Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 726: 150269, 2024 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-38909533

RESUMEN

Mitochondrial dysfunction is implicated in a wide range of human disorders including many neurodegenerative and cardiovascular diseases, metabolic diseases, cancers, and respiratory disorders. Studies have suggested the potential of l-ergothioneine (ET), a unique dietary thione, to prevent mitochondrial damage and improve disease outcome. Despite this, no studies have definitively demonstrated uptake of ET into mitochondria. Moreover, the expression of the known ET transporter, OCTN1, on the mitochondria remains controversial. In this study, we utilise mass spectrometry to demonstrate direct ET uptake in isolated mitochondria as well as its presence in mitochondria isolated from ET-treated cells and animals. Mitochondria isolated from OCTN1 knockout mice tissues, have impaired but still detectable ET uptake, raising the possibility of alternative transporter(s) which may facilitate ET uptake into the mitochondria. Our data confirm that ET can enter mitochondria, providing a basis for further work on ET in the prevention of mitochondrial dysfunction in human disease.


Asunto(s)
Ergotioneína , Ratones Noqueados , Mitocondrias , Ergotioneína/metabolismo , Ergotioneína/farmacología , Animales , Mitocondrias/metabolismo , Humanos , Ratones , Proteínas de Transporte de Catión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/genética , Simportadores/metabolismo , Simportadores/genética
2.
Int J Mol Sci ; 25(16)2024 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-39201429

RESUMEN

OCTN1 and OCTN2 are membrane transport proteins encoded by the SLC22A4 and SLC22A5 genes, respectively. Even though several transcripts have been predicted by bioinformatics for both genes, only one functional protein isoform has been described for each of them. Both proteins are ubiquitous, and depending on the physiopathological state of the cell, their expression is regulated by well-known transcription factors, although some aspects have been neglected. A plethora of missense variants with uncertain clinical significance are reported both in the dbSNP and the Catalogue of Somatic Mutations in Cancer (COSMIC) databases for both genes. Due to their involvement in human pathologies, such as inflammatory-based diseases (OCTN1/2), systemic primary carnitine deficiency (OCTN2), and drug disposition, it would be interesting to predict the impact of variants on human health from the perspective of precision medicine. Although the lack of a 3D structure for these two transport proteins hampers any speculation on the consequences of the polymorphisms, the already available 3D structures for other members of the SLC22 family may provide powerful tools to perform structure/function studies on WT and mutant proteins.


Asunto(s)
Regulación de la Expresión Génica , Miembro 5 de la Familia 22 de Transportadores de Solutos , Humanos , Miembro 5 de la Familia 22 de Transportadores de Solutos/genética , Miembro 5 de la Familia 22 de Transportadores de Solutos/metabolismo , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/química , Conformación Proteica , Simportadores/genética , Simportadores/metabolismo , Simportadores/química
3.
Neurochem Res ; 47(9): 2513-2521, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35788879

RESUMEN

Ergothioneine (ERGO) is a thiol contained in the food that exhibits an excellent antioxidant effect similar to that of glutathione. Although mammals lack a biosynthetic pathway for ERGO, the carnitine/organic cation transporter OCTN1/SLC22A4, which transports ERGO in vivo, is expressed throughout the body, and ERGO is distributed to various organs after oral intake. ERGO is a stable compound that remains in the body for a long time after ingestion. OCTN1 is also expressed in brain parenchymal cells, including neurons, and ERGO in the blood permeates the blood-brain barrier and is distributed to the brain, exhibiting a neuroprotective effect. Recently, the association between central nervous system (CNS) diseases and ERGO has become a research focus. ERGO concentrations in the blood components are lower in patients with cognitive impairment, Parkinson's disease, and frailty than in healthy subjects. ERGO exerts a protective effect against various neurotoxins and improves the symptoms of cognitive impairment, depression, and epilepsy in animal models. The promotion of neurogenesis and induction of neurotrophic factors, in addition to the antioxidant and anti-inflammatory effects, may be involved in the neuroprotective effect of ERGO. This review shows the association between ERGO and CNS diseases, discusses the possible biomarkers of peripheral ERGO in CNS diseases, and the possible preventive and improvement effects of ERGO on CNS diseases.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Ergotioneína , Fármacos Neuroprotectores , Simportadores , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Ergotioneína/farmacología , Humanos , Mamíferos/metabolismo , Proteínas de la Membrana/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Proteínas de Transporte de Catión Orgánico/metabolismo
4.
Int J Mol Sci ; 23(14)2022 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-35887262

RESUMEN

Bivalve molluscs are filter-feeding organisms that can accumulate paralytic shellfish toxins (PST) through ingesting toxic marine dinoflagellates. While the effects of PST accumulation upon the physiology of bivalves have been documented, the underlying molecular mechanism remains poorly understood. In this study, transcriptomic analysis was performed in the gills of Zhikong scallop (Chlamys farreri) after 1, 3, 5, 10, and 15 day(s) exposure of PST-producing dinoflagellate Alexandrium minutum. Higher numbers of differentially expressed genes (DEGs) were detected at day 1 (1538) and day 15 (989) than that at day 3 (77), day 5 (82), and day 10 (80) after exposure, and most of the DEGs were only regulated at day 1 or day 15, highlighting different response mechanisms of scallop to PST-producing dinoflagellate at different stages of exposure. Functional enrichment results suggested that PST exposure induced the alterations of nervous system development processes and the activation of xenobiotic metabolism and substance transport processes at the acute and chronic stages of exposure, respectively, while the immune functions were inhibited by PST and might ultimately cause the activation of apoptosis. Furthermore, a weighted gene co-expression network was constructed, and ten responsive modules for toxic algae exposure were identified, among which the yellow module was found to be significantly correlated with PST content. Most of the hub genes in the yellow module were annotated as solute carriers (SLCs) with eight being OCTN1s, implying their dominant roles in regulating PST accumulation in scallop gills. Overall, our results reveal the gene set responding to and involved in PST accumulation in scallop gills, which will deepen our understanding of the molecular mechanism of bivalve resistance to PST.


Asunto(s)
Bivalvos , Dinoflagelados , Pectinidae , Animales , Bivalvos/genética , Dinoflagelados/genética , Dinoflagelados/metabolismo , Branquias , Toxinas Marinas/toxicidad , Pectinidae/genética , Transcriptoma
5.
Clin Oral Investig ; 25(2): 701-709, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32964310

RESUMEN

OBJECTIVE: SLC22A4/5 single nucleotide polymorphisms (SNPs) have been reported to affect inflammatory diseases. We report the relationship of these polymorphisms with adiposity and tooth loss as elucidated in a 10-year follow-up study. METHODS: Participants of the Study of Health in Pomerania (SHIP, N = 4105) were genotyped for the polymorphisms c.1507C > T in SLC22A4 (rs1050152) and -207C > G in SLC22A5 (rs2631367) using allele-specific real-time PCR assays. A total of 1817 subjects, 934 female and 883 male aged 30-80 years, underwent follow-up 10 years later (SHIP-2) and were assessed for adiposity and tooth loss. RESULTS: The frequencies of the rarer SLC22A4 TT and SLC22A5 CC alleles were 16.7% and 20.3%, respectively. In women, tooth loss was associated with genotype TT vs. CC with incidence rate ratio IRR = 0.74 (95%C.I. 0.60-0.92) and CC vs. GG IRR = 0.79 (0.65-0.96) for SLC22A4 and SLC22A5 SNPs, respectively. In men, no such associations were observed. In the follow-up examination, the relationship between tooth loss and these SNPs was in parallel with measures of body shape such as BMI, body weight, waist circumference, or body fat accumulation. The association between muscle strength and body fat mass was modified by the genotypes studied. CONCLUSIONS: SLC22A4 c.150C > T and SLC22A5 -207C > G polymorphisms are associated with tooth loss and markers of body shape in women but not in men. CLINICAL RELEVANCE: Tooth loss may be related to obesity beyond inflammatory mechanisms, conceivably with a genetic background.


Asunto(s)
Carnitina , Pérdida de Diente , Adiposidad/genética , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Estudios de Seguimiento , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Obesidad/genética , Proteínas de Transporte de Catión Orgánico/genética , Polimorfismo de Nucleótido Simple , Miembro 5 de la Familia 22 de Transportadores de Solutos , Simportadores/genética , Pérdida de Diente/genética
6.
Int J Mol Sci ; 22(24)2021 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-34948211

RESUMEN

Several SLC22 transporters in the human kidney and other tissues are thought to regulate endogenous small antioxidant molecules such as uric acid, ergothioneine, carnitine, and carnitine derivatives. These transporters include those from the organic anion transporter (OAT), OCTN/OCTN-related, and organic cation transporter (OCT) subgroups. In mammals, it has been difficult to show a clear in vivo role for these transporters during oxidative stress. Ubiquitous knockdowns of related Drosophila SLC22s-including transporters homologous to those previously identified by us in mammals such as the "Fly-Like Putative Transporters" FLIPT1 (SLC22A15) and FLIPT2 (SLC22A16)-have shown modest protection against oxidative stress. However, these fly transporters tend to be broadly expressed, and it is unclear if there is an organ in which their expression is critical. Using two tissue-selective knockdown strategies, we were able to demonstrate much greater and longer protection from oxidative stress compared to previous whole fly knockdowns as well as both parent and WT strains (CG6126: p < 0.001, CG4630: p < 0.01, CG16727: p < 0.0001 and CG6006: p < 0.01). Expression in the Malpighian tubule and likely other tissues as well (e.g., gut, fat body, nervous system) appear critical for managing oxidative stress. These four Drosophila SLC22 genes are similar to human SLC22 transporters (CG6126: SLC22A16, CG16727: SLC22A7, CG4630: SLC22A3, and CG6006: SLC22A1, SLC22A2, SLC22A3, SLC22A6, SLC22A7, SLC22A8, SLC22A11, SLC22A12 (URAT1), SLC22A13, SLC22A14)-many of which are highly expressed in the kidney. Consistent with the Remote Sensing and Signaling Theory, this indicates an important in vivo role in the oxidative stress response for multiple SLC22 transporters within the fly renal system, perhaps through interaction with SLC22 counterparts in non-renal tissues. We also note that many of the human relatives are well-known drug transporters. Our work not only indicates the importance of SLC22 transporters in the fly renal system but also sets the stage for in vivo studies by examining their role in mammalian oxidative stress and organ crosstalk.


Asunto(s)
Drosophila melanogaster/metabolismo , Riñón/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Estrés Oxidativo/fisiología , Animales , Antioxidantes/metabolismo , Transporte Biológico/fisiología , Humanos , Transducción de Señal/fisiología
7.
Int J Mol Sci ; 21(3)2020 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-32041338

RESUMEN

The effect of cholesterol was investigated on the OCTN1 transport activity measured as [14C]-tetraethylamonium or [3H]-acetylcholine uptake in proteoliposomes reconstituted with native transporter extracted from HeLa cells or the human recombinant OCTN1 over-expressed in E. coli. Removal of cholesterol from the native transporter by MßCD before reconstitution led to impairment of transport activity. A similar activity impairment was observed after treatment of proteoliposomes harboring the recombinant (cholesterol-free) protein by MßCD, suggesting that the lipid mixture used for reconstitution contained some cholesterol. An enzymatic assay revealed the presence of 10 µg cholesterol/mg total lipids corresponding to 1% cholesterol in the phospholipid mixture used for the proteoliposome preparation. On the other way around, the activity of the recombinant OCTN1 was stimulated by adding the cholesterol analogue, CHS to the proteoliposome preparation. Optimal transport activity was detected in the presence of 83 µg CHS/ mg total lipids for both [14C]-tetraethylamonium or [3H]-acetylcholine uptake. Kinetic analysis of transport demonstrated that the stimulation of transport activity by CHS consisted in an increase of the Vmax of transport with no changes of the Km. Altogether, the data suggests a direct interaction of cholesterol with the protein. A further support to this interpretation was given by a docking analysis indicating the interaction of cholesterol with some protein sites corresponding to CARC-CRAC motifs. The observed direct interaction of cholesterol with OCTN1 points to a possible direct influence of cholesterol on tumor cells or on acetylcholine transport in neuronal and non-neuronal cells via OCTN1.


Asunto(s)
Acetilcolina/análisis , Colesterol/farmacología , Proteínas de Transporte de Catión Orgánico/metabolismo , Simportadores/metabolismo , Tetraetilamonio/análisis , Acetilcolina/química , Radioisótopos de Carbono/química , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Simulación del Acoplamiento Molecular , Proteolípidos/análisis , Proteolípidos/química , Tetraetilamonio/química , Tritio/química
8.
J Neurochem ; 148(3): 373-385, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30295925

RESUMEN

Oxaliplatin is widely used as a key drug in the treatment of colorectal cancer. However, its administration is associated with the dose-limiting adverse effect, peripheral neuropathy. Platinum accumulation in the dorsal root ganglion (DRG) is the major mechanism responsible for oxaliplatin-induced neuropathy. Some drug transporters have been identified as platinum complex transporters in kidney or tumor cells, but not yet in DRG. In the present study, we investigated oxaliplatin transporters and their contribution to peripheral neuropathy. We identified 12 platinum transporters expressed in DRG with real-time PCR, and their transiently overexpressing cells were established. After exposure to oxaliplatin, the accumulation of platinum in these overexpressing cells was evaluated using a coupled plasma mass spectrometer. Octn1/2- and Mate1-expressing cells showed the intracellular accumulation of oxaliplatin. In an animal study, peripheral neuropathy developed after the administration of oxaliplatin (4 mg/kg, intravenously, twice a week) to siRNA-injected rats (0.5 nmol, intrathecally, once a week) was demonstrated with the von Frey test. The knockdown of Octn1 in DRG ameliorated peripheral neuropathy, and decreased platinum accumulation in DRG, whereas the knockdown of Octn2 did not. Mate1 siRNA-injected rats developed more severe neuropathy than control rats. These results indicate that Octn1 and Mate1 are involved in platinum accumulation at DRG and oxaliplatin-induced peripheral neuropathy.


Asunto(s)
Antineoplásicos/toxicidad , Ganglios Espinales/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Oxaliplatino/toxicidad , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Animales , Antiportadores/metabolismo , Proteínas Portadoras/metabolismo , Ganglios Espinales/efectos de los fármacos , Células HEK293 , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Células PC12 , Enfermedades del Sistema Nervioso Periférico/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Transportadoras de Solutos , Simportadores
9.
Biochem Biophys Res Commun ; 513(1): 22-27, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-30929922

RESUMEN

Ergothioneine is a biologically important compound that has been shown to be transported by the organic cation transporter novel type 1 (OCTN1). Following this discovery, a variety of alternate functions for OCTN1 have been suggested including an integral function in the extra-neuronal cholinergic system. The present study reaffirms the primacy of ergothioneine over these alternate substrates using natively expressed OCTN1 in HeLa cells. Besides the general transport inhibitors, quinidine, verapamil and pyrilamine no other putative substrate inhibited ergothioneine transport significantly, with only a slight inhibition demonstrated by carnitine. Even compounds structurally similar to ergothioneine failed to inhibit ergothioneine uptake, suggesting high selectivity of OCTN1. Ergothioneine was found to be avidly accumulated even at low concentrations (300 nM) by HeLa cells.


Asunto(s)
Antioxidantes/metabolismo , Ergotioneína/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Simportadores/metabolismo , Células HeLa , Humanos , Especificidad por Sustrato
10.
Respir Res ; 19(1): 68, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29678179

RESUMEN

BACKGROUND: Several inhaled drugs are dependent on organic cation transporters to cross cell membranes. To further evaluate their potential to impact on inhaled drug disposition, the localization of MATE1, P-gp, OCTN1 and OCTN2 were investigated in human lung. METHODS: Transporter proteins were analysed by immunohistochemistry in lung tissue from healthy subjects and COPD patients. Transporter mRNA was analysed by qPCR in lung tissue and in bronchoalveolar lavage (BAL) cells from smokers and non-smokers. RESULTS: We demonstrate for the first time MATE1 protein expression in the lung with localization to the apical side of bronchial and bronchiolar epithelial cells. Interestingly, MATE1 was strongly expressed in alveolar macrophages as demonstrated both in lung tissue and in BAL cells, and in inflammatory cells including CD3 positive T cells. P-gp, OCTN1 and OCTN2 were also expressed in the alveolar epithelial cells and in inflammatory cells including alveolar macrophages. In BAL cells from smokers, MATE1 and P-gp mRNA expression was significantly lower compared to cells from non-smokers whereas no difference was observed between COPD patients and healthy subjects. THP-1 cells were evaluated as a model for alveolar macrophages but did not reflect the transporter expression observed in BAL cells. CONCLUSIONS: We conclude that MATE1, P-gp, OCTN1 and OCTN2 are expressed in pulmonary lung epithelium, in alveolar macrophages and in other inflammatory cells. This is important to consider in the development of drugs treating pulmonary disease as the transporters may impact drug disposition in the lung and consequently affect pharmacological efficacy and toxicity.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Proteínas de Transporte de Catión Orgánico/biosíntesis , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Miembro 5 de la Familia 22 de Transportadores de Solutos/biosíntesis , Células THP-1/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Adulto , Femenino , Expresión Génica , Voluntarios Sanos , Humanos , Inmunidad Celular/fisiología , Pulmón/citología , Pulmón/inmunología , Pulmón/metabolismo , Masculino , Persona de Mediana Edad , Proteínas de Transporte de Catión Orgánico/genética , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Miembro 5 de la Familia 22 de Transportadores de Solutos/genética , Simportadores , Células THP-1/inmunología , Adulto Joven
11.
Neurochem Res ; 43(1): 116-128, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28688036

RESUMEN

Brain immune cells, i.e., microglia, play an important role in the maintenance of brain homeostasis, whereas chronic overactivation of microglia is involved in the development of various neurodegenerative disorders. Therefore, the regulation of microglial activation may contribute to their treatment. The aim of the present study was to clarify the functional expression of carnitine/organic cation transporter OCTN1/SLC22A4, which recognizes the naturally occurring food-derived antioxidant ergothioneine (ERGO) as a substrate in vivo, in microglia and its role in regulation of microglial activation. Primary cultured microglia derived from wild-type mice (WT-microglia) and mouse microglial cell line BV2 exhibited time-dependent uptake of [3H]- or d9-labeled ERGO. The uptake was markedly decreased in cultured microglia from octn1 gene knockout mice (octn1 -/--microglia) and BV2 cells transfected with small interfering RNA targeting the mouse octn1 gene (siOCTN1). These results demonstrate that OCTN1 is functionally expressed in murine microglial cells. Exposure of WT-microglia to ERGO led to a significant decrease in cellular hypertrophy by LPS-stimulation with concomitant attenuation of intracellular reactive oxygen species (ROS), suggesting that OCTN1-mediated ERGO uptake may suppress cellular hypertrophy via the inhibition of ROS production with microglial activation. The expression of mRNA for interleukin-1ß (IL-1ß) after LPS-treatment was significantly increased in octn1 -/--microglia and siOCTN1-treated BV2 cells compared to the control cells. Meanwhile, treatment of ERGO minimally affected the induction of IL-1ß mRNA by LPS-stimulation in cultured microglia and BV2 cells. Thus, OCTN1 negatively regulated the induction of inflammatory cytokine IL-1ß, at least in part, via the transport of unidentified substrates other than ERGO in microglial cells.


Asunto(s)
Transporte Biológico/efectos de los fármacos , Proteínas Portadoras/metabolismo , Ergotioneína/farmacología , Proteínas de la Membrana/metabolismo , Microglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Carnitina , Proteínas Portadoras/genética , Células Cultivadas , Interleucina-1beta/metabolismo , Lipopolisacáridos/farmacología , Proteínas de la Membrana/genética , Ratones , Microglía/metabolismo , Neuronas/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Simportadores
12.
Pharm Res ; 35(11): 224, 2018 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-30280275

RESUMEN

PURPOSE: Solute carrier SLC22A4 encodes the carnitine/organic cation transporter OCTN1 and is associated with inflammatory bowel disease, although little is known about how this gene is linked to pathogenesis. The aim of the present study was to identify endogenous substrates that are associated with gastrointestinal inflammation. METHODS: HEK293/OCTN1 and mock cells were incubated with colon extracts isolated from dextran sodium sulfate-induced colitis mice; the subsequent cell lysates were mixed with the amino group selective reagent 3-aminopyridyl-N-hydroxysuccinimidyl carbamate (APDS), to selectively label OCTN1 substrates. Precursor ion scanning against the fragment ion of APDS was then used to identify candidate OCTN1 substrates. RESULTS: Over 10,000 peaks were detected by precursor ion scanning; m/z 342 had a higher signal in HEK293/OCTN1 compared to mock cells. This peak was detected as a divalent ion that contained four APDS-derived fragments and was identified as spermine. Spermine concentration in peripheral blood mononuclear cells from octn1 gene knockout mice (octn1-/-) was significantly lower than in wild-type mice. Lipopolysaccharide-induced gene expression of inflammatory cytokines in peritoneal macrophages from octn1-/- mice was lower than in wild-type mice. CONCLUSIONS: The combination metabolomics approach can provide a novel tool to identify endogenous substrates of OCTN1.


Asunto(s)
Proteínas de Transporte de Catión Orgánico/metabolismo , Animales , Colitis/inducido químicamente , Colitis/metabolismo , Colon/química , Colon/metabolismo , Citocinas/metabolismo , Células HEK293 , Humanos , Enfermedades Inflamatorias del Intestino/metabolismo , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Metabolómica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Transporte de Catión Orgánico/genética , Espermina/metabolismo , Simportadores , Extractos de Tejidos/farmacología
13.
Zygote ; 26(2): 149-161, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29607799

RESUMEN

SummaryThe objective of the study was to investigate the effect of l-ergothioneine (l-erg) (5 mM or 10 mM) supplementation in maturation medium on the developmental potential and OCTN1-dependant l-erg-mediated (10 mM) change in mRNA abundance of apoptotic (Bcl2, Bax, Casp3 and PCNA) and antioxidant (GPx, SOD1, SOD2 and CAT) genes in sheep oocytes and developmental stages of embryos produced in vitro. Oocytes matured with l-erg (10 mM) reduced their embryo toxicity by decreasing intracellular ROS and increasing intracellular GSH in matured oocytes that in turn improved developmental potential, resulting in significantly (P < 0.05) higher percentages of cleavage (53.72% vs 38.86, 46.56%), morulae (34.36% vs 20.62, 25.84%) and blastocysts (14.83% vs 6.98, 9.26%) compared with other lower concentrations (0 mM and 5 mM) of l-erg without change in maturation rate. l-Erg (10 mM) treatment did not influence the mRNA abundance of the majority of apoptotic and antioxidant genes studied in the matured oocytes and developmental stages of embryo. A gene expression study found that the SLC22A4 gene that encodes OCTN1, an integral membrane protein and specific transporter of l-erg was not expressed in oocytes and developmental stages of embryos. Therefore it was concluded from the study that although there was improvement in the developmental potential of sheep embryos by l-erg supplementation in maturation medium, there was no change in the expression of the majority of the genes studied due to the absence of the SLC22A4 gene in oocytes and embryos that encode OCTN1, which is responsible for transportation of l-erg across the membrane to alter gene expression.


Asunto(s)
Blastocisto/efectos de los fármacos , Ergotioneína/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Oocitos/efectos de los fármacos , Ovinos/embriología , Animales , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Blastocisto/citología , Medios de Cultivo/química , Medios de Cultivo/farmacología , Técnicas de Cultivo de Embriones , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/genética , Femenino , Glutatión/metabolismo , Técnicas de Maduración In Vitro de los Oocitos/métodos , Técnicas de Maduración In Vitro de los Oocitos/veterinaria , Masculino , Mórula , Oocitos/fisiología , Proteínas de Transporte de Catión Orgánico/genética , Testículo/efectos de los fármacos
14.
Acta Endocrinol (Buchar) ; 14(3): 294-299, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-31149274

RESUMEN

CONTEXT: Peripheral nerve lesions are a major complication of diabetes mellitus, the main clinical manifestations of which are numbness and pain involving the limbs. OBJECTIVE: To determine the correlation between pregabalin treatment and diabetic peripheral neuropathic pain. DESIGN: An experimental animal study in BALB/c mice. SUBJECTS AND METHODS: Diabetes models are established by injecting streptozotocin (STZ) into the abdominal cavities of mice. The correlation between the treatment effect, time, and dosage of pregabalin was determined. The effect of a type 1 organic cation transporter (Octn1) in the absorption of pregabalin was evaluated. RESULTS: Pregabalin reduced tactile allodynia in diabetic mice. The best analgesic effect occurred when intestinal absorption was increased. Octn1 mediated pregabalin entry into intestinal epithelial cells, which influenced the absorption of pregabalin with a time-dependent fluctuation in the small intestine. Peripheral nerve damage caused by diabetes was dependent on time and dose of pregabalin, which was related to the regular expression of Octn1 in small intestinal epithelium. CONCLUSIONS: Peripheral nerve damage caused by diabetes was dependent on time and dosage of pregabalin, which was related to the regular expression of Octn1 in small intestinal epithelium.

15.
Biochim Biophys Acta ; 1858(4): 653-60, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26724204

RESUMEN

A suitable experimental tool based on proteoliposomes for assaying Organic Cation Transporter Novel member 1 (OCTN1) of peritoneum was pointed out. OCTN1, recently acknowledged as acetylcholine transporter, was immunodetected in rat peritoneum. Transport was assayed following flux of radiolabelled TEA, acetylcholine or acetylcarnitine in proteoliposomes reconstituted with peritoneum extract. OCTN1 mediated, besides TEA, also acetylcholine and a slower acetylcarnitine transport. External sodium inhibited acetylcholine uptake but not its release from proteoliposomes. Differently, sodium did not affect acetylcarnitine uptake. These results suggested that physiologically, acetylcholine should be released while acetylcarnitine was taken up by peritoneum cells. Transport was impaired by OCTN1 inhibitors, butyrobetaine, spermine, and choline. Biotin was also found as acetylcholine transport inhibitor. Anti-OCTN1 antibody specifically inhibited acetylcholine transport confirming the involvement of OCTN1. The transporter was also immunodetected in human mesothelial primary cells. Extract from these cells was reconstituted in proteoliposomes. Transport features very similar to those found with rat peritoneum were observed. Validation of the proteoliposome model for peritoneal transport study was then achieved assaying transport in intact mesothelial cells. TEA, butyrobetaine and Na(+) inhibited acetylcholine transport in intact cells while efflux was Na(+) insensitive. Therefore transport features in intact cells overlapped those found in proteoliposomes.


Asunto(s)
Acetilcarnitina/metabolismo , Acetilcolina/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Peritoneo/metabolismo , Acetilcarnitina/química , Acetilcolina/química , Animales , Transporte Biológico Activo , Proteínas Portadoras/química , Humanos , Proteínas de la Membrana/química , Proteínas de Transporte de Catión Orgánico , Peritoneo/química , Proteolípidos/química , Proteolípidos/metabolismo , Ratas , Sodio/química , Proteínas Transportadoras de Solutos , Simportadores
16.
Biochim Biophys Acta ; 1858(2): 210-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26607009

RESUMEN

Carnitine plays a physiologically important role in the ß-oxidation of fatty acids, facilitating the transport of long-chain fatty acids across the inner mitochondrial membrane. Distribution of carnitine within the body tissues is mainly performed by novel organic cation transporter (OCTN) family, including the isoforms OCTN1 (SLC22A4) and OCTN2 (SLC22A5) expressed in human. We performed here a characterization of carnitine transport in human airway epithelial cells A549, Calu-3, NCl-H441, and BEAS-2B, by means of an integrated approach combining data of mRNA/protein expression with the kinetic and inhibition analyses of L-[(3)H]carnitine transport. Carnitine uptake was strictly Na(+)-dependent in all cell models. In A549 and BEAS-2B cells, carnitine uptake was mediated by one high-affinity component (Km<2 µM) identifiable with OCTN2. In both these cell models, indeed, carnitine uptake was maximally inhibited by betaine and strongly reduced by SLC22A5/OCTN2 silencing. Conversely, Calu-3 and NCl-H441 exhibited both a high (Km~20 µM) and a low affinity (Km>1 mM) transport component. While the high affinity component is identifiable with OCTN2, the low affinity uptake is mediated by ATB(0,+), a Na(+), and Cl(-)-coupled transport system for neutral and cationic amino acids, as demonstrated by the inhibition by leucine and arginine, as well as by SLC6A14/ATB(0,+) silencing. The presence of this transporter leads to a massive accumulation of carnitine inside the cells and may be of peculiar relevance in pathologic conditions of carnitine deficiency, such as those associated to OCTN2 defects.


Asunto(s)
Carnitina/metabolismo , Células Epiteliales/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Mucosa Respiratoria/metabolismo , Sistemas de Transporte de Aminoácidos , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Transporte Biológico Activo/fisiología , Carnitina/genética , Línea Celular Tumoral , Células Epiteliales/citología , Regulación de la Expresión Génica/fisiología , Humanos , Proteínas de Transporte de Catión Orgánico/genética , Mucosa Respiratoria/citología , Miembro 5 de la Familia 22 de Transportadores de Solutos , Simportadores
17.
Biochim Biophys Acta ; 1860(6): 1334-42, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26994919

RESUMEN

BACKGROUND: Organic cation transporters transfer solutes with a positive charge across the plasma membrane. The novel organic cation transporter 1 (OCTN1) and 2 (OCTN2) transport ergothioneine and carnitine, respectively. Mutations in the SLC22A5 gene encoding OCTN2 cause primary carnitine deficiency, a recessive disorders resulting in low carnitine levels and defective fatty acid oxidation. Variations in the SLC22A4 gene encoding OCTN1 are associated with rheumatoid arthritis and Crohn disease. METHODS: Here we evaluate the functional properties of the OCTN1 transporter using chimeric transporters constructed by fusing different portion of the OCTN1 and OCTN2 cDNAs. Their relative abundance and subcellular distribution was evaluated through western blot analysis and confocal microscopy. RESULTS: Substitutions of the C-terminal portion of OCTN1 with the correspondent residues of OCTN2 generated chimeric OCTN transporters more active than wild-type OCTN1 in transporting ergothioneine. Additional single amino acid substitutions introduced in chimeric OCTN transporters further increased ergothioneine transport activity. Kinetic analysis indicated that increased transport activity was due to an increased V(max), with modest changes in K(m) toward ergothioneine. CONCLUSIONS: Our results indicate that the OCTN1 transporter is tolerant to extensive amino acid substitutions. This is in sharp contrast to the OCTN2 carnitine transporter that has been selected for high functional activity through evolution, with almost all substitutions reducing carnitine transport activity. GENERAL SIGNIFICANCE: The widespread tolerance of OCTN1 to amino acid substitutions suggests that the corresponding SLC22A4 gene may have derived from a recent duplication of the SLC22A5 gene and might not yet have a defined physiological role.


Asunto(s)
Ergotioneína/farmacocinética , Proteínas de Transporte de Catión Orgánico/fisiología , Sustitución de Aminoácidos , Animales , Transporte Biológico , Western Blotting , Células CHO , Cricetulus , Humanos , Microscopía Confocal , Proteínas de Transporte de Catión Orgánico/química , Relación Estructura-Actividad , Simportadores
18.
Biol Pharm Bull ; 40(8): 1146-1152, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28768995

RESUMEN

Dysfunction in neurotransmission mediated by neurotransmitters causes various neurological disorders. Therefore, receptors and reuptake transporters of neurotransmitters have been focused on as a therapeutic target in neurological disorders. These membrane proteins have high affinity for a specific neurotransmitter and are highly expressed on synaptic membranes. In contrast, xenobiotic transporters have relatively lower affinity for neurotransmitters but widely recognize various organic cations and/or anions and are also expressed in brain neurons. However, it has been largely unknown why such xenobiotic transporters are expressed in neurons that play a key role in signal transduction. We have therefore attempted to clarify the physiological roles of one such xenobiotic organic cation transporter (OCT) in neural cells with the aim of obtaining new insight into the treatment of neurological disorders. Carnitine/organic cation transporter OCTN1/SLC22A4 is functionally expressed in neurons and neural stem cells. In particular, OCTN1 is expressed at much higher levels compared with other OCTs in neural stem cells and positively regulates their differentiation into neurons. OCTN1 accepts the naturally occurring food-derived antioxidant ergothioneine (ERGO) as a good in vivo substrate. Because ERGO is highly distributed into the brain after oral ingestion, OCTN1 may contribute to the alleviation of oxidative stress and promotion of neuronal differentiation via the uptake of ERGO in the brain, perhaps abating symptoms of neurological disorders. In this review, we introduce current topics on the physiological roles of OCTs with a focus on OCTN1 in neural cells and discuss its possible application to the treatment of neurological disorders.


Asunto(s)
Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Animales , Carnitina/metabolismo , Humanos , Enfermedades del Sistema Nervioso/metabolismo
19.
Korean J Physiol Pharmacol ; 21(1): 11-17, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28066136

RESUMEN

Crohn's disease (CD) is a chronic inflammatory bowel disease with multifactorial causes including environmental and genetic factors. Several studies have demonstrated that the organic cation/carnitine transporter 1 (OCTN1) non-synonymous variant L503F is associated with susceptibility to CD. However, it was reported that L503F is absent in Asian populations. Previously, we identified and functionally characterized genetic variants of the OCTN1 promoter region in Koreans. In that study, four variants demonstrated significant changes in promoter activity. In the present study, we determined whether four functional variants of the OCTN1 promoter play a role in the susceptibility to or clinical course of CD in Koreans. To examine it, the frequencies of the four variants of the OCTN1 promoter were determined by genotyping using DNA samples from 194 patients with CD and 287 healthy controls. Then, associations between genetic variants and the susceptibility to CD or clinical course of CD were evaluated. We found that susceptibility to CD was not associated with OCTN1 functional promoter variants or haplotypes showing altered promoter activities in in vitro assays. However, OCTN1 functional promoter haplotypes showing decreased promoter activities were significantly associated with a penetrating behavior in CD patients (HR=2.428, p=0.009). Our results suggest that the OCTN1 functional promoter haplotypes can influence the CD phenotype, although these might not be associated with susceptibility to this disease.

20.
Biochem Biophys Res Commun ; 470(2): 245-250, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26772879

RESUMEN

Ergothioneine (ET) is a diet-derived, thiolated derivative of histidine with antioxidant properties. Although ET is produced only by certain fungi and bacteria, it can be found at high concentrations in certain human and animal tissues and is absorbed through a specific, high affinity transporter (OCTN1). In liver, heart, joint and intestinal injury, elevated ET concentrations have been observed in injured tissues. The physiological role of ET remains unclear. We thus review current literature to generate a specific hypothesis: that the accumulation of ET in vivo is an adaptive mechanism, involving the regulated uptake and concentration of an exogenous natural compound to minimize oxidative damage.


Asunto(s)
Antioxidantes/metabolismo , Citoprotección , Ergotioneína/metabolismo , Inflamación/metabolismo , Adaptación Fisiológica , Animales , Humanos , Especificidad de Órganos , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA