Assuntos
Bioengenharia , Contração Muscular , Músculo Liso/fisiologia , Animais , Linhagem Celular , Técnicas de Cocultura , Humanos , Lactente , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso/citologia , Neuroglia/citologia , Neurônios/citologia , Fatores de Tempo
6.
PLoS One ; 11(1): e0148216, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26820624

RESUMO

BACKGROUND & AIMS: Intestinal microfold (M) cells are specialized epithelial cells that act as gatekeepers of luminal antigens in the intestinal tract. They play a critical role in the intestinal mucosal immune response through transport of viruses, bacteria and other particles and antigens across the epithelium to immune cells within Peyer's patch regions and other mucosal sites. Recent studies in mice have demonstrated that M cells are generated from Lgr5+ intestinal stem cells (ISCs), and that infection with Salmonella enterica serovar Typhimurium increases M cell formation. However, it is not known whether and how these findings apply to primary human small intestinal epithelium propagated in an in vitro setting. METHODS: Human intestinal crypts were grown as monolayers with growth factors and treated with recombinant RANKL, and assessed for mRNA transcripts, immunofluorescence and uptake of microparticles and S. Typhimurium. RESULTS: Functional M cells were generated by short-term culture of freshly isolated human intestinal crypts in a dose- and time-dependent fashion. RANKL stimulation of the monolayer cultures caused dramatic induction of the M cell-specific markers, SPIB, and Glycoprotein-2 (GP2) in a process primed by canonical WNT signaling. Confocal microscopy demonstrated a pseudopod phenotype of GP2-positive M cells that preferentially take up microparticles. Furthermore, infection of the M cell-enriched cultures with the M cell-tropic enteric pathogen, S. Typhimurium, led to preferential association of the bacteria with M cells, particularly at lower inoculum sizes. Larger inocula caused rapid induction of M cells. CONCLUSIONS: Human intestinal crypts containing ISCs can be cultured and differentiate into an epithelial layer with functional M cells with characteristic morphological and functional properties. This study is the first to demonstrate that M cells can be induced to form from primary human intestinal epithelium, and that S. Typhimurium preferentially infect these cells in an in vitro setting. We anticipate that this model can be used to generate large numbers of M cells for further functional studies of these key cells of intestinal immune induction and their impact on controlling enteric pathogens and the intestinal microbiome.


Assuntos
Mucosa Intestinal/citologia , Intestino Delgado/citologia , Nódulos Linfáticos Agregados/citologia , Células-Tronco/citologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células Cultivadas , Humanos , Imunidade nas Mucosas , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/microbiologia , Ligante RANK/imunologia , Salmonella typhimurium/imunologia , Células-Tronco/imunologia
7.
J Pediatr Gastroenterol Nutr ; 57(6): 759-67, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24280991

RESUMO

OBJECTIVES: Congenital diarrhea disorders are a group of genetically diverse and typically autosomal recessive disorders that have yet to be well characterized phenotypically or molecularly. Diagnostic assessments are generally limited to nutritional challenges and histologic evaluation, and many subjects eventually require a prolonged course of intravenous nutrition. Here we describe next-generation sequencing techniques to investigate a child with perplexing congenital malabsorptive diarrhea and other presumably unrelated clinical problems; this method provides an alternative approach to molecular diagnosis. METHODS: We screened the diploid genome of an affected individual, using exome sequencing, for uncommon variants that have observed protein-coding consequences. We assessed the functional activity of the mutant protein, as well as its lack of expression using immunohistochemistry. RESULTS: Among several rare variants detected was a homozygous nonsense mutation in the catalytic domain of the proprotein convertase subtilisin/kexin type 1 gene. The mutation abolishes prohormone convertase 1/3 endoprotease activity as well as expression in the intestine. These primary genetic findings prompted a careful endocrine reevaluation of the child at 4.5 years of age, and multiple significant problems were subsequently identified consistent with the known phenotypic consequences of proprotein convertase subtilisin/kexin type 1 (PCSK1) gene mutations. Based on the molecular diagnosis, alternate medical and dietary management was implemented for diabetes insipidus, polyphagia, and micropenis. CONCLUSIONS: Whole-exome sequencing provides a powerful diagnostic tool to clinicians managing rare genetic disorders with multiple perplexing clinical manifestations.


Assuntos
Códon sem Sentido , Diabetes Insípido/complicações , Diarreia/genética , Exoma , Síndromes de Malabsorção/genética , Pró-Proteína Convertase 1/genética , Diarreia/congênito , Diarreia/diagnóstico , Homozigoto , Humanos , Recém-Nascido , Síndromes de Malabsorção/congênito , Síndromes de Malabsorção/diagnóstico , Masculino , Pró-Proteína Convertase 1/metabolismo , Análise de Sequência de DNA
8.
Gastroenterology ; 145(1): 138-148, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23562752

RESUMO

BACKGROUND & AIMS: Proprotein convertase 1/3 (PC1/3) deficiency, an autosomal-recessive disorder caused by rare mutations in the proprotein convertase subtilisin/kexin type 1 (PCSK1) gene, has been associated with obesity, severe malabsorptive diarrhea, and certain endocrine abnormalities. Common variants in PCSK1 also have been associated with obesity in heterozygotes in several population-based studies. PC1/3 is an endoprotease that processes many prohormones expressed in endocrine and neuronal cells. We investigated clinical and molecular features of PC1/3 deficiency. METHODS: We studied the clinical features of 13 children with PC1/3 deficiency and performed sequence analysis of PCSK1. We measured enzymatic activity of recombinant PC1/3 proteins. RESULTS: We identified a pattern of endocrinopathies that develop in an age-dependent manner. Eight of the mutations had severe biochemical consequences in vitro. Neonates had severe malabsorptive diarrhea and failure to thrive, required prolonged parenteral nutrition support, and had high mortality. Additional endocrine abnormalities developed as the disease progressed, including diabetes insipidus, growth hormone deficiency, primary hypogonadism, adrenal insufficiency, and hypothyroidism. We identified growth hormone deficiency, central diabetes insipidus, and male hypogonadism as new features of PCSK1 insufficiency. Interestingly, despite early growth abnormalities, moderate obesity, associated with severe polyphagia, generally appears. CONCLUSIONS: In a study of 13 children with PC1/3 deficiency caused by disruption of PCSK1, failure of enteroendocrine cells to produce functional hormones resulted in generalized malabsorption. These findings indicate that PC1/3 is involved in the processing of one or more enteric hormones that are required for nutrient absorption.


Assuntos
Diarreia/etiologia , Doenças do Sistema Endócrino/etiologia , Síndromes de Malabsorção/etiologia , Obesidade/complicações , Pró-Proteína Convertase 1/deficiência , Adolescente , Hormônio Adrenocorticotrópico/sangue , Criança , Pré-Escolar , Estudos de Coortes , Doenças do Sistema Endócrino/complicações , Doenças do Sistema Endócrino/congênito , Feminino , Humanos , Lactente , Masculino , Mutação , Obesidade/congênito , Pró-Proteína Convertase 1/genética
9.
PLoS One ; 8(1): e55065, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23383060

RESUMO

BACKGROUND: Common single nucleotide polymorphisms (SNPs) in proprotein convertase subtilisin/kexin type 1 with modest effects on PC1/3 in vitro have been associated with obesity in five genome-wide association studies and with diabetes in one genome-wide association study. We here present a novel SNP and compare its biosynthesis, secretion and catalytic activity to wild-type enzyme and to SNPs that have been linked to obesity. METHODOLOGY/PRINCIPAL FINDINGS: A novel PC1/3 variant introducing an Arg to Gln amino acid substitution at residue 80 (within the secondary cleavage site of the prodomain) (rs1799904) was studied. This novel variant was selected for analysis from the 1000 Genomes sequencing project based on its predicted deleterious effect on enzyme function and its comparatively more frequent allele frequency. The actual existence of the R80Q (rs1799904) variant was verified by Sanger sequencing. The effects of this novel variant on the biosynthesis, secretion, and catalytic activity were determined; the previously-described obesity risk SNPs N221D (rs6232), Q665E/S690T (rs6234/rs6235), and the Q665E and S690T SNPs (analyzed separately) were included for comparative purposes. The novel R80Q (rs1799904) variant described in this study resulted in significantly detrimental effects on both the maturation and in vitro catalytic activity of PC1/3. CONCLUSION/SIGNIFICANCE: Our findings that this novel R80Q (rs1799904) variant both exhibits adverse effects on PC1/3 activity and is prevalent in the population suggests that further biochemical and genetic analysis to assess its contribution to the risk of metabolic disease within the general population is warranted.


Assuntos
Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Polimorfismo de Nucleotídeo Único , Biocatálise , Bases de Dados Genéticas , Diabetes Mellitus/genética , Feminino , Humanos , Neuropeptídeos/biossíntese , Obesidade/genética , Análise de Sequência de DNA , Relação Estrutura-Atividade
10.
Mol Genet Metab ; 85(4): 301-7, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15905112

RESUMO

In human cells, biotin is essential to maintain metabolic homeostasis and as regulator of gene expression. The enzyme holocarboxylase synthetase (HCS) transforms biotin into its active form 5'-biotinyl-AMP and this compound is used to biotinylate five biotin-dependent carboxylases or to activate a soluble guanylate cyclase (sGC) and a cGMP-dependent protein kinase (PKG). The HCS-sGC-PKG pathway is responsible for maintaining the mRNA levels of enzymes involved in biotin utilization including HCS, carboxylases, and a biotin carrier known as sodium-dependent multivitamin transporter (SMVT). To understand the role of SMVT in the control of biotin utilization, we have studied the effect of biotin availability on SMVT protein and mRNA expression levels in HepG2 cells by Western blot analysis and rtPCR, respectively; and their functional impact on the rate of [3H]biotin uptake in human cells. Our results showed that human HepG2 cells grown in a biotin-deficient medium have a lower rate of biotin uptake than normal cells. The impairment in biotin uptake is associated with a reduction in the amount of both SMVT protein mass and mRNA levels. Transfection of HepG2 cells with a vector containing a luciferase reporter gene under the control of the rat SMVT promoter demonstrated that its transcriptional activity is regulated by biotin availability through activation of the HCS-sGC-PKG pathway. Our results support the proposed role of SMVT in the altruistic regulation of biotin utilization in liver cells that has been associated with sparing biotin depletion of the brain.


Assuntos
Biotina/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Fígado/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Simportadores/metabolismo , Ativação Transcricional , Animais , Biotina/deficiência , Biotina/genética , Biotinilação , Western Blotting , Técnicas de Cultura de Células , Proteínas Quinases Dependentes de GMP Cíclico/genética , Transportador de Glucose Tipo 1 , Humanos , Proteínas de Membrana Transportadoras , Proteínas de Transporte de Monossacarídeos/genética , RNA Mensageiro , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Simportadores/genética
11.
J Biol Chem ; 279(50): 52312-8, 2004 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-15456772

RESUMO

Holocarboxylase synthetase (HCS) catalyzes the biotinylation of five carboxylases in human cells, and mutations of HCS cause multiple carboxylase deficiency (MCD). Although HCS also participates in the regulation of its own mRNA levels, the relevance of this mechanism to normal metabolism or to the MCD phenotype is not known. In this study, we show that mRNA levels of enzymes involved in biotin utilization, including HCS, are down-regulated during biotin deficiency in liver while remaining constitutively expressed in brain. We propose that this mechanism of regulation is aimed at sparing the essential function of biotin in the brain at the expense of organs such as liver and kidney during biotin deprivation. In MCD, it is possible that some of the manifestations of the disease may be associated with down-regulation of biotin utilization in liver because of the impaired activity of HCS and that high dose biotin therapy may in part be important to overcoming the adverse regulatory impact in such organs.


Assuntos
Biotina/metabolismo , Encéfalo/metabolismo , Fígado/metabolismo , Deficiência Múltipla de Carboxilase/genética , Deficiência Múltipla de Carboxilase/metabolismo , Animais , Sequência de Bases , Biotina/deficiência , Carbono-Nitrogênio Ligases/genética , Carbono-Nitrogênio Ligases/metabolismo , Linhagem Celular , DNA Complementar/genética , Deficiência de Holocarboxilase Sintetase/genética , Deficiência de Holocarboxilase Sintetase/metabolismo , Humanos , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Distribuição Tecidual
12.
Am J Physiol Gastrointest Liver Physiol ; 286(6): G922-31, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15132949

RESUMO

The regulatory elements that control the transcriptional regulation of the intestinal Fc receptor (FcRn) have not been elucidated. The objective of this study was to characterize the core promoter region of the rat FcRn gene. Chimeric clones that contained various regions of the promoter located upstream of the luciferase reporter were transiently transfected into either IEC-6 or Caco-2 cell lines and nuclear extracts were used to perform DNase I footprint and DNA binding assays (EMSA). Transfection of chimeric upstream nested deletions-luciferase reporter clones into either of these cell lines supported robust reporter activity and identified the location of the minimal promoter at -157/+135. DNase I footprint analysis revealed two complexes located within the gene's core promoter region, and site-directed mutagenesis identified two regions that were critical to maintain basal expression. EMSA identified the presence of five Sp elements within the immediate promoter region that are capable of binding members of the Sp family of proteins. Among the five Sp elements, one element appears to not bind Sp1, Sp2, or Sp3 while influencing the interaction of Sp proteins with an adjacent Sp site. Overexpression of either Sp1 or Sp3 augments activity of the minimal promoter in Sp-deficient Drosophila SL2 cells. In summary, we report on the characterization of the rat FcRn minimal promoter, including the characterization of five Sp elements within this region that interact with members of the Sp family of transcriptional factors and drive promoter activity in intestinal cell lines.


Assuntos
Mucosa Intestinal/metabolismo , Família Multigênica , Regiões Promotoras Genéticas/genética , Receptores Fc/genética , Fatores de Transcrição/fisiologia , Transcrição Gênica , Animais , Sequência de Bases , Linhagem Celular , Células Clonais , Drosophila , Eletroforese , Regulação da Expressão Gênica , Homeostase , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Estereoisomerismo , Fatores de Transcrição/genética , Transfecção
13.
Arch Med Res ; 33(5): 439-47, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12459313

RESUMO

Biotin, a water-soluble vitamin, is used as cofactor of enzymes involved in carboxylation reactions. In humans, there are five biotin-dependent carboxylases: propionyl-CoA carboxylase; methylcrotonyl-CoA carboxylase; pyruvate carboxylase, and two forms of acetyl-CoA carboxylase. These enzymes catalyze key reactions in gluconeogenesis, fatty acid metabolism, and amino acid catabolism; thus, biotin plays an essential role in maintaining metabolic homeostasis. In recent years, biotin has been associated with several diseases in humans. Some are related to enzyme deficiencies involved in biotin metabolism. However, not all biotin-responsive disorders can be explained based on the classical role of the vitamin in cell metabolism. Several groups have suggested that biotin may be involved in regulating transcription or protein expression of different proteins. Biotinylation of histones and triggering of transduction signaling cascades have been suggested as underlying mechanisms behind these non-classical biotin-deficiency manifestation in humans.


Assuntos
Biotina/metabolismo , Biotina/fisiologia , Sequência de Aminoácidos , Carbono-Nitrogênio Ligases/metabolismo , Catálise , Feminino , Regulação Enzimológica da Expressão Gênica , Histonas/metabolismo , Humanos , Masculino , Modelos Biológicos , Modelos Químicos , Dados de Sequência Molecular , Deficiência Múltipla de Carboxilase/genética , Gravidez , Transdução de Sinais , Transcrição Gênica
14.
Am J Physiol Gastrointest Liver Physiol ; 283(2): G415-25, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12121890

RESUMO

The polymeric Ig receptor (pIgR) is a critical component of the mucosal immune system and is expressed in largest amounts in the small intestine. In this study, we describe the initial characterization of the core promoter region of this gene. Expression of chimeric promoter-reporter constructs was supported in Caco-2 and HT-29 cells, and DNase I footprint analysis revealed a large protein complex within the core promoter region. Site-directed mutagenesis experiments determined that elements within this region serve to either augment or repress basal activity of the human pIgR promoter. Band shift assays of overlapping oligonucleotides within the core promoter identified eight distinct complexes; the abundance of most complexes was enhanced in post-confluent cells. In summary, we report the characterization of the human pIgR promoter and the essential role that eight different nuclear complexes have in controlling basal expression of this gene in Caco-2 cells.


Assuntos
Receptores de Imunoglobulina Polimérica/genética , Receptores de Imunoglobulina Polimérica/metabolismo , Fatores de Transcrição/fisiologia , Animais , Sequência de Bases/genética , Células CHO , Células CACO-2 , Clonagem Molecular , Cricetinae , Pegada de DNA , Eletroforese , Genes Reporter , Células HT29 , Humanos , Dados de Sequência Molecular , Mutação , Regiões Promotoras Genéticas/genética , Transfecção
15.
Proc Natl Acad Sci U S A ; 99(8): 5325-30, 2002 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-11959985

RESUMO

Holocarboxylase synthetase (HCS) catalyzes the covalent attachment of biotin to five biotin-dependent carboxylases in human cells. Multiple carboxylase deficiency (MCD) is a life-threatening disease characterized by the lack of carboxylase activities because of deficiency of HCS activity. Here, we report the obligatory participation of HCS in the biotin-dependent stimulation of the level of HCS mRNA and those of acetyl-CoA carboxylase and the alpha subunit of propionyl-CoA carboxylase in human cells. Fibroblasts from patients with MCD are unable to increase HCS mRNA in response to biotin unless the vitamin concentration is raised 100-fold, in keeping with mutations that cause a reduced affinity for biotin by the mutant enzyme. The outcome is deficient synthesis of biotinyl-5'-AMP, the active form of the vitamin in the biotinylation reaction. HCS and carboxylase mRNA levels in normal and MCD fibroblasts and HepG2 cells can be restored by the addition of the cGMP analogue, 8-Br-cGMP, and can be abolished by the addition of inhibitors of the soluble form of guanylate cyclase. We propose a regulatory role for biotin in the control of HCS and carboxylase mRNA levels through a signaling cascade that requires HCS, guanylate cyclase, and cGMP-dependent protein kinase.


Assuntos
Biotina/farmacologia , Carbono-Carbono Ligases/metabolismo , Carbono-Nitrogênio Ligases/química , Carbono-Nitrogênio Ligases/metabolismo , GMP Cíclico/análogos & derivados , RNA Mensageiro/metabolismo , Linhagem Celular , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Dactinomicina/metabolismo , Densitometria , Relação Dose-Resposta a Droga , Fibroblastos/metabolismo , Guanilato Ciclase/metabolismo , Humanos , Mutação , Ligação Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Tempo , Vitaminas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA