Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Placenta ; 54: 45-51, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28089504

RESUMEN

Nutrient sulfate has numerous roles in mammalian physiology and is essential for healthy fetal growth and development. The fetus has limited capacity to generate sulfate and relies on sulfate supplied from the maternal circulation via placental sulfate transporters. The placenta also has a high sulfate requirement for numerous molecular and cellular functions, including sulfate conjugation (sulfonation) to estrogen and thyroid hormone which leads to their inactivation. Accordingly, the ratio of sulfonated (inactive) to unconjugated (active) hormones modulates endocrine function in fetal, placental and maternal tissues. During pregnancy, there is a marked increase in the expression of genes involved in transport and generation of sulfate in the mouse placenta, in line with increasing fetal and placental demands for sulfate. The maternal circulation also provides a vital reservoir of sulfate for the placenta and fetus, with maternal circulating sulfate levels increasing by 2-fold from mid-gestation. However, despite evidence from animal studies showing the requirement of maternal sulfate supply for placental and fetal physiology, there are no routine clinical measurements of sulfate or consideration of dietary sulfate intake in pregnant women. This is also relevant to certain xenobiotics or pharmacological drugs which when taken by the mother use significant quantities of circulating sulfate for detoxification and clearance, and thereby have the potential to decrease sulfonation capacity in the placenta and fetus. This article will review the physiological adaptations of the placenta for maintaining sulfate homeostasis in the fetus and placenta, with a focus on pathophysiological outcomes in animal models of disturbed sulfate homeostasis.


Asunto(s)
Adaptación Fisiológica , Placenta/metabolismo , Sulfatos/metabolismo , Animales , Dieta , Femenino , Desarrollo Fetal , Humanos , Enfermedades Placentarias/metabolismo , Embarazo
2.
Artículo en Inglés | MEDLINE | ID: mdl-31565540

RESUMEN

OBJECTIVES: Breast feeding protects infants from many diseases, including necrotizing enterocolitis, peptic ulceration and infectious diarrhea. Conversely, maternal separation stress and Non-Steroidal Anti-Inflammatory Drugs (NSAID's) can induce intestinal injury and bleeding. This study aimed to evaluate in suckling rats if maternal separation/formula feeding leads to increased intestinal sensitivity to indomethacin (indo)-induced intestinal injury and to look at potential mechanisms involved. METHODS: Nine-day-old rats were dam-fed or separated/trained to formula-feed for 6 days prior to indo administration (5 mg/kg/day) or saline (control) for 3 days. Intestinal bleeding and injury were assessed by measuring luminal and Fecal Hemoglobin (Hob) and jejunal histology. Maturation of the intestine was assessed by measuring luminal bile acids, jejunal sucrase, serum corticosterone, and mRNA expression of ileal Apical Sodium-Dependent Bile Acid Transporter (ASBT). RESULTS: At 17 days, formula-fed indo-treated pups had a 2-fold increase in luminal Hb compared to formula-fed control pups and had evidence of morphological injury to the small intestinal mucosa as observed at the light microscopic level, whereas indo had no effect on dam-fed littermates. In addition, formula-fed rats had significant increases in luminal bile acid, sucrase specific activity, serum corticosterone, and expression of ASBT mRNA compared to dam-fed rats. CONCLUSION: Maternal separation stress may cause early intestinal maturational changes induced by corticosteroid release, including increased epithelial exposure to bile acids. These maturational changes may have a sensitizing rather than protective effect against indo-induced injury in the new-born.

3.
Placenta ; 36(8): 956-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26119969

RESUMEN

Nutrient sulfate is important for fetal development. The fetus has a limited capacity to generate sulfate and relies on maternal sulfate supplied via the placenta. The gestational age when fetal sulfate generation begins is unknown but would require cysteine dioxygenase (CDO1) which mediates a major step of sulfate production from cysteine. We investigated the ontogeny of Cdo1 mRNA expression in mouse fetal and placental tissues, which showed increasing levels from embryonic day 10.5 and was localised to the decidua and several fetal tissues including nasal cavities and brain. These findings suggest a role for Cdo1 in sulfate generation from mid-gestation.


Asunto(s)
Cisteína-Dioxigenasa/genética , Feto/metabolismo , Expresión Génica , Placenta/metabolismo , Animales , Encéfalo/metabolismo , Cisteína-Dioxigenasa/metabolismo , Femenino , Ratones , Embarazo
4.
Placenta ; 34(4): 381-4, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23453247

RESUMEN

Sulfate is an important nutrient for fetal growth and development. The fetus has no mechanism for producing its own sulfate and is therefore totally reliant on sulfate from the maternal circulation via placental sulfate transport. To build a model of directional sulfate transport in the placenta, we investigated the relative abundance of the 10 known sulfate transporter mRNAs in human placenta from uncomplicated term pregnancies. SLC13A4 and SLC26A2 were the most abundant sulfate transporter mRNAs, which localized to syncytiotrophoblast and cytotrophoblast cells, respectively. These findings indicate important physiological roles for SLC13A4 and SLC26A2 in human placental sulfate transport.


Asunto(s)
Proteínas de Transporte de Anión/biosíntesis , Placenta/metabolismo , Simportadores/biosíntesis , Trofoblastos/metabolismo , Transporte Biológico , Femenino , Humanos , Embarazo , ARN Mensajero/metabolismo , Transportadores de Sulfato , Sulfatos/metabolismo , Transcriptoma
5.
Biochem Biophys Res Commun ; 433(1): 79-83, 2013 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-23485456

RESUMEN

The human solute linked carrier (SLC) 13A4 gene is primarily expressed in the placenta where it is proposed to mediate the transport of nutrient sulfate from mother to fetus. The molecular mechanisms involved in the regulation of SLC13A4 expression remain unknown. To investigate the regulation of SLC13A4 gene expression, we analysed the transcriptional activity of the human SLC13A4 5'-flanking region in the JEG-3 placental cell line using luciferase reporter assays. Basal transcriptional activity was identified in the region -57 to -192 nucleotides upstream of the SLC13A4 transcription initiation site. Mutational analysis of the minimal promoter region identified Nuclear factor Y (NFY), Specificity protein 1 (SP1) and Krüppel like factor 7 (KLF7) motifs which conferred positive transcriptional activity, as well as Zinc finger protein of the cerebellum 2 (ZIC2) and helix-loop-helix protein 1 (HEN1) motifs that repressed transcription. The conserved NFY, SP1, KLF7, ZIC2 and HEN1 motifs in the SLC13A4 promoter of placental species but not in non-placental species, suggests a potential role for these putative transcriptional factor binding motifs in the physiological control of SLC13A4 mRNA expression.


Asunto(s)
Proteínas de Transporte de Anión/genética , Simportadores/genética , Región de Flanqueo 5' , Secuencia de Bases , Línea Celular , Secuencia Conservada , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Humanos , Luciferasas/genética , Mutagénesis Sitio-Dirigida , Filogenia , Placenta/metabolismo , Embarazo , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transportadores de Sulfato , Transcripción Genética
6.
Br J Pharmacol ; 170(8): 1449-58, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24528237

RESUMEN

The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties from the IUPHAR database. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. This compilation of the major pharmacological targets is divided into seven areas of focus: G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors & Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and GRAC and provides a permanent, citable, point-in-time record that will survive database updates.


Asunto(s)
Bases de Datos Farmacéuticas , Terapia Molecular Dirigida , Farmacología , Humanos , Ligandos , Preparaciones Farmacéuticas/química
7.
Artículo en Inglés | MEDLINE | ID: mdl-21111708

RESUMEN

This article has been withdrawn at the request of the editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

8.
Gut ; 58(7): 910-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19201772

RESUMEN

OBJECTIVE: Sulfate (SO(4)(2-)) is an abundant component of intestinal mucins and its content is decreased in certain gastrointestinal diseases, including inflammatory bowel disease. In this study, the hyposulfataemic NaS1 sulfate transporter null (Nas1(-/-)) mice were used to investigate the physiological consequences of disturbed sulfate homeostasis on (1) intestinal sulfomucin content and mRNA expression; (2) intestinal permeability and proliferation; (3) dextran sulfate sodium (DSS)-induced colitis; and (4) intestinal barrier function against the bacterial pathogen, Campylobacter jejuni. METHODS: Intestinal sulfomucins and sialomucins were detected by high iron diamine staining, permeability was assessed by fluorescein isothiocyanate (FITC)-dextran uptake, and proliferation was assessed by 5-bromodeoxyuridine (BrdU) incorporation. Nas1(-/-) and wild-type (Nas1(+/+)) mice received DSS in drinking water, and intestinal damage was assessed by histological, clinical and haematological measurements. Mice were orally inoculated with C jejuni, and intestinal and systemic infection was assessed. Ileal mRNA expression profiles of Nas1(-/-) and Nas1(+/+) mice were determined by cDNA microarrays and validated by quantitative real-time PCR. RESULTS: Nas1(-/-) mice exhibited reduced intestinal sulfomucin content, enhanced intestinal permeability and DSS-induced colitis, and developed systemic infections when challenged orally with C jejuni. The transcriptional profile of 41 genes was altered in Nas1(-/-) mice, with the most upregulated gene being pancreatic lipase-related protein 2 and the most downregulated gene being carbonic anhydrase 1 (Car1). CONCLUSION: Sulfate homeostasis is essential for maintaining a normal intestinal metabolic state, and hyposulfataemia leads to reduced intestinal sulfomucin content, enhanced susceptibility to toxin-induced colitis and impaired intestinal barrier to bacterial infection.


Asunto(s)
Colitis/metabolismo , Mucosa Intestinal/metabolismo , Mucinas/metabolismo , Animales , Infecciones por Campylobacter/microbiología , Campylobacter jejuni/aislamiento & purificación , Colitis/inducido químicamente , Colitis/microbiología , Inmunohistoquímica , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Noqueados , Factores de Tiempo
9.
Anal Biochem ; 374(1): 106-11, 2008 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-18082126

RESUMEN

An HPLC-MS/MS method has been developed for the selective quantitative analysis of paracetamol and its two major metabolites. The use of tandem MS enabled the detection and quantitation of metabolites in small sample sizes with high sensitivity and selectivity. Isocratic elution using acetonitrile and water containing formic acid combined with electrospray-tandem MS enabled the separation and accurate quantitation of each analyte and the internal standard 3-acetamidophenol. The on-column limits of detection for paracetamol, paracetamol sulfate, and paracetamol glucuronide were 2.4, 1.2, and 1.2 pmol, respectively. The method was applied to quantitate paracetamol and its metabolites in mouse urine. It is highly specific, sensitive, and easily adaptable to measure these analytes in biological fluids of other animals.


Asunto(s)
Acetaminofén/orina , Cromatografía Líquida de Alta Presión/métodos , Espectrometría de Masas en Tándem/métodos , Acetaminofén/análogos & derivados , Animales , Masculino , Ratones , Sensibilidad y Especificidad
10.
Curr Med Chem ; 12(4): 385-96, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15720248

RESUMEN

Over the past decade, 11 human genes belonging to the solute linked carrier (SLC) 26 family of transporters, have been identified. The SLC26 proteins, which include SAT-1, DTDST, DRA/CLD, pendrin, prestin, PAT-1/CFEX and Tat-1, are structurally related and have been shown to transport one or more of the following substrates: sulfate, chloride, bicarbonate, iodide, oxalate, formate, hydroxyl or fructose. Special interest has focused on four members of the SLC26 family that are associated with distinct recessive diseases: (i) Mutations in SLC26A2 lead to four different chondrodysplasias (diastrophic dysplasia, atelosteogenesis type II, achondrogenesis type IB and multiple epiphyseal dysplasia); (ii) SLC26A3 is associated with congenital chloride diarrhea; (iii) SLC26A4 is associated with Pendred syndrome and non-syndromic deafness, DFNB4; and (iv) SLC26A5 is defective in non-syndromic hearing impairment. This review article summarizes current information on the pathophysiological consequences of mutations in the human SLC26A2 to A5 genes.


Asunto(s)
Condrodisplasia Punctata/genética , Diarrea/genética , Pérdida Auditiva/genética , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Anión , Proteínas Portadoras/genética , Humanos , Mutación , Transportadores de Sulfato
11.
Pflugers Arch ; 444(3): 353-9, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12111243

RESUMEN

The renal sodium-sulfate cotransporter, NaS(i)-1, a protein implicated to control serum sulfate levels, has been shown to be regulated in vivo by 1,25-dihydroxyvitamin D(3) (1,25-(OH)(2)D(3)) and tri-iodothyronine (T(3)). Recently, we cloned the mouse NaS(i)-1 gene ( Nas1) and in the present study identified a 1,25-(OH)(2)D(3)- and T(3)-responsive element located within the Nas1 promoter. Mutational analysis of the Nas1 promoter resulted in identification of a direct repeat 6-type vitamin-D-responsive element (DR6 VDRE) at -525 to -508 and an imperfect inverted repeat 0-type T(3)-responsive element (IR0 T(3)RE) at -436 to -425 which conferred 1,25-(OH)(2)D(3) and T(3) responsiveness, respectively. In summary, we have identified responsive elements that mediate the enhanced transcription of Nas1 by 1,25-(OH)(2)D(3) and T(3), and these mechanisms may provide important clues to the physiological control of sulfate homeostasis.


Asunto(s)
Calcitriol/farmacología , Agonistas de los Canales de Calcio/farmacología , Proteínas de Transporte de Catión , Regiones Promotoras Genéticas/fisiología , Simportadores/genética , Triyodotironina/farmacología , Animales , Células Cultivadas , Homeostasis/fisiología , Riñón/citología , Luciferasas/genética , Ratones , Zarigüeyas , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Mensajero/análisis , Elementos de Respuesta , Cotransportador de Sodio-Sulfato , Sulfatos/metabolismo , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/fisiología
12.
J Interferon Cytokine Res ; 21(11): 899-904, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11747621

RESUMEN

Genes in the interleukin-1 (IL-1) gene cluster on human chromosome 2 play an important role in mediating inflammatory responses and are associated with numerous diseases. We have identified a novel IL-1-like gene, IL-1F10, on human chromosome 2q13-14.1 near the IL-1 receptor antagonist gene (IL-1RN). The IL1F10 gene is encoded by 5 exons spanning over 7.8 kb of genomic DNA. The 1008-bp IL-1F10 cDNA encodes a 152-amino acid protein that shares between 41% and 43% amino acid identity with human IL-1 receptor antagonist (IL-1Ra) and FIL-1delta, respectively. IL-1F10 shares characteristics of the IL-1Ra family, including key amino acid consensus sequences and a similar genomic structure. By multitissue first-strand cDNA PCR analysis, IL-1F10 mRNA is expressed in heart, placenta, fetal liver, spleen, thymus, and tonsil. The expression in a variety of immune tissues and similarity to IL-1Ra suggest a role of IL-1F10 in the inflammatory response.


Asunto(s)
Cromosomas Humanos Par 2 , Interleucina-1/genética , Familia de Multigenes , Secuencia de Aminoácidos , Mapeo Cromosómico , Humanos , Proteína Antagonista del Receptor de Interleucina 1 , Interleucina-1/biosíntesis , Datos de Secuencia Molecular , Filogenia , ARN Mensajero/biosíntesis , Homología de Secuencia de Aminoácido , Sialoglicoproteínas/genética , Distribución Tisular
13.
Arterioscler Thromb Vasc Biol ; 21(12): 2039-45, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11742882

RESUMEN

Familial hypertriglyceridemia (FHTG), a disease characterized by elevated plasma very low density lipoprotein triglyceride levels, has been associated with impaired intestinal absorption of bile acids. The aim of this study was to test the hypothesis that defects in the active ileal absorption of bile acids are a primary cause of FHTG. Single-stranded conformation polymorphism analysis was used to screen the ileal Na(+)/bile acid cotransporter gene (SLC10A2) for FHTG-associated mutations. Analysis of 20 hypertriglyceridemic patients with abnormal bile acid metabolism revealed 3 missense mutations (V98I, V159I, and A171S), a frame-shift mutation (646insG) at codon 216, and 4 polymorphisms in the 5' flanking sequence of SLC10A2. The SLC10A2 missense mutations and 5' flanking sequence polymorphisms were not correlated with bile acid production or turnover in the hypertriglyceridemic patients and were equally prevalent in the unaffected control subjects. In transfected COS cells, the V98I, V159I, and A171S isoforms all transported bile acids similar to the wild-type SLC10A2. The 646insG frame-shift mutation abolished bile acid transport activity in transfected COS cells but was found in only a single FHTG patient. These findings indicate that the decreased intestinal bile acid absorption in FHTG patients is not commonly associated with inherited defects in SLC10A2.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Proteínas Portadoras/análisis , Hiperlipoproteinemia Tipo IV/genética , Hiperlipoproteinemia Tipo IV/metabolismo , Ilion/fisiopatología , Transportadores de Anión Orgánico Sodio-Dependiente , Simportadores , Adulto , Femenino , Mutación del Sistema de Lectura , Frecuencia de los Genes , Humanos , Absorción Intestinal , Masculino , Persona de Mediana Edad
14.
Mol Genet Metab ; 74(1-2): 186-99, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11592815

RESUMEN

We have carried out a detailed sequence and functional analysis of a novel human facilitative glucose transporter, designated GLUT10, located in the Type 2 diabetes-linked region of human chromosome 20q12-13.1. The GLUT10 gene is located between D20S888 and D20S891 and is encoded by 5 exons spanning 26.8 kb of genomic DNA. The human GLUT10 cDNA encodes a 541 amino acid protein that shares between 31 and 35% amino acid identity with human GLUT1-8. The predicted amino acid sequence of GLUT10 is nearly identical in length to the recently described GLUT9 homologue, but is longer than other known members of the GLUT family. In addition, we have cloned the mouse cDNA homolog of GLUT10 that encodes a 537 amino acid protein that shares 77.3% identity with human GLUT10. The amino acid sequence probably has 12 predicted transmembrane domains and shares characteristics of other mammalian glucose transporters. Human and mouse GLUT10 retain several sequence motifs characteristic of mammalian glucose transporters including VP497ETKG in the cytoplasmic C-terminus, G73R[K,R] between TMD2 and TMD3 (PROSITE PS00216), VD92RAGRR between TMD8 and TMD9 (PROSITE PS00216), Q242QLTG in TMD7, and tryptophan residues W430 (TMD10) and W454 (TMD11), that correspond to trytophan residues previously implicated in GLUT1 cytochalasin B binding and hexose transport. Neither human nor mouse GLUT10 retains the full P[E,D,N]SPR motif after Loop6 but instead is replaced with P186AG[T,A]. A PROSITE search also shows that GLUT10 has lost the SUGAR TRANSPORT 2 pattern (PS00217), a result of the substitution G113S in TMD4, while all other known human GLUTs retain the glycine and the pattern match. The significance of this substitution is unknown. Sites for N-linked glycosylation are predicted at N334ATG between TMD8 and TMD9 and N526STG in the cytoplasmic C-terminus. Northern hybridization analysis identified a single 4.4-kb transcript for GLUT10 in human heart, lung, brain, liver, skeletal muscle, pancreas, placenta, and kidney. By RT-PCR analysis, GLUT10 mRNA was also detected in fetal brain and liver. When expressed in Xenopus oocytes, human GLUT10 exhibited 2-deoxy-D-glucose transport with an apparent Km of approximately 0.3 mM. D-Glucose and D-galactose competed with 2-deoxy-D-glucose and transport was inhibited by phloretin. The gene localization and functional properties suggest a role for GLUT10 in glucose metabolism and Type 2 diabetes.


Asunto(s)
Cromosomas Humanos Par 20/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Secuencia de Aminoácidos , Animales , Femenino , Proteínas Facilitadoras del Transporte de la Glucosa , Humanos , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Proteínas de Transporte de Monosacáridos/biosíntesis , Proteínas de Transporte de Monosacáridos/fisiología , Oocitos , Especificidad de Órganos/genética , Análisis de Secuencia de ADN , Xenopus laevis
15.
Scand J Gastroenterol ; 36(10): 1077-80, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11589382

RESUMEN

BACKGROUND: A congenital form of idiopathic intestinal bile acid malabsorption (IBAM) has been associated with dysfunctional mutations in the ileal apical sodium-dependent bile acid transporter (ASBT). The aim of this study was to determine whether mutations in the ASBT gene (SLC10A2) predispose to the development of adult-onset idiopathic bile acid malabsorption and chronic watery diarrhea. METHODS: Genomic DNA was obtained from 13 adult IBAM patients previously diagnosed on the basis of clinical data, response to cholestyramine, and abnormal 75Se-homocholic acid taurine (SeHCAT) test values. The ASBT gene was screened for the presence of mutations or polymorphisms by single-stranded conformation polymorphism analysis (SSCP) and DNA sequencing. RESULTS: ASBT gene polymorphisms were detected in 5 of the 13 adult IBAM patients. Four patients were heterozygous for a common polymorphism in exon 3, leading to an alanine to serine substitution at codon 171 (A171S). An additional subject was heterozygous for a polymorphism in exon 1 that causes a valine to isoleucine substitution at codon 98 (V981). These functional polymorphisms were also found in unaffected subjects and do not appear to affect ASBT function. CONCLUSIONS: Adult-onset IBAM is not directly related to dysfunctional mutations in the coding region or intron/exon junctions of the SLC10A2 gene. In the absence of apparent ileal disease or intestinal motility defects, inappropriate down-regulation of the ileal bile acid transporter or defects in ileocyte transfer of bile acids into the portal circulation could explain this form of adult IBAM.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Proteínas Portadoras/genética , Diarrea/genética , Síndromes de Malabsorción/genética , Transportadores de Anión Orgánico Sodio-Dependiente , Simportadores , Adulto , Regulación hacia Abajo/fisiología , Femenino , Humanos , Masculino , Mutación/genética
16.
Am J Physiol Gastrointest Liver Physiol ; 279(6): G1188-200, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11093941

RESUMEN

The rat and mouse organic anion-transporting polypeptides (oatp) subtype 3 (oatp3) were cloned to further define components of the intestinal bile acid transport system. In transfected COS cells, oatp3 mediated Na(+)-independent, DIDS-inhibited taurocholate uptake (Michaelis-Menten constant approximately 30 microM). The oatp3-mediated uptake rates and affinities were highest for glycine-conjugated dihydroxy bile acids. In stably transfected, polarized Madin-Darby canine kidney (MDCK) cells, oatp3 mediated only apical uptake of taurocholate. RT-PCR analysis revealed that rat oatp3, but not oatp1 or oatp2, was expressed in small intestine. By RNase protection assay, oatp3 mRNA was readily detected down the length of the small intestine as well as in brain, lung, and retina. An antibody directed to the carboxy terminus localized oatp3 to the apical brush-border membrane of rat jejunal enterocytes. The mouse oatp3 gene was localized to a region of mouse chromosome 6. This region is syntenic with human chromosome 12p12, where the human OATP-A gene was mapped, suggesting that rodent oatp3 is orthologous to the human OATP-A. These transport and expression properties suggest that rat oatp3 mediates the anion exchange-driven absorption of bile acids previously described for the proximal small intestine.


Asunto(s)
Proteínas Portadoras/genética , Cromosomas , Transportadores de Anión Orgánico Sodio-Independiente , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Secuencia de Aminoácidos , Animales , Proteínas de Transporte de Anión , Secuencia de Bases , Bilis/metabolismo , Células COS , Proteínas Portadoras/farmacocinética , Perros , Humanos , Intestinos/química , Cinética , Masculino , Ratones , Datos de Secuencia Molecular , Ratas , Ratas Sprague-Dawley , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Sodio/metabolismo
17.
Nucleic Acids Res ; 28(20): 4013-20, 2000 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11024182

RESUMEN

Ribozymes are a promising agent for the gene therapy of dominant negative genetic disorders by allele-specific mRNA suppression. To test allele-specific mRNA suppression in cells, we used fibroblasts from a patient with osteogenesis imperfecta (OI). These cells contain a mutation in one alpha1(I) collagen allele which both causes the skeletal disorder and generates a novel ribozyme cleavage site. In a preliminary in vitro assay, ribozymes cleaved mutant RNA substrate whereas normal substrate was left intact. For the studies in cell culture we generated cell lines stably expressing active (AR) and inactive (IR) ribozymes targeted to mutant alpha1(I) collagen mRNA. Quantitative competitive RT-PCR analyses of type I collagen mRNA, normalized to beta-actin expression levels, revealed that the level of mutant alpha1(I) collagen mRNA was significantly decreased by approximately 50% in cells expressing AR. Normal alpha1(I) collagen mRNA showed no significant reduction when AR or IR was expressed from the pHbetaAPr-1-neo vector and a small (10-20%) but significant reduction when either ribozyme was expressed from the pCI.neo vector. In clonal lines derived from cells expressing AR the level of ribozyme expression correlated with the extent of reduction in the mutant:normal alpha1(I) mRNA ratio, ranging from 0.33 to 0.96. Stable expression of active ribozyme did not affect cell viability, as assessed by growth rates. Ribozyme cleavage of mutant mRNA results in a reduction in mutant type I collagen protein, as demonstrated by SDS-urea-PAGE. This is the first report of ribozymes causing specific suppression of an endogenous mutant mRNA in cells derived from a patient with a dominant negative genetic disorder.


Asunto(s)
Colágeno/genética , Terapia Genética , Osteogénesis Imperfecta/genética , Mutación Puntual/genética , ARN Catalítico/metabolismo , Alelos , Secuencia de Bases , Unión Competitiva , División Celular , Supervivencia Celular , Células Cultivadas , Colágeno/metabolismo , Fibroblastos , Genes Dominantes/genética , Heterocigoto , Humanos , Cinética , Conformación de Ácido Nucleico , Osteogénesis Imperfecta/terapia , Pepsina A/metabolismo , Plásmidos/genética , ARN Catalítico/química , ARN Catalítico/genética , ARN Catalítico/uso terapéutico , ARN Mensajero/análisis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad por Sustrato , Moldes Genéticos , Transfección
18.
Proc Natl Acad Sci U S A ; 97(20): 11092-7, 2000 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-10984521

RESUMEN

Bile secretion involves the structural and functional interplay of hepatocytes and cholangiocytes, the cells lining the intrahepatic bile ducts. Hepatocytes actively secrete bile acids into the canalicular space and cholangiocytes then transport bile acids in a vectorial manner across their apical and basolateral plasma membranes. The initial step in the transepithelial transport of bile acids across rat cholangiocytes is apical uptake by a Na(+)-dependent bile acid transporter (ASBT). To date, the molecular basis of the obligate efflux mechanism for extrusion of bile acids across the cholangiocyte basolateral membrane remains unknown. We have identified an exon-2 skipped, alternatively spliced form of ASBT, designated t-ASBT, expressed in rat cholangiocytes, ileum, and kidney. Alternative splicing causes a frameshift that produces a 154-aa protein. Antipeptide antibodies detected the approximately 19 kDa t-ASBT polypeptide in rat cholangiocytes, ileum, and kidney. The t-ASBT was specifically localized to the basolateral domain of cholangiocytes. Transport studies in Xenopus oocytes revealed that t-ASBT can function as a bile acid efflux protein. Thus, alternative splicing changes the cellular targeting of ASBT, alters its functional properties, and provides a mechanism for rat cholangiocytes and other bile acid-transporting epithelia to extrude bile acids. Our work represents an example in which a single gene appears to encode via alternative splicing both uptake and obligate efflux carriers in a bile acid-transporting epithelial cell.


Asunto(s)
Empalme Alternativo/fisiología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Hígado/metabolismo , Transportadores de Anión Orgánico Sodio-Dependiente , Simportadores , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Transporte Biológico/fisiología , Compartimento Celular , Hígado/citología , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Datos de Secuencia Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Endogámicas F344 , Sodio/metabolismo
19.
Carcinogenesis ; 20(10): 1905-11, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10506103

RESUMEN

This study was undertaken to investigate the influence of the peroxisome proliferator-activated receptor gamma (PPARgamma) agonists on the proliferation, apoptosis and tumorigenesis of breast cancer cells. PPARgamma investigation has been largely restricted to adipose tissue, where it plays a key role in differentiation, but recent data reveal that PPARgamma is expressed in several transformed cells. However, the function of PPARgamma activation in neoplastic cells is unclear. Activation of PPARgamma with the known prostanoid agonist 15-deoxy-Delta12,14-prostaglandin J(2) (15dPGJ(2)) or the thiazolidinedione (TZD) agonist troglitazone (TGZ) attenuated cellular proliferation of the estrogen receptor-negative breast cancer cell line MDA-MB-231, as well as the estrogen receptor-positive breast cancer cell line MCF-7. This was marked by a decrease in total cell number and by an inhibition of cell cycle progression. Addition of 15dPGJ(2) was not associated with an increase in cellular differentiation, as has been seen in other neoplastic cells, but rather induction of cellular events associated with programmed cell death, apoptosis. Video time-lapse microscopy revealed that 15dPGJ(2) induced morphological changes associated with apoptosis, including cellular rounding, blebbing, the production of echinoid spikes, blistering and cell lysis. In contrast, TGZ caused only a modest induction of apoptosis. These results were verified by histochemistry using the specific DNA stain DAPI to observe nuclear condensation, a marker of apoptosis. Finally, a brief exposure of MDA-MB-231 cells to 15dPGJ(2) initiated an irreversible apoptotic pathway that inhibited the growth of tumors in a nude mouse model. These findings illustrate that induction of apoptosis may be the primary biological response resulting from PPARgamma activation in some breast cancer cells and further suggests a potential role for PPARgamma ligands for the treatment of breast cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Transformación Celular Neoplásica/efectos de los fármacos , Prostaglandina D2/análogos & derivados , Animales , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , División Celular , Cartilla de ADN , Humanos , Ratones , Prostaglandina D2/farmacología , ARN Mensajero/genética , Receptores Citoplasmáticos y Nucleares/genética , Factores de Transcripción/genética , Células Tumorales Cultivadas
20.
Curr Opin Lipidol ; 10(4): 315-20, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10482134

RESUMEN

The strong association between intestinal cholesterol absorption and total plasma cholesterol level has renewed interest in the absorptive process and stimulated the generation of new animal models. Increasingly, new studies suggest that cholesterol absorption is genetically controlled and supports a protein-mediated mechanism for cholesterol uptake into the intestinal mucosal cell. Insights into potential mechanisms are predicted to lead to novel pharmacological approaches to inhibit cholesterol absorption.


Asunto(s)
Colesterol/metabolismo , Absorción Intestinal/fisiología , Animales , Colesterol/sangre , Humanos , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/genética , Primates
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA