Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 179(12): 3007-3023, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34935131

RESUMO

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.


Assuntos
Pré-Eclâmpsia , Animais , Fatores Biológicos , Citrulina/metabolismo , Citrulina/farmacologia , Citrulina/uso terapêutico , Feminino , Humanos , Masculino , Placenta/metabolismo , Pré-Eclâmpsia/tratamento farmacológico , Gravidez , Proteinúria/complicações , Proteinúria/tratamento farmacológico , Proteinúria/metabolismo , Ratos , Ratos Endogâmicos Dahl
2.
Biomedicines ; 9(11)2021 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-34829915

RESUMO

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.

3.
Ophthalmologe ; 118(7): 684-690, 2021 Jul.
Artigo em Alemão | MEDLINE | ID: mdl-32940851

RESUMO

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.


Assuntos
Hipertensão , Oclusão da Veia Retiniana , Idoso , Arginina/análogos & derivados , Humanos , Oclusão da Veia Retiniana/diagnóstico , Oclusão da Veia Retiniana/epidemiologia , Fatores de Risco
4.
Amino Acids ; 52(3): 411-424, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32008091

RESUMO

We have previously shown that particle-mediated epidermal delivery (PMED) of plasmids encoding ß-galactosidase (ßGal) under control of the fascin-1 promoter (pFascin-ßGal) yielded selective production of the protein in skin dendritic cells (DCs), and suppressed Th2 responses in a mouse model of type I allergy by inducing Th1/Tc1 cells. However, intranasal challenge of mice immunized with pFascin-ßGal induced airway hyperreactivity (AHR) and neutrophilic inflammation in the lung. The tryptophan-catabolizing enzyme indoleamine 2,3-dioxygenase (IDO) has been implicated in immune suppression and tolerance induction. Here we investigated the consequences of co-application of an IDO-encoding vector on the modulatory effect of DNA vaccination by PMED using pFascin-ßGal in models of eosinophilic allergic and non-eosinophilic intrinsic airway inflammation. IDO-encoding plasmids and pFascin-ßGal or pCMV-ßGal were co-applied to abdominal skin of BALB/c mice without, before or after sensitization with ßGal protein. Immune responses in the lung were analysed after intranasal provocation and airway reactivity was determined by whole body plethysmography. Co-application of pCMV-IDO with pFascin-ßGal, but not pCMV-ßGal inhibited the Th1/Tc1 immune response after PMED. Moreover, AHR in those mice was attenuated following intranasal challenge. Therapeutic vaccination of ßGal-sensitized mice with pFascin-ßGal plus pCMV-IDO slightly suppressed airway inflammation and AHR after provocation with ßGal protein, while prophylactic vaccination was not effective. Altogether, our data suggest that only the combination of DC-restricted antigen and ubiquitous IDO expression attenuated asthma responses in mice, most probably by forming a tryptophan-depleted and kynurenine-enriched micromilieu known to affect neutrophils and T cells.


Assuntos
Asma/terapia , Biolística/métodos , Hiper-Reatividade Brônquica/terapia , Células Dendríticas/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Vacinação/métodos , Animais , Antígenos/genética , Antígenos/imunologia , Asma/imunologia , Hiper-Reatividade Brônquica/imunologia , Proteínas de Transporte/genética , Vetores Genéticos , Camundongos , Camundongos Endogâmicos BALB C , Proteínas dos Microfilamentos/genética , Células NIH 3T3 , Infiltração de Neutrófilos , Linfócitos T Citotóxicos/imunologia , Células Th1/imunologia , Transgenes , beta-Galactosidase/genética , beta-Galactosidase/imunologia
5.
Amino Acids ; 52(3): 499-503, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32008093

RESUMO

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.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Transportador 1 de Aminoácidos Catiônicos/metabolismo , Cisteína/metabolismo , Óxido Nítrico/metabolismo , Animais , Arginina/metabolismo , Humanos , Oócitos , Xenopus laevis
6.
Front Immunol ; 11: 617699, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33717053

RESUMO

Myeloid cell arginase-mediated arginine depletion with consecutive inhibition of T cell functions is a key component of tumor immune escape. Both, granulocytic myeloid-derived suppressor cells (G-MDSC) and conventional mature human polymorphonuclear neutrophil granulocytes (PMN) express high levels of arginase 1 and can act as suppressor cells of adaptive anti-cancer immunity. Here we demonstrate that pharmacological inhibition of PMN-derived arginase 1 not only prevents the suppression of T cell functions but rather leads to a strong hyperactivation of T cells. Human PMN were incubated in cell culture medium in the absence or presence of an arginase inhibitor. T cells from healthy donors were then activated either polyclonally or in an antigen-specific manner in the supernatants of the PMN cultures at different PMN-T cell ratios. T cell proliferation was completely suppressed in these supernatants in the absence of an arginase inhibitor. Arginase inhibition led to a strong hyperinduction of T cell proliferation, which exceeded control activation conditions up to 25-fold. The hyperinduction was correlated with higher PMN-T cell ratios and was only apparent when PMN arginase activity was blocked sufficiently. The T cell stimulatory factor was liberated very early by PMN and was present in the < 3 kDa fraction of the PMN supernatants. Increased T cell production of specific proinflammatory cytokines by PMN supernatant in the presence of arginase inhibitor was apparent. Upon arginase inhibition, downregulation of important T cell membrane activation and costimulation proteins was completely prevented or de novo induction accelerated. Antigen-specific T cell cytotoxicity against tumor cells was enhanced by PMN supernatant itself and could be further increased by PMN arginase blockade. Finally, we analyzed anergic T cells from multiple myeloma patients and noticed a complete reversal of anergy and the induction of strong proliferation upon T cell activation in PMN supernatants by arginase inhibition. In summary, we discovered a potent PMN-mediated hyperactivation of human T cells, which is apparent only when PMN arginase-mediated arginine depletion is concurrently inhibited. Our findings are clearly relevant for the analysis and prevention of human tumor immune escape in conjunction with the application of arginase inhibitors already being developed clinically.


Assuntos
Arginase/metabolismo , Ativação Linfocitária/imunologia , Neutrófilos/enzimologia , Linfócitos T/imunologia , Proliferação de Células/fisiologia , Células Cultivadas , Humanos , Mieloma Múltiplo/imunologia , Neutrófilos/imunologia , Evasão Tumoral/imunologia
7.
Front Oncol ; 9: 1268, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31824848

RESUMO

Interfering with tumor metabolism by specifically restricting the availability of extracellular nutrients is a rapidly emerging field of cancer research. A variety of tumor entities depend on the uptake of the amino acid arginine since they have lost the ability to synthesize it endogenously, that is they do not express the rate limiting enzyme for arginine synthesis, argininosuccinate synthase (ASS). Arginine transport through the plasma membrane of mammalian cells is mediated by eight different transporters that belong to two solute carrier (SLC) families. In the present study we found that the proliferation of primary as well as immortalized chronic lymphocytic leukemia (CLL) cells depends on the availability of extracellular arginine and that primary CLL cells do not express ASS and are therefore arginine-auxotrophic. The cationic amino acid transporter-1 (CAT-1) was the only arginine importer expressed in CLL cells. Lentiviral-mediated downregulation of the CAT-1 transporter in HG3 CLL cells significantly reduced arginine uptake, abolished cell proliferation and impaired cell viability. In a murine CLL xenograft model, tumor growth was significantly suppressed upon induced downregulation of CAT-1 in the CLL cells. Our results suggest that inhibition of CAT-1 is a promising new therapeutic approach for CLL.

9.
Front Immunol ; 8: 864, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28791021

RESUMO

In the tumor microenvironment, arginine is metabolized by arginase-expressing myeloid cells. This arginine depletion profoundly inhibits T cell functions and is crucially involved in tumor-induced immunosuppression. Reconstitution of adaptive immune functions in the context of arginase-mediated tumor immune escape is a promising therapeutic strategy to boost the immunological antitumor response. Arginine can be recycled in certain mammalian tissues from citrulline via argininosuccinate (ASA) in a two-step enzymatic process involving the enzymes argininosuccinate synthase (ASS) and argininosuccinate lyase (ASL). Here, we demonstrate that anti-CD3/anti-CD28-activated human primary CD4+ and CD8+ T cells upregulate ASS expression in response to low extracellular arginine concentrations, while ASL is expressed constitutively. ASS expression peaked under moderate arginine restriction (20 µM), but no relevant induction was detectable in the complete absence of extracellular arginine. The upregulated ASS correlated with a reconstitution of T cell proliferation upon supplementation of citrulline, while the suppressed production of IFN-γ was refractory to citrulline substitution. In contrast, ASA reconstituted proliferation and cytokine synthesis even in the complete absence of arginine. By direct quantification of intracellular metabolites we show that activated primary human T cells import citrulline but only metabolize it further to ASA and arginine when ASS is expressed in the context of low amounts of extracellular arginine. We then clarify that citrulline transport is largely mediated by the L-type amino acid transporter 1 (LAT1), induced upon human T cell activation. Upon siRNA-mediated knockdown of LAT1, activated T cells lost the ability to import citrulline. These data underline the potential of citrulline substitution as a promising pharmacological way to treat immunosuppression in settings of arginine deprivation.

10.
Oncotarget ; 8(70): 115667-115680, 2017 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-29383190

RESUMO

BACKGROUND: Organic cation transporters (OCT) are responsible for the uptake of a broad spectrum of endogenous and exogenous substrates. Downregulation of OCT is frequently observed in human hepatocellular carcinoma (HCC) and is associated with a poor outcome. The aim of our current study was to elucidate the impact of OCT3 on hepatocarcinogenesis. METHODS: Transcriptional and functional loss of OCT was investigated in primary murine hepatocytes, derived from Oct3-knockout (Oct3-/-; FVB.Slc22a3tm1Dpb ) and wildtype (WT) mice. Liver tumors were induced in Oct3-/- and WT mice with Diethylnitrosamine and Phenobarbital over 10 months and characterized macroscopically and microscopically. Key survival pathways were investigated by Western Blot analysis. RESULTS: Loss of Oct3-/- in primary hepatocytes resulted in significantly reduced OCT activity determined by [3H]MPP+ uptake in vivo. Furthermore, tumor size and quantity were markedly enhanced in Oct3-/- mice (p<0.0001). Oct3-/- tumors showed significant higher proliferation (p<0.0001). Ki-67 and Cyclin D expression were significantly increased in primary Oct3-/- hepatocytes after treatment with the OCT inhibitors quinine or verapamil (p<0.05). Functional inhibition of OCT by quinine resulted in an activation of c-Jun N-terminal kinase (Jnk), especially in Oct3-/- hepatocytes. CONCLUSION: Loss of Oct3 leads to enhanced proliferation and hepatocarcinogenesis in vivo.

11.
Arterioscler Thromb Vasc Biol ; 36(1): 78-85, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26586660

RESUMO

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.


Assuntos
Tecido Adiposo/enzimologia , Aorta Torácica/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico/metabolismo , Obesidade/enzimologia , Vasodilatação , Adipócitos/enzimologia , Adipocinas/metabolismo , Tecido Adiposo/fisiopatologia , Adiposidade , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/fisiopatologia , Arginase/antagonistas & inibidores , Arginase/metabolismo , Arginina/deficiência , Citocinas/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Mediadores da Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Obesidade/fisiopatologia , Comunicação Parácrina , Fosforilação , Transdução de Sinais , Superóxidos/metabolismo , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
12.
Amino Acids ; 48(2): 427-36, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26403849

RESUMO

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.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Arginina/análogos & derivados , Proteínas de Transporte/metabolismo , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Arginina/metabolismo , Transporte Biológico , Linhagem Celular Tumoral , Humanos , Túbulos Renais Proximais/metabolismo , Transaminases
13.
Eur J Immunol ; 46(1): 92-103, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26449889

RESUMO

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.


Assuntos
Arginina/metabolismo , Transportador 1 de Aminoácidos Catiônicos/imunologia , Ativação Linfocitária/imunologia , Linfócitos T/imunologia , Western Blotting , Transportador 1 de Aminoácidos Catiônicos/metabolismo , Proliferação de Células , Células Cultivadas , Humanos , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T/metabolismo , Transfecção
14.
J Geriatr Cardiol ; 12(5): 528-39, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26512245

RESUMO

OBJECTIVE: To find out whether dexamethasone induces an uncoupling of the endothelial nitric oxide synthase (eNOS). METHODS & RESULTS: A major cause of eNOS uncoupling is a deficiency of its cofactor tetrahydrobiopterin (BH4). Treatment of human EA.hy 926 endothelial cells with dexamethasone decreased mRNA and protein expression of both BH4-synthesizing enzymes: GTP cyclohydrolase I and dihydrofolate reductase. Consistently, a concentration- and time-dependent reduction of BH4, dihydrobiopterin (BH2) as well as BH4: BH2 ratio was observed in dexamethasone-treated cells. Surprisingly, no evidence for eNOS uncoupling was found. We then analyzed the expression and phosphorylation of the eNOS enzyme. Dexamethasone treatment led to a down-regulation of eNOS protein and a reduction of eNOS phosphorylation at serine 1177. A reduction of eNOS expression may lead to a relatively normal BH4: eNOS molar ratio in dexamethasone-treated cells. Because the BH4-eNOS stoichiometry rather than the absolute BH4 amount is the key determinant of eNOS functionality (i.e., coupled or uncoupled), the down-regulation of eNOS may represent an explanation for the absence of eNOS uncoupling. Phosphorylation of eNOS at serine 1177 is needed for both the NO-producing activity of the coupled eNOS and the superoxide-producing activity of the uncoupled eNOS. Thus, a reduction of serine 1177 phosphorylation may render a potentially uncoupled eNOS hardly detectable. CONCLUSIONS: Although dexamethasone reduces BH4 levels in endothelial cells, eNOS uncoupling is not evident. The reduction of NO production in dexamethasone-treated endothelial cells is mainly attributable to reduced eNOS expression and decreased eNOS phosphorylation at serine 1177.

15.
Amino Acids ; 47(12): 2647-58, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26215737

RESUMO

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.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos/genética , Transtorno do Espectro Autista/genética , Sequência de Aminoácidos , Animais , Biotinilação , Encéfalo/metabolismo , Membrana Celular/metabolismo , Criança , Cromossomos Humanos X/genética , Epilepsia/complicações , Epilepsia/genética , Frequência do Gene , Humanos , Perda de Heterozigosidade , Masculino , Conformação Molecular , Dados de Sequência Molecular , Mutação , Mutação de Sentido Incorreto , Oócitos/metabolismo , Linhagem , Fenótipo , Xenopus laevis
17.
Amino Acids ; 47(2): 335-44, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25385314

RESUMO

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.


Assuntos
Glioblastoma/metabolismo , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Proteínas de Neoplasias/metabolismo , Tirosina/análogos & derivados , Animais , Linhagem Celular Tumoral , Meios de Contraste , Cadeia Pesada da Proteína-1 Reguladora de Fusão/genética , Cadeia Pesada da Proteína-1 Reguladora de Fusão/metabolismo , Glioblastoma/diagnóstico por imagem , Glioblastoma/genética , Humanos , Transportador 1 de Aminoácidos Neutros Grandes/genética , Proteínas de Neoplasias/genética , Tomografia por Emissão de Pósitrons , Radiografia , Tirosina/farmacocinética , Tirosina/farmacologia , Xenopus laevis
18.
Front Immunol ; 5: 532, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25386178

RESUMO

Macrophages play a major role in the immune system, both as antimicrobial effector cells and as immunoregulatory cells, which induce, suppress or modulate adaptive immune responses. These key aspects of macrophage biology are fundamentally driven by the phenotype of macrophage arginine metabolism that is prevalent in an evolving or ongoing immune response. M1 macrophages express the enzyme nitric oxide synthase, which metabolizes arginine to nitric oxide (NO) and citrulline. NO can be metabolized to further downstream reactive nitrogen species, while citrulline might be reused for efficient NO synthesis via the citrulline-NO cycle. M2 macrophages are characterized by expression of the enzyme arginase, which hydrolyzes arginine to ornithine and urea. The arginase pathway limits arginine availability for NO synthesis and ornithine itself can further feed into the important downstream pathways of polyamine and proline syntheses, which are important for cellular proliferation and tissue repair. M1 versus M2 polarization leads to opposing outcomes of inflammatory reactions, but depending on the context, M1 and M2 macrophages can be both pro- and anti-inflammatory. Notably, M1/M2 macrophage polarization can be driven by microbial infection or innate danger signals without any influence of adaptive immune cells, secondarily driving the T helper (Th)1/Th2 polarization of the evolving adaptive immune response. Since both arginine metabolic pathways cross-inhibit each other on the level of the respective arginine break-down products and Th1 and Th2 lymphocytes can drive or amplify macrophage M1/M2 dichotomy via cytokine activation, this forms the basis of a self-sustaining M1/M2 polarization of the whole immune response. Understanding the arginine metabolism of M1/M2 macrophage phenotypes is therefore central to find new possibilities to manipulate immune responses in infection, autoimmune diseases, chronic inflammatory conditions, and cancer.

19.
J Leukoc Biol ; 96(6): 1047-53, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25104794

RESUMO

Arginine depletion via myeloid cell arginase is critically involved in suppression of the adaptive immune system during cancer or chronic inflammation. On the other hand, arginine depletion is being developed as a novel anti-tumor metabolic strategy to deprive arginine-auxotrophic cancer cells of this amino acid. In human immune cells, arginase is mainly expressed constitutively in PMNs. We therefore purified human primary PMNs from healthy donors and analyzed PMN function as the main innate effector cell and arginase producer in the context of arginine deficiency. We demonstrate that human PMN viability, activation-induced IL-8 synthesis, chemotaxis, phagocytosis, generation of ROS, and fungicidal activity are not impaired by the absence of arginine in vitro. Also, profound pharmacological arginine depletion in vivo via ADI-PEG20 did not inhibit PMN functions in a mouse model of pulmonary invasive aspergillosis; PMN invasion into the lung, activation, and successful PMN-dependent clearance of Aspergillus fumigatus and survival of mice were not impaired. These novel findings add to a better understanding of immunity during inflammation-associated arginine depletion and are also important for the development of therapeutic arginine depletion as anti-metabolic tumor therapy.


Assuntos
Arginina/fisiologia , Neutrófilos/imunologia , Animais , Antineoplásicos/farmacologia , Apoptose , Arginase/sangue , Arginase/fisiologia , Arginina/análise , Arginina/farmacologia , Aspergillus fumigatus/imunologia , Líquido da Lavagem Broncoalveolar/química , Células Cultivadas , Quimiotaxia de Leucócito , Citrulina/análise , Humanos , Hidrolases/farmacologia , Imunidade Inata , Interleucina-8/biossíntese , Interleucina-8/genética , Pulmão/imunologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Fagocitose , Polietilenoglicóis/farmacologia , Cultura Primária de Células , Aspergilose Pulmonar/imunologia , Aspergilose Pulmonar/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Explosão Respiratória
20.
PLoS One ; 8(11): e82594, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24312428

RESUMO

Tetrahydrobiopterin (BH4), which fosters the formation of and stabilizes endothelial NO synthase (eNOS) as an active dimer, tightly regulates eNOS coupling / uncoupling. Moreover, studies conducted in genetically-modified models demonstrate that BH4 pulmonary deficiency is a key determinant in the pathogenesis of pulmonary hypertension. The present study thus investigates biopterin metabolism and eNOS expression, as well as the effect of sepiapterin (a precursor of BH4) and eNOS gene deletion, in a mice model of hypoxic pulmonary hypertension. In lungs, chronic hypoxia increased BH4 levels and eNOS expression, without modifying dihydrobiopterin (BH2, the oxidation product of BH4) levels, GTP cyclohydrolase-1 or dihydrofolate reductase expression (two key enzymes regulating BH4 availability). In intrapulmonary arteries, chronic hypoxia also increased expression of eNOS, but did not induce destabilisation of eNOS dimers into monomers. In hypoxic mice, sepiapterin prevented increase in right ventricular systolic pressure and right ventricular hypertrophy, whereas it modified neither remodelling nor alteration in vasomotor responses (hyper-responsiveness to phenylephrine, decrease in endothelium-dependent relaxation to acetylcholine) in intrapulmonary arteries. Finally, deletion of eNOS gene partially prevented hypoxia-induced increase in right ventricular systolic pressure, right ventricular hypertrophy and remodelling of intrapulmonary arteries. Collectively, these data demonstrate the absence of BH4/BH2 changes and eNOS dimer destabilisation, which may induce eNOS uncoupling during hypoxia-induced pulmonary hypertension. Thus, even though eNOS gene deletion and sepiapterin treatment exert protective effects on hypoxia-induced pulmonary vascular remodelling, increase on right ventricular pressure and / or right ventricular hypertrophy, these effects appear unrelated to biopterin-dependent eNOS uncoupling within pulmonary vasculature of hypoxic wild-type mice.


Assuntos
Biopterinas/metabolismo , Hipertensão Pulmonar/metabolismo , Hipóxia/complicações , Óxido Nítrico Sintase Tipo III/metabolismo , Animais , Modelos Animais de Doenças , Hipertensão Pulmonar/complicações , Hipertensão Pulmonar/enzimologia , Camundongos , Óxido Nítrico Sintase Tipo III/genética , Tetra-Hidrofolato Desidrogenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...