Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Amino Acids ; 52(3): 499-503, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32008093

RESUMEN

A direct inhibiting effect of NO on the function of CAT-1 and -2A has been postulated to occur via nitrosylation of cysteine residues in the transporters. Neither the NO donor SNAP nor a mixture of SIN-1 and Spermine NONOate, that generates the strong nitrosating agent N2O3, reduced CAT-mediated L-arginine transport. Direct nitros(yl)ation does either not occur in CATs or does not affect their transport function. A regulatory effect of NO or nitrosating agents on CAT-mediated transport under physiological conditions seems, therefore, unlikely.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Transportador de Aminoácidos Catiónicos 1/metabolismo , Cisteína/metabolismo , Óxido Nítrico/metabolismo , Animales , Arginina/metabolismo , Humanos , Oocitos , Xenopus laevis
2.
Eur J Immunol ; 46(1): 92-103, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26449889

RESUMEN

Availability of the semiessential amino acid arginine is fundamental for the efficient function of human T lymphocytes. Tumor-associated arginine deprivation, mainly induced by myeloid-derived suppressor cells, is a central mechanism of tumor immune escape from T-cell-mediated antitumor immune responses. We thus assumed that transmembranous transport of arginine must be crucial for T-cell function and studied which transporters are responsible for arginine influx into primary human T lymphocytes. Here, we show that activation via CD3 and CD28 induces arginine transport into primary human T cells. Both naïve and memory CD4(+) T cells as well as CD8(+) T cells specifically upregulated the human cationic amino acid transporter-1 (hCAT-1), with an enhanced and persistent expression under arginine starvation. When hCAT-1 induction was suppressed via siRNA transfection, arginine uptake, and cellular proliferation were impaired. In summary, our results demonstrate that hCAT-1 is a key component of efficient T-cell activation and a novel potential target structure to modulate adaptive immune responses in tumor immunity or inflammation.


Asunto(s)
Arginina/metabolismo , Transportador de Aminoácidos Catiónicos 1/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Western Blotting , Transportador de Aminoácidos Catiónicos 1/metabolismo , Proliferación Celular , Células Cultivadas , Humanos , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/metabolismo , Transfección
3.
Arterioscler Thromb Vasc Biol ; 36(1): 78-85, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26586660

RESUMEN

OBJECTIVE: The present study was conducted to investigate the contribution of perivascular adipose tissue (PVAT) to vascular dysfunction in a mouse model of diet-induced obesity. APPROACH AND RESULTS: Obesity was induced in male C57BL/6J mice with a high-fat diet for 20 weeks, and vascular function was studied with myograph. In PVAT-free aortas isolated from obese mice, the endothelium-dependent, nitric oxide-mediated vasodilator response to acetylcholine remained normal. In contrast, a clear reduction in the vasodilator response to acetylcholine was observed in aortas from obese mice when PVAT was left in place. Adipocytes in PVAT were clearly positive in endothelial nitric oxide synthase (eNOS) staining, and PVAT nitric oxide production was significantly reduced in obese mice. High-fat diet had no effect on eNOS expression but led to eNOS uncoupling, evidenced by diminished superoxide production in PVAT after eNOS inhibition. As mechanisms for eNOS uncoupling, arginase induction and l-arginine deficiency were observed in PVAT. Obesity-induced vascular dysfunction could be reversed by ex vivo l-arginine treatment and arginase inhibition. CONCLUSIONS: Diet-induced obesity leads to l-arginine deficiency and eNOS uncoupling in PVAT. The combination therapy with l-arginine and arginase inhibitors may represent a novel therapeutic strategy for obesity-induced vascular disease.


Asunto(s)
Tejido Adiposo/enzimología , Aorta Torácica/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Obesidad/enzimología , Vasodilatación , Adipocitos/enzimología , Adipoquinas/metabolismo , Tejido Adiposo/fisiopatología , Adiposidad , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiopatología , Arginasa/antagonistas & inhibidores , Arginasa/metabolismo , Arginina/deficiencia , Citocinas/metabolismo , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Obesidad/fisiopatología , Comunicación Paracrina , Fosforilación , Transducción de Señal , Superóxidos/metabolismo , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
4.
Amino Acids ; 48(2): 427-36, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26403849

RESUMEN

Asymmetric dimethyl L-arginine (ADMA) is generated within cells and in mitochondria when proteins with dimethylated arginine residues are degraded. The aim of this study was to identify the carrier protein(s) that transport ADMA across the inner mitochondrial membrane. It was found that the recombinant, purified mitochondrial solute carrier SLC25A2 when reconstituted into liposomes efficiently transports ADMA in addition to its known substrates arginine, lysine, and ornithine and in contrast to the other known mitochondrial amino acid transporters SLC25A12, SLC25A13, SLC25A15, SLC25A18, SLC25A22, and SLC25A29. The widely expressed SLC25A2 transported ADMA across the liposomal membrane in both directions by both unidirectional transport and exchange against arginine or lysine. The SLC25A2-mediated ADMA transport followed first-order kinetics, was nearly as fast as the transport of the best SLC25A2 substrates known so far, and was highly specific as symmetric dimethylarginine (SDMA) was not transported at all. Furthermore, ADMA inhibited SLC25A2 activity with an inhibition constant of 0.38 ± 0.04 mM, whereas SDMA inhibited it poorly. We propose that a major function of SLC25A2 is to export ADMA from mitochondria missing the mitochondrial ADMA-metabolizing enzyme AGXT2. There is evidence that ADMA can also be imported into mitochondria, e.g., in kidney proximal tubulus cells, to be metabolized by AGXT2. SLC25A2 may also mediate this transport function.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Arginina/análogos & derivados , Proteínas Portadoras/metabolismo , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Arginina/metabolismo , Transporte Biológico , Línea Celular Tumoral , Humanos , Túbulos Renales Proximales/metabolismo , Transaminasas
5.
Amino Acids ; 47(2): 335-44, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25385314

RESUMEN

O-(2-fluoroethyl)-L-tyrosine (FET) labeled with fluorine-18 is an important and specific tracer for diagnostics of glioblastoma via positron emission tomography (PET). However, the mechanism of its quite specific accumulation in tumor tissue has not been understood so far. In this work we demonstrate that [(3)H]L-tyrosine is primarily transported by the system L transporter LAT1 in human LN229 glioblastoma cells. FET reduced tyrosine transport, suggesting that it shares the same uptake pathway. More importantly, accumulation of FET was significantly reduced after siRNA-mediated downregulation of LAT1. Xenopus laevis oocytes expressing human LAT1 together with the glycoprotein 4F2hc (necessary to pull LAT-1 to the plasma membrane) exhibited a similar accumulation of FET as observed in glioblastoma cells. In contrast, no accumulation was observed in control oocytes, not overexpressing an exogenous transporter. Because LAT1 works exclusively as an exchanger of amino acids, substrates at one side of the membrane stimulate exchange against substrates at the other side. Extracellular FET stimulated the efflux of intracellular [(3)H]L-leucine, demonstrating that FET is indeed an influx substrate for LAT1. However, FET injected into oocytes was not able to stimulate uptake of extracellular [(3)H]L-leucine, indicating that FET is not a good efflux substrate. Our data, therefore, suggest that FET is trapped within cells due to the asymmetry of its intra- and extracellular recognition by LAT1. If also found for other transporters in tumor cells, asymmetric substrate recognition may be further exploited for tumor-specific accumulation of PET-tracers and/or other tumor-related drugs.


Asunto(s)
Glioblastoma/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Proteínas de Neoplasias/metabolismo , Tirosina/análogos & derivados , Animales , Línea Celular Tumoral , Medios de Contraste , Cadena Pesada de la Proteína-1 Reguladora de Fusión/genética , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Glioblastoma/diagnóstico por imagen , Glioblastoma/genética , Humanos , Transportador de Aminoácidos Neutros Grandes 1/genética , Proteínas de Neoplasias/genética , Tomografía de Emisión de Positrones , Radiografía , Tirosina/farmacocinética , Tirosina/farmacología , Xenopus laevis
6.
Amino Acids ; 47(12): 2647-58, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26215737

RESUMEN

Cationic amino acid transporters (CATs) mediate the entry of L-type cationic amino acids (arginine, ornithine and lysine) into the cells including neurons. CAT-3, encoded by the SLC7A3 gene on chromosome X, is one of the three CATs present in the human genome, with selective expression in brain. SLC7A3 is highly intolerant to variation in humans, as attested by the low frequency of deleterious variants in available databases, but the impact on variants in this gene in humans remains undefined. In this study, we identified a missense variant in SLC7A3, encoding the CAT-3 cationic amino acid transporter, on chromosome X by exome sequencing in two brothers with autism spectrum disorder (ASD). We then sequenced the SLC7A3 coding sequence in 148 male patients with ASD and identified three additional rare missense variants in unrelated patients. Functional analyses of the mutant transporters showed that two of the four identified variants cause severe or moderate loss of CAT-3 function due to altered protein stability or abnormal trafficking to the plasma membrane. The patient with the most deleterious SLC7A3 variant had high-functioning autism and epilepsy, and also carries a de novo 16p11.2 duplication possibly contributing to his phenotype. This study shows that rare hypomorphic variants of SLC7A3 exist in male individuals and suggest that SLC7A3 variants possibly contribute to the etiology of ASD in male subjects in association with other genetic factors.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/genética , Trastorno del Espectro Autista/genética , Secuencia de Aminoácidos , Animales , Biotinilación , Encéfalo/metabolismo , Membrana Celular/metabolismo , Niño , Cromosomas Humanos X/genética , Epilepsia/complicaciones , Epilepsia/genética , Frecuencia de los Genes , Humanos , Pérdida de Heterocigocidad , Masculino , Conformación Molecular , Datos de Secuencia Molecular , Mutación , Mutación Missense , Oocitos/metabolismo , Linaje , Fenotipo , Xenopus laevis
7.
J Biol Chem ; 288(42): 30411-30419, 2013 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-24019517

RESUMEN

In most cells, cationic amino acids such as l-arginine, l-lysine, and l-ornithine are transported by cationic (CAT) and y(+)L (y(+)LAT) amino acid transporters. In human erythrocytes, the cysteine-modifying agent N-ethylmaleimide (NEM) has been shown to inhibit system y(+) (most likely CAT-1), but not system y(+)L (Devés, R., Angelo, S., and Chávez, P. (1993) J. Physiol. 468, 753-766). We thus wondered if sensitivity to NEM distinguishes generally all CAT and y(+)LAT isoforms. Transport assays in Xenopus laevis oocytes established that indeed all human CATs (including the low affinity hCAT-2A), but neither y(+)LAT isoform, are inhibited by NEM. hCAT-2A inhibition was not due to reduced transporter expression in the plasma membrane, indicating that NEM reduces the intrinsic transporter activity. Individual mutation of each of the seven cysteine residues conserved in all CAT isoforms did not lead to NEM insensitivity of hCAT-2A. However, a cysteine-less mutant was no longer inhibited by NEM, suggesting that inhibition occurs through modification of more than one cysteine in hCAT-2A. Indeed, also the double mutant C33A/C273A was insensitive to NEM inhibition, whereas reintroduction of a cysteine at either position 33 or 273 in the cysteine-less mutant led to NEM sensitivity. We thus identified Cys-33 and Cys-273 in hCAT-2A as the targets of NEM inhibition. In addition, all proteins with Cys-33 mutations showed a pronounced reduction in transport activity, suggesting that, surprisingly, this residue, located in the cytoplasmic N terminus, is important for transporter function.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/antagonistas & inhibidores , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Cisteína/metabolismo , Inhibidores Enzimáticos/farmacología , Etilmaleimida/farmacología , Sustitución de Aminoácidos , Sistemas de Transporte de Aminoácidos Básicos/química , Animales , Cisteína/química , Cisteína/genética , Inhibidores Enzimáticos/química , Etilmaleimida/química , Humanos , Mutación Missense , Oocitos , Estructura Terciaria de Proteína , Xenopus laevis
8.
J Biol Chem ; 287(36): 30853-60, 2012 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-22787143

RESUMEN

In human skin fibroblasts, a lysosomal transport system specific for cationic amino acids has been described and named system c. We asked if SLC7A14 (solute carrier family 7 member A14), an orphan protein assigned to the SLC7 subfamily of cationic amino acid transporters (CATs) due to sequence homology, may represent system c. Fusion proteins between SLC7A14 and enhanced GFP localized to intracellular vesicles, co-staining with the lysosomal marker LysoTracker(®). To perform transport studies, we first tried to redirect SLC7A14 to the plasma membrane (by mutating putative lysosomal targeting motifs) but without success. We then created a chimera carrying the backbone of human (h) CAT-2 and the protein domain of SLC7A14 corresponding to the so-called "functional domain" of the hCAT proteins, a protein stretch of 81 amino acids that determines the apparent substrate affinity, sensitivity to trans-stimulation, and (as revealed in this study) pH dependence. The chimera mediated arginine transport and exhibited characteristics similar but not identical to hCAT-2A (the low affinity hCAT-2 isoform). Western blot and microscopic analyses confirmed localization of the chimera in the plasma membrane of Xenopus laevis oocytes. Noticeably, arginine transport by the hCAT-2/SLC7A14 chimera was pH-dependent, trans-stimulated, and inhibited by α-trimethyl-L-lysine, properties assigned to lysosomal transport system c in human skin fibroblasts. Expression analysis showed strong expression of SLC7A14 mRNA in these cells. Taken together, these data strongly suggest that SLC7A14 is a lysosomal transporter for cationic amino acids.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Arginina/metabolismo , Fibroblastos/metabolismo , Lisosomas/metabolismo , Piel/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/genética , Animales , Arginina/genética , Transporte Biológico Activo/fisiología , Línea Celular , Fibroblastos/citología , Humanos , Concentración de Iones de Hidrógeno , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Piel/citología , Xenopus laevis
9.
Am J Physiol Renal Physiol ; 305(12): F1645-55, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24107421

RESUMEN

Cystinuria is an autosomal recessive disease caused by mutations in SLC3A1 (rBAT) and SLC7A9 (b(0,+)AT). Gene targeting of the catalytic subunit (Slc7a9) in mice leads to excessive excretion of cystine, lysine, arginine, and ornithine. Here, we studied this non-type I cystinuria mouse model using gene expression analysis, Western blotting, clearance, and brush-border membrane vesicle (BBMV) uptake experiments to further characterize the renal and intestinal consequences of losing Slc7a9 function. The electrogenic and BBMV flux studies in the intestine suggested that arginine and ornithine are transported via other routes apart from system b(0,+). No remarkable gene expression changes were observed in other amino acid transporters and the peptide transporters in the intestine and kidney. Furthermore, the glomerular filtration rate (GFR) was reduced by 30% in knockout animals compared with wild-type animals. The fractional excretion of arginine was increased as expected (∼100%), but fractional excretions of lysine (∼35%), ornithine (∼16%), and cystine (∼11%) were less affected. Loss of function of b(0,+)AT reduced transport of cystine and arginine in renal BBMVs and completely abolished the exchanger activity of dibasic amino acids with neutral amino acids. In conclusion, loss of Slc7a9 function decreases the GFR and increases the excretion of several amino acids to a lesser extent than expected with no clear regulation at the mRNA and protein level of alternative transporters and no increased renal epithelial uptake. These observations indicate that transporters located in distal segments of the kidney and/or metabolic pathways may partially compensate for Slc7a9 loss of function.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/deficiencia , Aminoácidos Diaminos/metabolismo , Cistina/metabolismo , Cistinuria/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/genética , Animales , Cistinuria/fisiopatología , Modelos Animales de Enfermedad , Tasa de Filtración Glomerular/fisiología , Riñón/metabolismo , Riñón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
10.
Biochem Biophys Res Commun ; 423(2): 218-23, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22609206

RESUMEN

A 37-year old male patient presented with frequent angina attacks (up to 40/day) largely resistant to classical vasodilator therapy. The patient showed severe coronary and peripheral endothelial dysfunction, increased platelet aggregation and increased platelet-derived superoxide production. The endothelial nitric oxide synthase (eNOS)-inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME) reduced superoxide formation in platelets identifying "uncoupled" eNOS as a superoxide source. Oral L-arginine normalized coronary and peripheral endothelial dysfunction and reduced platelet aggregation and eNOS-derived superoxide production. Plasma concentrations of the endogenous NOS inhibitor asymmetric dimethyl-L-arginine (ADMA), representing an independent risk factor for cardiovascular disease, were normal in the patient. However, immediately after oral administration of cationic amino acid (CAA), plasma ADMA levels rose markedly, demonstrating increased ADMA efflux from intracellular stores. ADMA efflux from mononuclear cells of the patient was accelerated by CAA, but not neutral amino acids (NAA) demonstrating impairment of y(+)LAT (whose expression was found reduced in these cells). These data suggest that impairment of y(+)LAT may cause intracellular (endothelial) ADMA accumulation leading to systemic endothelial dysfunction. This may represent a novel mechanism underlying vasospastic angina and vascular dysfunction in general. Moreover, these new findings contribute to the understanding of the l-arginine paradox, the improvement of eNOS activity by oral L-arginine despite sufficient cellular l-arginine levels to ensure proper function of this enzyme.


Asunto(s)
Angina de Pecho/metabolismo , Arginina/análogos & derivados , Vasoespasmo Coronario/metabolismo , Endotelio Vascular/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Adulto , Angina de Pecho/sangre , Angina de Pecho/tratamiento farmacológico , Arginina/administración & dosificación , Arginina/sangre , Arginina/metabolismo , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Vasoespasmo Coronario/sangre , Vasoespasmo Coronario/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Humanos , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Superóxidos/metabolismo
11.
J Immunol ; 184(11): 6025-34, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20421643

RESUMEN

Dendritic cells (DCs) are the most potent APCs of the immune system. Understanding the intercellular and intracellular signaling processes that lead to DC maturation is critical for determining how these cells initiate T cell-mediated immune processes. NO synthesized by the inducible NO synthase (iNOS) is important for the function of murine DCs. In our study, we investigated the regulation of the arginine/NO-system in human monocyte-derived DCs. Maturation of DCs induced by inflammatory cytokines (IL-1beta, TNF, IL-6, and PGE(2)) resulted in a pronounced expression of neuronal NOS (nNOS) but only minimal levels of iNOS and endothelial NOS were detected in human mature DCs. In addition, reporter cell assays revealed the production of NO by mature DCs. Specific inhibitors of NOS (N-nitro-L-arginine methyl ester) or of the NO target guanylyl cyclase (H-(1,2,4)-oxadiazolo [4,3-a] quinoxalin-1-one) prevented DC maturation (shown by decreased expression of MHC class II, costimulatory and CD83 molecules and reduced IL-12 production) and preserved an immature phenotype, indicating an autocrine effect of nNOS-derived NO on human DC maturation. Notably, inhibitor-treated DCs were incapable of inducing efficient T cell responses after primary culture and generated an anergic T cell phenotype. In conclusion, our results suggest that, in the human system, nNOS-, but not iNOS-derived NO, plays an important regulatory role for the maturation of DCs and, thus, the induction of pronounced T cell responses.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/citología , Óxido Nítrico Sintasa de Tipo I/inmunología , Línea Celular , Separación Celular , Citocinas/biosíntesis , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Immunoblotting , Activación de Linfocitos/inmunología , Óxido Nítrico/biosíntesis , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Br J Pharmacol ; 179(12): 3007-3023, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34935131

RESUMEN

BACKGROUND AND PURPOSE: Preeclampsia, characterized by hypertension, proteinuria and restriction of fetal growth, is one of the leading causes of maternal and perinatal mortality. So far, there is no effective pharmacological therapy for preeclampsia. The present study was conducted to investigate the effects of supplementation with l-citrulline in Dahl salt-sensitive rats, a model of superimposed preeclampsia. EXPERIMENTAL APPROACH: Parental Dahl salt-sensitive rats were treated with l-citrulline (2.5 g·L-1 in drinking water) from the day of mating to the end of lactation period. Blood pressure was monitored throughout pregnancy and markers of preeclampsia were assessed. Endothelial function of the pregnant Dahl salt-sensitive rats was assessed by wire myograph. KEY RESULTS: In Dahl salt-sensitive rats, l-citrulline supplementation significantly reduced maternal blood pressure, proteinuria and levels of circulating soluble fms-like tyrosine kinase 1. l-Citrulline improved maternal endothelial function by augmenting the production of nitric oxide in the aorta and improving endothelium-derived hyperpolarizing factor-mediated vasorelaxation in resistance arteries. l-Citrulline supplementation improved placental insufficiency and fetal growth, which were associated with an enhancement of angiogenesis and reduction of fibrosis and senescence in the placentas. In addition, l-citrulline down-regulated genes involved in the TLR4 and NF-κB signalling pathways. CONCLUSION AND IMPLICATIONS: This study shows that l-citrulline supplementation reduced gestational hypertension and improved placentation and fetal growth in a rat model of superimposed preeclampsia. l-Citrulline supplementation may provide an effective and safe therapeutic strategy for preeclampsia that benefits both the mother and the fetus.


Asunto(s)
Preeclampsia , Animales , Factores Biológicos , Citrulina/metabolismo , Citrulina/farmacología , Citrulina/uso terapéutico , Femenino , Humanos , Masculino , Placenta/metabolismo , Preeclampsia/tratamiento farmacológico , Embarazo , Proteinuria/complicaciones , Proteinuria/tratamiento farmacológico , Proteinuria/metabolismo , Ratas , Ratas Endogámicas Dahl
13.
J Mol Cell Cardiol ; 51(5): 855-61, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21839088

RESUMEN

In certain cases of endothelial dysfunction l-arginine becomes rate-limiting for NO synthesis in spite of sufficiently high plasma concentrations of the amino acid. To better understand this phenomenon, we investigated routes of substrate supply to endothelial nitric oxide synthase (eNOS). Our previous data with human umbilical vein (HUVEC) and EA.hy.926 endothelial cells demonstrated that eNOS can obtain its substrate from the conversion of l-citrulline to l-arginine and from protein breakdown. In the present study, we determined the quantitative contribution of proteasomal and lysosomal protein degradation and investigated to what extent extracellular peptides and l-citrulline can provide substrate to eNOS. The RFL-6 reporter cell assay was used to measure eNOS activity in human EA.hy926 endothelial cells. Individual proteasome and lysosome inhibition reduced eNOS activity in EA.hy926 cells only slightly. However, the combined inhibition had a pronounced reducing effect. eNOS activity was fully restored by supplementing either l-citrulline or l-arginine-containing dipeptides. Histidine prevented the restoration of eNOS activity by the dipeptide, suggesting that a transporter accepting both, peptides and histidine, mediates the uptake of the extracellular peptide. In fact, the peptide and histidine transporter PHT1 was expressed in EA.hy926 cells and HUVECs (qRT/PCR). Our study thus demonstrates that l-citrulline and l-arginine-containing peptides derived from either intracellular protein breakdown or from the extracellular space seem to be good substrate sources for eNOS.


Asunto(s)
Arginina , Aterosclerosis/metabolismo , Citrulina , Endotelio Vascular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/biosíntesis , Arginina/metabolismo , Arginina/farmacología , Aterosclerosis/patología , Aterosclerosis/prevención & control , Transporte Biológico/efectos de los fármacos , Línea Celular , Cloroquina/farmacología , Cromatografía Líquida de Alta Presión , Citrulina/metabolismo , Citrulina/farmacología , Dipéptidos/metabolismo , Dipéptidos/farmacología , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Genes Reporteros , Histidina/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Leupeptinas/farmacología , Lisosomas/metabolismo , Proteínas de Transporte de Membrana/genética , Óxido Nítrico Sintasa de Tipo III/genética , Oligopéptidos/farmacología , Inhibidores de Proteasas/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis/efectos de los fármacos
14.
Gastroenterology ; 139(5): 1686-98, 1698.e1-6, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20600019

RESUMEN

BACKGROUND & AIMS: Helicobacter pylori-induced immune responses fail to eradicate the bacterium. Nitric oxide (NO) can kill H pylori. However, translation of inducible NO synthase (iNOS) and NO generation by H pylori-stimulated macrophages is inhibited by the polyamine spermine derived from ornithine decarboxylase (ODC), and is dependent on availability of the iNOS substrate L-arginine (L-Arg). We determined if spermine inhibits iNOS-mediated immunity by reducing L-Arg uptake into macrophages. METHODS: Levels of the inducible cationic amino acid transporter (CAT)2, ODC, and iNOS were measured in macrophages and H pylori gastritis tissues. L-Arg uptake, iNOS expression, and NO levels were assessed in cells with small interfering RNA knockdown of CAT2 or ODC, and in gastric macrophages. The ODC inhibitor, α-difluoromethylornithine, was administered to H pylori-infected mice for 4 months after inoculation. RESULTS: H pylori induced CAT2 and uptake of L-Arg in RAW 264.7 or primary macrophages. Addition of spermine or knockdown of CAT2 inhibited L-Arg uptake, NO production, and iNOS protein levels, whereas knockdown of ODC had the opposite effect. CAT2 and ODC were increased in mouse and human H pylori gastritis tissues and localized to macrophages. Gastric macrophages from H pylori-infected mice showed increased ODC expression, and attenuated iNOS and NO levels upon ex vivo H pylori stimulation versus cells from uninfected mice. α-Difluoromethylornithine treatment of infected mice restored L-Arg uptake, iNOS protein expression, and NO production in gastric macrophages, and significantly reduced both H pylori colonization levels and gastritis severity. CONCLUSIONS: Up-regulation of ODC in gastric macrophages impairs host defense against H pylori by suppressing iNOS-derived NO production.


Asunto(s)
Arginina/antagonistas & inhibidores , Mucosa Gástrica/metabolismo , Infecciones por Helicobacter/inmunología , Helicobacter pylori/patogenicidad , Inmunidad Celular/fisiología , Óxido Nítrico/biosíntesis , Espermina/farmacología , Animales , Arginina/metabolismo , Transportador de Aminoácidos Catiônicos 2/biosíntesis , Transportador de Aminoácidos Catiônicos 2/genética , Células Cultivadas , Modelos Animales de Enfermedad , Mucosa Gástrica/microbiología , Gastritis/metabolismo , Gastritis/microbiología , Gastritis/patología , Regulación de la Expresión Génica , Infecciones por Helicobacter/metabolismo , Infecciones por Helicobacter/microbiología , Helicobacter pylori/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Ornitina Descarboxilasa/biosíntesis , Ornitina Descarboxilasa/genética , Poliaminas/farmacología , ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Ophthalmologe ; 118(7): 684-690, 2021 Jul.
Artículo en Alemán | MEDLINE | ID: mdl-32940851

RESUMEN

BACKGROUND: Asymmetric dimethylarginine (ADMA) is considered an independent cardiovascular risk factor (cvRF) and thus represents a potential new biomarker for retinal vein occlusion (RVO). METHODS: Overall, 92 patients with RVO and the same number of matched controls were included in the Gutenberg RVO study. All patients underwent a standardized examination for cvRF at the study center of the population-based Gutenberg health study (GHS) as well as ophthalmological examinations and intensive laboratory tests. This article presents a substudy of patients (≤65 years old) and the controls in whom ADMA was additionally determined by high performance liquid chromatography (HPLC) at baseline and 4-6 weeks later. RESULTS: Out of 44 patients with RVO 22 had central retinal vein occlusion (CRVO), 15 had branch retinal vein occlusion (BRVO) and 7 had hemiretinal vein occlusion (hemi-RVO). The ADMA levels were 0.383 ± 0.094 µM (mean ± standard deviation) in RVO patients at baseline and 0.380 ± 0.093 µM (p = 0.514, initial vs. follow-up) after the follow-up period versus 0.360 ± 0.077 µM (p = 0.175, controls vs. RVO) in controls (n = 44). Arterial hypertension was the most prevalent risk factor in 22 (50%) of the patients and in 11 (25%) of the controls (odds ratio, OR 2.77, 95% confidence interval, CI 0.97-7.95; p = 0.058). The ADMA values above the 95th percentile (>0.530 µM) were detected in 4 patients with RVO (9.1%) but not in any of the controls (p = 0.041, RVO vs. controls). CONCLUSION: Hypertension is the most important risk factor for RVO. Due to the high number of hypertensive patients in the cohort, the relevance of ADMA as an independent risk factor could neither be confirmed nor disproved.


Asunto(s)
Hipertensión , Oclusión de la Vena Retiniana , Anciano , Arginina/análogos & derivados , Humanos , Oclusión de la Vena Retiniana/diagnóstico , Oclusión de la Vena Retiniana/epidemiología , Factores de Riesgo
17.
Biomedicines ; 9(11)2021 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-34829915

RESUMEN

B lymphocytes have been implicated in the development of insulin resistance, atherosclerosis and certain types of hypertension. In contrast to these studies, which were performed under pathological conditions, the present study provides evidence for the protective effect of B lymphocytes in maintaining vascular homeostasis under physiological conditions. In young mice not exposed to any known risk factors, the lack of B cells led to massive endothelial dysfunction. The vascular dysfunction in B cell-deficient mice was associated with an increased number of neutrophils in the circulating blood. Neutrophil depletion in B cell-deficient mice resulted in the complete normalization of vascular function, indicating a causal role of neutrophilia. Moreover, vascular function in B cell-deficient mice could be restored by adoptive transfer of naive B-1 cells isolated from wild-type mice. Interestingly, B-1 cell transfer also reduced the number of neutrophils in the recipient mice, further supporting the involvement of neutrophils in the vascular pathology caused by B cell-deficiency. In conclusion, we report in the present study the hitherto undescribed role of B lymphocytes in regulating vascular function. B cell dysregulation may represent a crucial mechanism in vascular pathology.

18.
J Pharmacol Exp Ther ; 335(1): 149-54, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20610621

RESUMEN

A crucial cause of the decreased bioactivity of nitric oxide (NO) in cardiovascular diseases is the uncoupling of the endothelial NO synthase (eNOS) caused by the oxidative stress-mediated deficiency of the NOS cofactor tetrahydrobiopterin (BH(4)). The reversal of eNOS uncoupling might represent a novel therapeutic approach. The treatment of apolipoprotein E knockout (ApoE-KO) mice with resveratrol resulted in the up-regulation of superoxide dismutase (SOD) isoforms (SOD1-SOD3), glutathione peroxidase 1 (GPx1), and catalase and the down-regulation of NADPH oxidases NOX2 and NOX4 in the hearts of ApoE-KO mice. This was associated with reductions in superoxide, 3-nitrotyrosine, and malondialdehyde levels. In parallel, the cardiac expression of GTP cyclohydrolase 1 (GCH1), the rate-limiting enzyme in BH(4) biosynthesis, was enhanced by resveratrol. This enhancement was accompanied by an elevation in BH(4) levels. Superoxide production from ApoE-KO mice hearts was reduced by the NOS inhibitor L-N(G)-nitro-arginine methyl ester, indicating eNOS uncoupling in this pathological model. Resveratrol treatment resulted in a reversal of eNOS uncoupling. Treatment of human endothelial cells with resveratrol led to an up-regulation of SOD1, SOD2, SOD3, GPx1, catalase, and GCH1. Some of these effects were preventable with sirtinol, an inhibitor of the protein deacetylase sirtuin 1. In summary, resveratrol decreased superoxide production and enhanced the inactivation of reactive oxygen species. The resulting reduction in BH(4) oxidation, together with the enhanced biosynthesis of BH(4) by GCH1, probably was responsible for the reversal of eNOS uncoupling. This novel mechanism (reversal of eNOS uncoupling) might contribute to the protective effects of resveratrol.


Asunto(s)
Antioxidantes/farmacología , Apolipoproteínas E/fisiología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estilbenos/farmacología , Animales , Apolipoproteínas E/genética , Biopterinas/análogos & derivados , Biopterinas/metabolismo , GTP Ciclohidrolasa/genética , GTP Ciclohidrolasa/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Noqueados , Miocardio/metabolismo , Estrés Oxidativo/efectos de los fármacos , ARN/biosíntesis , ARN/genética , Resveratrol , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
19.
Pflugers Arch ; 458(6): 1163-73, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19562367

RESUMEN

Since arginine metabolites, such as nitric oxide and polyamines, influence the expression of genes involved in erythroid differentiation, the transport of the cationic amino acid may play an important role in erythroid cells. However, available data only concern the presence in these cells of CAT1 transporter (system y(+)), while no information exists on the role of the heterodimeric transporters of system y(+)L (4F2hc/y(+)LAT1 and 4F2hc/y(+)LAT2) which operates transmembrane arginine fluxes cis-inhibited by neutral amino acids in the presence of sodium. Using erythroleukemia K562 cells and normal erythroid precursors, we demonstrate here that arginine transport in human erythroid cells is due to the additive contributions of a leucine-sensitive and leucine-insensitive component. In both cell types, leucine inhibition of arginine influx is much less evident in the absence of sodium, a hallmark of system y(+)L. In K562 cells, N-ethylmaleimide, a known inhibitor of CAT transporters (system y(+)), suppresses only a fraction of arginine influx corresponding to leucine-insensitive uptake. Moreover, in Xenopus oocytes coexpressing 4F2hc and y(+)LAT2, leucine exerts a marked inhibition of arginine transport, partially dependent on sodium, while no inhibition is seen in oocytes expressing CAT1. Lastly, silencing of SLC7A6, the gene for y(+)LAT2, lowers arginine transport and doubles the intracellular content of the cationic amino acid in K562 cells. We conclude that arginine transport in human erythroid cells is due to both system y(+) (CAT1 transporter) and system y(+)L (4F2hc/y(+)LAT2 isoform), which mainly contribute, respectively, to the influx and to the efflux of the cationic amino acid.


Asunto(s)
Células Precursoras Eritroides/metabolismo , Cadena Pesada de la Proteína-1 Reguladora de Fusión/fisiología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Sistema de Transporte de Aminoácidos y+/metabolismo , Sistemas de Transporte de Aminoácidos Básicos/fisiología , Animales , Arginina/metabolismo , Transportador de Aminoácidos Catiónicos 1/metabolismo , Silenciador del Gen , Humanos , Células K562 , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Oocitos/metabolismo , Xenopus laevis
20.
Biochem Biophys Res Commun ; 386(4): 650-5, 2009 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-19545540

RESUMEN

We have recently shown that inhibition of nitric oxide (NO) synthesis by asymmetrical dimethylarginine (ADMA) accelerated endothelial cell (EC) senescence which was prevented by coincubation with L-arginine; however the effect of long-term treatment of l-arginine alone on senescence of ECs have not been investigated. Human ECs were cultured in medium containing different concentrations of L-arginine until senescence. L-Arginine paradoxically accelerated senescence indicated by inhibiting telomerase activity. Moreover, L-arginine decreased NO metabolites, increased peroxynitrite, and 8-iso-prostaglandin F(2alpha) formation. In old cells, the mRNA expression of human amino acid transporter (hCAT)2B, the activity and protein expression of arginase II were upregulated indicated by enhanced urea, L-ornithine, and L-arginine consumption. Inhibition of arginase activity, or transfection with arginase II siRNA prevented L-arginine-accelerated senescence. The most possible explanation for the paradoxical acceleration of senescence by L-arginine so far may be the translational and posttranslational activation of arginase II.


Asunto(s)
Arginina/análogos & derivados , Arginina/metabolismo , Senescencia Celular/fisiología , Células Endoteliales/fisiología , Endotelio Vascular/fisiología , Arginasa/antagonistas & inhibidores , Arginasa/biosíntesis , Arginasa/genética , Arginina/farmacología , Transportador de Aminoácidos Catiónicos 1/biosíntesis , Transportador de Aminoácidos Catiônicos 2/biosíntesis , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Humanos , Óxido Nítrico/biosíntesis , Estrés Oxidativo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA