Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Drug Metab Rev ; 46(3): 261-82, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24483608

RESUMO

The historical phasing concept of drug metabolism and elimination was introduced to comprise the two phases of metabolism: phase I metabolism for oxidations, reductions and hydrolyses, and phase II metabolism for synthesis. With this concept, biological membrane barriers obstructing the accessibility of metabolism sites in the cells for drugs were not considered. The concept of two phases was extended to a concept of four phases when drug transporters were detected that guided drugs and drug metabolites in and out of the cells. In particular, water soluble or charged drugs are virtually not able to overcome the phospholipid membrane barrier. Drug transporters belong to two main clusters of transporter families: the solute carrier (SLC) families and the ATP binding cassette (ABC) carriers. The ABC transporters comprise seven families with about 20 carriers involved in drug transport. All of them operate as pumps at the expense of ATP splitting. Embedded in the former phase concept, the term "phase III" was introduced by Ishikawa in 1992 for drug export by ABC efflux pumps. SLC comprise 52 families, from which many carriers are drug uptake transporters. Later on, this uptake process was referred to as the "phase 0 transport" of drugs. Transporters for xenobiotics in man and animal are most expressed in liver, but they are also present in extra-hepatic tissues such as in the kidney, the adrenal gland and lung. This review deals with the function of drug carriers in various organs and their impact on drug metabolism and elimination.


Assuntos
Transporte Biológico/fisiologia , Inativação Metabólica/fisiologia , Proteínas de Membrana Transportadoras/metabolismo , Xenobióticos/metabolismo , Animais , Portadores de Fármacos/metabolismo , Humanos , Fígado/metabolismo
2.
Drug Metab Rev ; 46(3): 291-324, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24666398

RESUMO

In general, xenobiotic metabolizing enzymes (XMEs) are expressed in lower levels in the extrahepatic tissues than in the liver, making the former less relevant for the clearance of xenobiotics. Local metabolism, however, may lead to tissue-specific adverse responses, e.g. organ toxicities, allergies or cancer. This review summarizes the knowledge on the expression of phase I and phase II XMEs and transporters in extrahepatic tissues at the body's internal-external interfaces. In the lung, CYPs of families 1, 2, 3 and 4 and epoxide hydrolases are important phase I enzymes, while conjugation is less relevant. In skin, phase I-related enzymatic reactions are considered less relevant. Predominant skin XMEs are phase II enzymes, whereby glucuronosyltransferases (UGT) 1, glutathione-S-transferase (GST) and N-acetyltransferase (NAT) 1 are important for detoxification. The intestinal epithelium expresses many transporters and phase I XME with high levels of CYP3A4 and CYP3A5 and phase II metabolism is mainly related to UGT, NAT and Sulfotransferases (SULT). In the kidney, conjugation reactions and transporters play a major role for excretion processes. In the bladder, CYPs are relevant and among the phase II enzymes, NAT1 is involved in the activation of bladder carcinogens. Expression of XMEs is regulated by several mechanisms (nuclear receptors, epigenetic mechanisms, microRNAs). However, the understanding why XMEs are differently expressed in the various tissues is fragmentary. In contrast to the liver - where for most XMEs lower expression is demonstrated in early life - the XME ontogeny in the extrahepatic tissues remains to be investigated.


Assuntos
Transporte Biológico/fisiologia , Xenobióticos/metabolismo , Animais , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Proteínas de Membrana Transportadoras/metabolismo
3.
World J Urol ; 31(1): 219-24, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22120415

RESUMO

PURPOSE: To analyse whether the permeability of the blood-brain barrier to the antimuscarinic drug trospium chloride is altered with ageing. This is a relevant question for elderly patients with overactive bladder syndrome who are treated with trospium chloride as the occurrence of adverse effects on the central nervous system (CNS) highly depends on the absolute drug concentration in the brain. METHODS: Trospium chloride at 1 mg/kg was intravenously administered to adult, middle-aged, and aged mice at 6, 12, and 24 months of age, respectively, and the absolute drug concentrations in the brain were analysed after 2 h. Furthermore, mRNA expression levels of relevant markers of blood-brain barrier integrity (occludin, claudin-5, and the drug efflux carrier P-glycoprotein) were analysed in brain samples from adult and aged mice. RESULTS: The absolute brain concentrations of the drug were identical in adult and middle-aged mice (13 ± 2 ng/g vs. 13 ± 2 ng/g) and were slightly, but significantly, lower in aged mice (8 ± 4 ng/g). The brain/plasma drug concentration ratios were not different between the age groups and demonstrated the generally low capability of trospium chloride in permeating the blood-brain barrier. Occludin, claudin-5, and P-glycoprotein showed identical mRNA expression levels in the brains of adult and aged mice. CONCLUSION: Based on our in vivo data in a mouse model, we conclude that trospium chloride permeation across the BBB is not increased in ageing per se, and therefore, the occurrence of adverse CNS drug effects is also not expected to increase with ageing.


Assuntos
Envelhecimento/metabolismo , Benzilatos/farmacocinética , Barreira Hematoencefálica/metabolismo , Antagonistas Muscarínicos/farmacocinética , Nortropanos/farmacocinética , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Animais , Encéfalo/metabolismo , Claudina-5/genética , Claudina-5/metabolismo , Feminino , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ocludina/genética , Ocludina/metabolismo , Permeabilidade
4.
Curr Top Membr ; 70: 105-68, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23177985

RESUMO

The SLC10 family represents seven genes containing 1-12 exons that encode proteins in humans with sequence lengths of 348-477 amino acids. Although termed solute carriers (SLCs), only three out of seven (i.e. SLC10A1, SLC10A2, and SLC10A6) show sodium-dependent uptake of organic substrates across the cell membrane. These include the uptake of bile salts, sulfated steroids, sulfated thyroidal hormones, and certain statin drugs by SLC10A1 (Na(+)-taurocholate cotransporting polypeptide (NTCP)), the uptake of bile salts by SLC10A2 (apical sodium-dependent bile acid transporter (ASBT)), and uptake of sulfated steroids and sulfated taurolithocholate by SLC10A6 (sodium-dependent organic anion transporter (SOAT)). The other members of the family are orphan carriers not all localized in the cell membrane. The name "bile acid transporter family" arose because the first two SLC10 members (NTCP and ASBT) are carriers for bile salts that establish their enterohepatic circulation. In recent years, information has been obtained on their 2D and 3D membrane topology, structure-transport relationships, and on the ligand and sodium-binding sites. For SLC10A2, the putative 3D morphology was deduced from the crystal structure of a bacterial SLC10A2 analog, ASBT(NM). This information was used in this chapter to calculate the putative 3D structure of NTCP. This review provides first an introduction to recent knowledge about bile acid synthesis and newly found bile acid hormonal functions, and then describes step-by-step each individual member of the family in terms of expression, localization, substrate pattern, as well as protein topology with emphasis on the three functional SLC10 carrier members.


Assuntos
Transportadores de Ânions Orgânicos Dependentes de Sódio/química , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Sódio/metabolismo , Simportadores/química , Simportadores/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Humanos , Ácido Taurolitocólico/metabolismo
5.
Drug Metab Dispos ; 37(7): 1371-4, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19389858

RESUMO

The aim of the present study was to characterize the role of the drug-efflux transporter P-glycoprotein (P-gp) for the disposition of trospium chloride, a widely used anticholinergic drug for the treatment of overactive bladder. P-gp-deficient mdr1a,b(-/-) knockout mice were given either 1 mg/kg trospium chloride orally or 1 mg/kg intravenously to analyze brain penetration, intestinal secretion, and hepatobiliary excretion of the drug. The concentrations of trospium chloride in the brain were up to 7 times higher in the mdr1a,b(-/-) knockout mice compared with wild-type mice (p < 0.05), making P-gp a limiting factor for the blood-brain barrier penetration of this drug. Moreover, the residence time of the drug in the central nervous system was significantly prolonged in mdr1a,b(-/-) knockout mice. Apart from the blood-brain barrier, P-gp also had significant effects on the overall pharmacokinetics of trospium chloride. In the mdr1a,b(-/-) knockout mice, hepatobiliary excretion and intestinal secretion were significantly reduced compared with the wild-type mice. Our study indicates that the multidrug resistance transporter P-gp is a major determinant for the distribution of trospium chloride in the body and highly restricts its entry into the brain.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Transporte Biológico/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Camundongos , Nortropanos/farmacologia , Bexiga Urinária Hiperativa/metabolismo , Animais , Benzilatos , Barreira Hematoencefálica/fisiologia , Encéfalo/metabolismo , Radioisótopos de Carbono/metabolismo , Glicoproteínas/metabolismo , Humanos , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/metabolismo , Masculino , Taxa de Depuração Metabólica , Camundongos Knockout , Nortropanos/urina , Distribuição Tecidual/efeitos dos fármacos
6.
Biochim Biophys Acta ; 1768(9): 2070-7, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17707334

RESUMO

To determine which efflux carriers are involved in hepatic phalloidin elimination, hepatobiliary [(3)H]-demethylphalloin (DMP) excretion was studied in normal Wistar rats and in Mrp2 deficient TR(-) Wistar rats as well as in normal wild-type FVB mice, Mdr1a,b(-/-) knockout mice, and Bcrp1(-/-) knockout mice by in situ bile duct/gallbladder cannulation. A subtoxic dose of 0.03 mg DMP/kg b.w. was used, which did not induce cholestasis in any tested animal. Excretion of DMP into bile was not altered in Mdr1a,b(-/-) mice or in Bcrp1(-/-) mice compared with wild-type FVB mice. Whereas 17.6% of the applied dose was excreted into bile of normal Wistar rats, hepatobiliary excretion decreased to 7.9% in TR(-) rats within 2 h after intravenous application. This decrease was not due to reduced cellular DMP uptake, as shown by normal expression of Oatp1b2 in livers of TR(-) rats and functional DMP uptake into isolated TR(-) rat hepatocytes. Tissue concentrations of phalloidin were also not altered in any of the transgenic mice. Interestingly, the decrease of biliary DMP excretion in the TR(-) rats was not followed by any increase of phalloidin accumulation in the liver but yielded a compensatory excretion of the toxin into urine, indicating that hepatocytes of TR(-) rats expelled phalloidin back into blood circulation.


Assuntos
Alcaloides/metabolismo , Alcaloides/farmacologia , Amanita/metabolismo , Bile/metabolismo , Fígado/metabolismo , Proteínas dos Microfilamentos/metabolismo , Alcaloides/farmacocinética , Animais , Bile/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Ratos , Ratos Wistar
7.
Eur J Cell Biol ; 86(8): 445-60, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17628207

RESUMO

The 'Solute Carrier Family SLC10' consists of six annotated members in humans, comprising two bile acid carriers (SLC10A1 and SLC10A2), one steroid sulfate transporter (SLC10A6), and three orphan carriers (SLC10A3 to SLC10A5). In this study we report molecular characterization and expression analysis of a novel member of the SLC10 family, SLC10A7, previously known as C4orf13. SLC10A7 proteins consist of 340-343 amino acids in humans, mice, rats, and frogs and show an overall amino acid sequence identity of >85%. SLC10A7 genes comprise 12 coding exons and show broad tissue expression pattern. When expressed in Xenopus laevis oocytes and HEK293 cells, SLC10A7 was detected in the plasma membrane but revealed no transport activity for bile acids and steroid sulfates. By immunofluorescence analysis of dual hemagglutinin (HA)- and FLAG-labeled SLC10A7 proteins in HEK293 cells, we established a topology of 10 transmembrane domains with an intracellular cis orientation of the N-terminal and C-terminal ends. This topology pattern is clearly different from the seven-transmembrane domain topology of the other SLC10 members but similar to hitherto uncharacterized non-vertebrate SLC10A7-related proteins. In contrast to the established SLC10 members, which are restricted to the taxonomic branch of vertebrates, SLC10A7-related proteins exist also in yeasts, plants, and bacteria, making SLC10A7 taxonomically the most widespread member of this carrier family. Vertebrate and bacterial SLC10A7 proteins exhibit >20% sequence identity, which is higher than the sequence identity of SLC10A7 to any other member of the SLC10 carrier family.


Assuntos
Bactérias/genética , Sequência Conservada , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Filogenia , Vertebrados/genética , Processamento Alternativo/genética , Sequência de Aminoácidos , Animais , Transporte Biológico , Membrana Celular/metabolismo , Clonagem Molecular , Éxons/genética , Genoma , Humanos , Imunoprecipitação , Camundongos , Dados de Sequência Molecular , Oócitos , Transportadores de Ânions Orgânicos Dependentes de Sódio/química , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Simportadores/metabolismo , Xenopus
8.
Naunyn Schmiedebergs Arch Pharmacol ; 372(6): 465-75, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16532306

RESUMO

This review deals with the drug transporters allowing drugs to enter and leave cells by carrier-mediated pathways. Emphasis is put on liver transporters but systems in gut, kidney, and blood-brain barrier are mentioned as well. Drug-drug interactions on carriers may provoke significant modification in pharmacokinetics as do carrier gene polymorphisms yielding functional carrier protein mutations. An integrated phase concept should reflect the interplay between drug metabolism and drug transport.


Assuntos
Proteínas de Membrana Transportadoras/metabolismo , Preparações Farmacêuticas/metabolismo , Animais , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Humanos , Rim/metabolismo , Fígado/metabolismo , Proteínas de Membrana Transportadoras/química , Modelos Biológicos , Xenobióticos/metabolismo , Xenobióticos/farmacocinética
9.
Mol Cell Endocrinol ; 428: 133-41, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27033324

RESUMO

The sodium-dependent organic anion transporter SOAT specifically transports sulfated steroid hormones and is supposed to play a role in testicular steroid regulation and male fertility. The present study aimed to identify novel specific SOAT inhibitors for further in vitro and in vivo studies on SOAT function. More than 100 compounds of different molecular structures were screened for inhibition of the SOAT-mediated transport of dehydroepiandrosterone sulfate in stably transfected SOAT-HEK293 cells. Twenty-five of these with IC50 values covering four orders of magnitude were selected as training set for 3D pharmacophore modelling. The SOAT pharmacophore features were calculated by CATALYST and consist of three hydrophobic sites and two hydrogen bond acceptors. By substrate database screening, compound T 0511-1698 was predicted as a novel SOAT inhibitor with an IC50 of 15 µM. This value was confirmed by cell-based transport assays. Therefore, the developed SOAT pharmacophore model demonstrated its suitability in predicting novel SOAT inhibitors.


Assuntos
Sulfato de Desidroepiandrosterona/metabolismo , Avaliação Pré-Clínica de Medicamentos , Modelos Moleculares , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Transportadores de Ânions Orgânicos/química , Relação Quantitativa Estrutura-Atividade , Ácidos e Sais Biliares/química , Ácidos e Sais Biliares/farmacologia , Células HEK293 , Humanos , Concentração Inibidora 50 , Reprodutibilidade dos Testes
10.
Biochem Pharmacol ; 66(4): 565-77, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12906921

RESUMO

As therapeutic antisense tools, oligonucleotides (ODNs) must enter cells to bind to their target structures. ODNs distribute in nearly each tissue with relatively high concentrations in kidney and liver from where excretion into urine and bile occurs. To investigate mechanisms involved in hepatic ODN transport, normal mixed backbone phosphodiester/phosphorothioate ODNs (n-ODN) and two different bile acid-conjugated mixed backbone ODNs (1BA-ODN and 2BA-ODN) were applied to two different rat strains, normal Wistar rats and Wistar TR- rats. In normal Wistar rats, concentration-dependent hepatobiliary elimination of the ODNs was observed with a remarkable increase of excretion of the cholic acid BA-ODN conjugates. In contrast to normal Wistar rats, n-ODN excretion into bile by TR- rats, a mutant Wistar rat strain lacking a functional multidrug resistance-associated protein 2 (mrp2) at the canalicular membrane, was strongly diminished, whereas these rats excreted an ODN conjugated with two cholic acid molecules (2BA-ODN) into bile. Concomitant application of substrates transported by mrp2 such as bromosulfophthalein (BSP) or the synthetic chlorogenic acid derivative S 3025 significantly reduced the biliary appearance of normal ODN and 2BA-ODN in Wistar rats and also in TR- rats. To inhibit the expression of cRNA derived from the Na+ -dependent taurocholate cotransporting polypeptide (Ntcp), antisense ODNs were constructed which fully retained the antisense properties when coupled with two bile acid molecules. The results indicate that ODNs are secreted via the mrp2 into bile. In the absence of mrp2, further excretory transport systems with affinity for bile acids seem to be relevant for their excretion. The results further indicate that bile acid tagged ODNs are useful tools for liver specific antisense therapy.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Ácidos e Sais Biliares/farmacocinética , Bile/metabolismo , Proteínas de Transporte/fisiologia , Fígado/metabolismo , Oligodesoxirribonucleotídeos/farmacocinética , Animais , Masculino , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Ratos , Ratos Wistar
11.
Comp Biochem Physiol B Biochem Mol Biol ; 137(3): 317-29, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15050519

RESUMO

We describe the cloning, functional characterization and tissue localization of a novel membrane transporter of the OATP/Oatp-gene family obtained from liver and kidney of cattle (Bos taurus). The carrier protein exhibits highest sequence identity to the human OATP1A2 (previously called OATP-A) and is, therefore, named bovine Oatp1a2. Bovine Oatp1a2 received the gene symbol Slco1a2 that is identical to the SLC classification of human OATP1A2 (SLCO1A2, previously called SLC21A3) and is likely an orthologue of the human gene. Two different full-length bOatp1a2 cDNAs of 2316-bp and 3504-bp were obtained and encoded for a 666 amino acid membrane protein, which contains twelve putative transmembrane spanning domains. Bovine Oatp1a2 expression was detected in liver, kidney, brain and adrenal gland. Uptake studies in cRNA-injected oocytes demonstrated that bOatp1a2 transports estrone-3-sulfate and taurocholate, with K(m) values of 9.6 microM and 51 microM, respectively, and estradiol-17beta-glucuronide. However, the structurally-related heart glycosides ouabain (1 microM) and digoxin (1 microM) are neither transported by bovine Oatp1a2 nor by human OATP1A2. We conclude that based on the tested substrates bovine Oatp1a2 shows functional homology to human OATP1A2.


Assuntos
Clonagem Molecular/métodos , Estradiol/análogos & derivados , Estrona/análogos & derivados , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Animais , Sequência de Bases , Transporte Biológico , Bovinos , DNA Complementar/genética , DNA Complementar/isolamento & purificação , Estradiol/metabolismo , Estrona/metabolismo , Cinética , Proteínas de Membrana , Dados de Sequência Molecular , Transportadores de Ânions Orgânicos/química , Alinhamento de Sequência , Ácido Taurocólico/metabolismo , Distribuição Tecidual
12.
Chem Biol Interact ; 224: 36-41, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25451572

RESUMO

The ATP-binding cassette transporters Breast Cancer Resistance Protein (Abcg2) and Multidrug Resistance-associated Protein 2 (Abcc2) play an important role for the hepatobiliary elimination of drugs and toxins as well as their metabolites. Previous in vitro transport studies showed that both transporters are involved in the active efflux of phase II metabolites of carcinogenic benzo[a]pyrene (BP), however the role of these carriers in hepatobiliary elimination in vivo is still unknown. In the present study, Abcg2(-/-) and Abcc2(-/-) knockout mice were used to elucidate the role of Abcg2 and Abcc2 for the hepatobiliary excretion of BP and its metabolites. After intravenous application of [(3)H]BP the hepatobiliary excretion was significantly reduced in these mice: whereas wild type mice excreted on average 25.4% of the applied dose into the bile over 90min, Abcg2(-/-) knockout mice only excreted 10.7% and Abcc2(-/-) knockout mice 8.6%. As a consequence, [(3)H]BP concentrations were in general higher in the plasma and in most of the organs of the Abcg2 and Abcc2 knockout mice. Both transporters may have a protective function for BP-induced carcinogenesis in humans, due to its crucial importance for the hepatobiliary elimination of BP via bile. Subjects with reduced ABCG2 or ABCC2 expression might have higher oral bioavailability for BP due to a reduced excretion and so might be more susceptible to BP-induced carcinogenesis.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Benzo(a)pireno/metabolismo , Bile/metabolismo , Fígado/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Animais , Benzo(a)pireno/administração & dosagem , Transporte Biológico , Encéfalo/metabolismo , Mucosa Gástrica/metabolismo , Pulmão/metabolismo , Masculino , Taxa de Depuração Metabólica , Camundongos , Camundongos Knockout , Proteína 2 Associada à Farmacorresistência Múltipla , Miocárdio/metabolismo , RNA Mensageiro/metabolismo
13.
J Steroid Biochem Mol Biol ; 138: 90-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23562556

RESUMO

The sodium-dependent organic anion transporter SOAT is a member of the Solute Carrier Family SLC10. In man, this carrier is predominantly expressed in the testis and has transport activity for sulfoconjugated steroid hormones. Here, we report on cloning, expression analysis and functional characterization of the mouse Soat (mSoat) and compare its characteristics with the human SOAT carrier. Quantitative mRNA expression analysis for mSoat in male mice revealed very high expression in lung and further high expression in testis and skin. Immunohistochemical studies showed expression of the mSoat protein in bronchial epithelial cells of the lung, in primary and secondary spermatocytes as well as round spermatids within the seminiferous tubules of the testis, in the epidermis of the skin, and in the urinary epithelium of the bladder. Stably transfected mSoat-HEK293 cells revealed sodium-dependent transport for dehydroepiandrosterone sulfate (DHEAS), estrone-3-sulfate, and pregnenolone sulfate (PREGS) with apparent Km values of 60.3µM, 2.1µM, and 2.5µM, respectively. In contrast to human SOAT, which has a preference for DHEAS as a substrate, mSoat exhibits the highest transport rate for PREGS, likely reflecting differences in the steroid pattern between both species. In conclusion, although certain differences between human SOAT and mSoat exist regarding quantitative gene expression in endocrine and non-endocrine tissues, as well as in the transport kinetics for steroid sulfates, in general, both can be regarded as homologous carriers.


Assuntos
Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Animais , Western Blotting , Linhagem Celular , Clonagem Molecular , Imunofluorescência , Hormônios Esteroides Gonadais/metabolismo , Humanos , Imuno-Histoquímica , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Pregnenolona/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pele/metabolismo , Testículo/metabolismo , Bexiga Urinária/metabolismo
14.
Toxins (Basel) ; 4(11): 1139-56, 2012 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-23202309

RESUMO

In cultures of primary rat hepatocytes, apoptosis occurred after application of 20 ng/mL tumor necrosis factor alpha (TNF-α). However, this was only in the presence of 200 ng/mL of the transcriptional inhibitor actinomycin D (ActD). This toxic effect was completely prevented in the presence of 25 µg/mL soluble TNF-α receptor I (sTNFR I) in the supernatant of hepatocyte cell cultures. Apoptosis also occurred after application of 12.5 µmol/L ochratoxin A (OTA). However, that was not prevented by up to 500 µg/mL sTNFR I, indicating that TNF-α/TNFR I is not involved in OTA mediated apoptosis in hepatocytes. The antioxidative flavanolignan silibinin in doses from 130 to 260 µmol/L prevented chromatin condensation, caspase-3 activation, and apoptotic DNA fragmentation that were induced by OTA, by 10 mmol/L hydrogen peroxide (H(2)O(2)) and by ultraviolet (UV-C) light (50 mJ/cm2), respectively. To achieve protection by silibinin, the drug was applied to the cell cultures for 2 h in advance. OTA stimulated lipid peroxidation on cultured immortalized rat liver HPCT cells, as was revealed by malondialdehyde (MDA) production. Lipid peroxidation occurred further by H(2)O(2) and ActD/TNF-α incubation. These reactions were also suppressed by silibinin pretreatment. We conclude that the anti-apoptotic activity of silibinin against OTA, H(2)O(2) and ActD/ TNF-α is caused in vitro by the antioxidative effects of the flavanolignan. Furthermore, cytotoxicity of the pro-apoptotic toxins was revealed by MTT-test. When applied separately, ActD and TNF-α showed no cytotoxic effects after 24 h, but were cytotoxic if applied in combination. The used concentrations of OTA, H(2)O(2) and the dose of UV-C caused a substantial decrease in viability within 36 h that was prevented mostly by silibinin. We conclude that silibinin is a potent protective compound against apoptosis and cytotoxicity caused by OTA and the investigated compounds.


Assuntos
Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Ocratoxinas/toxicidade , Silimarina/farmacologia , Fator de Necrose Tumoral alfa/toxicidade , Animais , Apoptose/efeitos da radiação , Caspase 3/metabolismo , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Cromatina/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Dactinomicina/farmacologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Hepatócitos/efeitos da radiação , Peróxido de Hidrogênio/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Malondialdeído/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ligação Proteica , Ratos , Ratos Wistar , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Silibina , Fator de Necrose Tumoral alfa/metabolismo , Raios Ultravioleta
16.
Vet J ; 189(1): 67-71, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20655253

RESUMO

A 4-bp deletion mutation associated with multiple drug sensitivity exists in the canine multidrug resistance (MDR1) gene. This mutation has been detected in more than 10 purebred dog breeds as well as in mixed breed dogs. To evaluate the breed distribution of this mutation in Germany, 7378 dogs were screened, including 6999 purebred and 379 mixed breed dogs. The study included dog breeds that show close genetic relationship or share breeding history with one of the predisposed breeds but in which the occurrence of the MDR1 mutation has not been reported. The breeds comprised Bearded Collies, Anatolian Shepherd Dog, Greyhound, Belgian Tervuren, Kelpie, Borzoi, Australian Cattle Dog and the Irish Wolfhound. The MDR1 mutation was not detected is any of these breeds, although it was found as expected in the Collie, Longhaired Whippet, Shetland Sheepdog, Miniature Australian Shepherd, Australian Shepherd, Wäller, White Swiss Shepherd, Old English Sheepdog and Border Collie with varying allelic frequencies for the mutant MDR1 allele of 59%, 45%, 30%, 24%, 22%, 17%, 14%, 4% and 1%, respectively. Allelic frequencies of 8% and 2% were determined in herding breed mixes and unclassified mixed breeds, respectively. Because of its widespread breed distribution and occurrence in many mixed breed dogs, it is difficult for veterinarians and dog owners to recognise whether MDR1-related drug sensitivity is relevant for an individual animal. This study provides a comprehensive overview of all affected dog breeds and many dog breeds that are probably unaffected on the basis of ∼15,000 worldwide MDR1 genotyping data.


Assuntos
Cães/genética , Genes MDR/genética , Mutação , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Animais , Resistência a Múltiplos Medicamentos/genética , Frequência do Gene , Testes Genéticos , Genótipo , Alemanha , Especificidade da Espécie
17.
Toxins (Basel) ; 2(6): 1279-99, 2010 06.
Artigo em Inglês | MEDLINE | ID: mdl-22069638

RESUMO

The release of tumor necrosis factor α (TNF-α) by ochratoxin A (OTA) was studied in various macrophage and non-macrophage cell lines and compared with E. coli lipopolysaccharide (LPS) as a standard TNF-α release agent. Cells were exposed either to 0, 2.5 or 12.5 µmol/L OTA, or to 0.1 µg/mL LPS, for up to 24 h. OTA at 2.5 µmol/L and LPS at 0.1 µg/mL were not toxic to the tested cells as indicated by viability markers. TNF-α was detected in the incubated cell medium of rat Kupffer cells, peritoneal rat macrophages, and the mouse monocyte macrophage cell line J774A.1: TNF-α concentrations were 1,000 pg/mL, 1,560 pg/mL, and 650 pg/mL, respectively, for 2.5 µmol/L OTA exposure and 3,000 pg/mL, 2,600 pg/mL, and 2,115 pg/mL, respectively, for LPS exposure. Rat liver sinusoidal endothelial cells, rat hepatocytes, human HepG2 cells, and mouse L929 cells lacked any cytokine response to OTA, but showed a significant release of TNF-α after LPS exposure, with the exception of HepG2 cells. In non-responsive cell lines, OTA lacked both any activation of NF-κB or the translocation of activated NF-κB to the cell nucleus, i.e., in mouse L929 cells. In J774A.1 cells, OTA mediated TNF-α release via the pRaf/MEK 1/2-NF-κB and p38-NF-κB pathways, whereas LPS used pRaf/MEK 1/2-NF-κB, but not p38-NF-κB pathways. In contrast, in L929 cells, LPS used other pathways to activate NF-κB. Our data indicate that only macrophages and macrophage derived cells respond to OTA and are considered as sources for TNF-α release upon OTA exposure.


Assuntos
Lipopolissacarídeos/toxicidade , Ocratoxinas/toxicidade , Fator de Necrose Tumoral alfa/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Interações Medicamentosas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos , Ratos , Ratos Wistar
18.
Mol Nutr Food Res ; 53(4): 460-6, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19156713

RESUMO

We studied the inhibitory effect of silibinin on ochratoxin A (OTA) and LPS-mediated tumor necrosis factor alpha (TNF-alpha) release and the leakage of cytotoxic markers glutamate dehydrogenase (GLDH) and lactate dehydrogenase (LDH), from isolated blood-free perfused rat livers, and from isolated pure rat Kupffer cells. In the recirculation perfusion model at the end point 90 min, 2.5 micromol/L OTA released 2600 pg/mL TNF-alpha without effects on liver vitality. LPS at 0.1 microg/mL induced 3000 pg TNF-alpha/mL with slight leakage of GLDH and LDH. Under similar experimental conditions, the addition of silibinin 10 min prior to OTA and LPS showed dose-dependent protection against OTA or LPS-induced hepatic TNF-alpha release. High-dose of silibinin (12.5 microg/mL) also completely restored GLDH and LDH levels in the perfusate. Pretreatment of isolated Kupffer cells with 0.02, 0.1, 0.5, 2.5, and 12.5 microg silibinin/mL 30 min prior to OTA reduced OTA-induced TNF-alpha levels to 90, 70, 25, 25, and 25% at 4 h, respectively, and abrogated any TNF-alpha release at 24 h. Similarly, in the presence of silibinin LPS-induced TNF-alpha levels decreased at 4 h to 71, 57, 18, 22, and 18%, respectively. However, after 24 h of LPS exposition the protection by silibinin vanished and TNF-alpha partially recurred into the incubation medium under LPS. In summary, silibinin had hepatoprotective effects against OTA- or LPS-mediated TNF-alpha release and also reduced the cytotoxicity of both toxins. Isolated Kupffer cells were even more sensitive to the protective effect than perfused livers and responded to very low concentrations of silibinin with a strong inhibition of toxins-mediated TNF-alpha release.


Assuntos
Células de Kupffer/efeitos dos fármacos , Fígado/efeitos dos fármacos , Micotoxinas/toxicidade , Ocratoxinas/toxicidade , Silybum marianum/química , Fator de Necrose Tumoral alfa/metabolismo , Animais , Relação Dose-Resposta a Droga , Células de Kupffer/metabolismo , Lipopolissacarídeos/toxicidade , Fígado/metabolismo , Masculino , NF-kappa B/metabolismo , Perfusão , Ratos , Ratos Wistar , Silibina , Silimarina/farmacologia
19.
Biochem Biophys Res Commun ; 361(1): 26-32, 2007 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-17632081

RESUMO

Here we report the identification, cloning, and characterization of SLC10A5, which is a new member of Solute Carrier Family 10 (SLC10), also known as the "sodium/bile acid cotransporter family". Expression of SLC10A5/Slc10a5 was examined by quantitative real-time PCR and revealed its highest expression levels in liver and kidney in humans, rat and mouse. In rat liver and kidney, Slc10a5 expression was localized by in situ hybridization to hepatocytes and proximal tubules, respectively. A SLC10A5-FLAG fusion protein was expressed in HEK293 cells and showed an apparent molecular weight of 42 kDa after immunoprecipitation. When expressed in Xenopus laevis oocytes, the SLC10A5-FLAG protein was detected in the oocyte's plasma membrane but showed no transport activity for taurocholate, cholate, estrone-3-sulfate, or dehydroepiandrosterone sulfate. As bile acid carriers are the most related carriers to SLC10A5 though, we strongly suppose that SLC10A5 is an orphan carrier with yet non-identified substrates.


Assuntos
Rim/metabolismo , Fígado/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Simportadores/metabolismo , Sequência de Aminoácidos , Animais , Clonagem Molecular , Expressão Gênica , Humanos , Camundongos , Dados de Sequência Molecular , Oócitos/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/química , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Ratos , Simportadores/química , Simportadores/genética , Xenopus laevis
20.
J Biol Chem ; 282(27): 19728-41, 2007 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-17491011

RESUMO

We have cloned human sodium-dependent organic anion transporter (SOAT) cDNA, which consists of 1502 bp and encodes a 377-amino acid protein. SOAT shows 42% sequence identity to the ileal apical sodium-dependent bile acid transporter ASBT and 33% sequence identity to the hepatic Na(+)/taurocholate-cotransporting polypeptide NTCP. Immunoprecipitation of a SOAT-FLAG-tagged protein revealed a glycosylated form at 46 kDa that decreased to 42 kDa after PNGase F treatment. SOAT exhibits a seven-transmembrane domain topology with an outside-to-inside orientation of the N-terminal and C-terminal ends. SOAT mRNA is most highly expressed in testis. Relatively high SOAT expression was also detected in placenta and pancreas. We established a stable SOAT-HEK293 cell line that showed sodium-dependent transport of dehydroepiandrosterone sulfate, estrone-3-sulfate, and pregnenolone sulfate with apparent K(m) values of 28.7, 12.0, and 11.3 microm, respectively. Although bile acids, such as taurocholic acid, cholic acid, and chenodeoxycholic acid, were not substrates of SOAT, the sulfoconjugated bile acid taurolithocholic acid-3-sulfate was transported by SOAT-HEK293 cells in a sodium-dependent manner and showed competitive inhibition of SOAT transport with an apparent K(i) value of 0.24 mum. Several nonsteroidal organosulfates also strongly inhibited SOAT, including 1-(omega-sulfooxyethyl)pyrene, bromosulfophthalein, 2- and 4-sulfooxymethylpyrene, and alpha-naphthylsulfate. Among these inhibitors, 2- and 4-sulfooxymethylpyrene were competitive inhibitors of SOAT, with apparent K(i) values of 4.3 and 5.5 microm, respectively, and they were also transported by SOAT-HEK293 cells.


Assuntos
Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Sequência de Aminoácidos , Transporte Biológico Ativo/efeitos dos fármacos , Transporte Biológico Ativo/fisiologia , Linhagem Celular , Clonagem Molecular , DNA Complementar/genética , DNA Complementar/metabolismo , Feminino , Humanos , Masculino , Moduladores de Transporte de Membrana/farmacologia , Dados de Sequência Molecular , Especificidade de Órgãos/fisiologia , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Pâncreas/metabolismo , Placenta/metabolismo , Gravidez , Homologia de Sequência de Aminoácidos , Simportadores/genética , Testículo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA