Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Immunol ; 195(11): 5237-50, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26491198

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature cells that expand during benign and cancer-associated inflammation and are characterized by their ability to inhibit T cell immunity. Increased metabolism of l-Arginine (l-Arg), through the enzymes arginase 1 and NO synthase 2 (NOS2), is well documented as a major MDSC suppressive mechanism. Therefore, we hypothesized that restricting MDSC uptake of l-Arg is a critical control point to modulate their suppressor activity. Using murine models of prostate-specific inflammation and cancer, we have identified the mechanisms by which extracellular l-Arg is transported into MDSCs. We have shown that MDSCs recruited to localized inflammation and tumor sites upregulate cationic amino acid transporter 2 (Cat2), coordinately with Arg1 and Nos2. Cat2 expression is not induced in MDSCs in peripheral organs. CAT2 contributes to the transport of l-Arg in MDSCs and is an important regulator of MDSC suppressive function. MDSCs that lack CAT2 have significantly reduced suppressive ability ex vivo and display impaired capacity for regulating T cell responses in vivo as evidenced by increased T cell expansion and decreased tumor growth in Cat2(-/-) mice. The abrogation of suppressive function is due to low intracellular l-Arg levels, which leads to the impaired ability of NOS2 to catalyze l-Arg-dependent metabolic processes. Together, these findings demonstrate that CAT2 modulates MDSC function. In the absence of CAT2, MDSCs display diminished capacity for controlling T cell immunity in prostate inflammation and cancer models, where the loss of CAT2 results in enhanced antitumor activity.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/genética , Transportador de Aminoácidos Catiônicos 2/biosíntesis , Células Mieloides/inmunología , Neoplasias de la Próstata/inmunología , Linfocitos T/inmunología , Sistemas de Transporte de Aminoácidos Básicos/biosíntesis , Animales , Arginasa/biosíntesis , Arginina/metabolismo , Transporte Biológico , Transportador de Aminoácidos Catiônicos 2/genética , Línea Celular Tumoral , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Neoplasias de la Próstata/patología , Especies Reactivas de Oxígeno/metabolismo
2.
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
3.
Anesthesiology ; 105(6): 1201-10; discussion 5A, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17122583

RESUMEN

BACKGROUND: L-Arginine transport mediated by type 2 cationic amino acid transporter (CAT-2) is one crucial mechanism that regulates nitric oxide production mediated by inducible nitric oxide synthase. Heme oxygenase (HO)-1 induction has been reported to significantly attenuate inducible nitric oxide synthase expression and nitric oxide production. The authors sought to explore the effects of HO-1 induction on CAT-2 expression and L-arginine transport. The effects of HO-1 induction on nuclear factor E2-related factor 2 (Nrf2) and nuclear factor kappaB (NF-kappaB) were also investigated. METHODS: Murine macrophages (RAW264.7 cells) were randomized to receive lipopolysaccharide, lipopolysaccharide plus hemin (an HO-1 inducer; 5, 50, or 500 microm), lipopolysaccharide plus hemin (5, 50, or 500 microm) plus tin protoporphyrin (an HO-1 inhibitor), or lipopolysaccharide plus hemin (5, 50, or 500 microm) plus hemoglobin (a carbon monoxide scavenger). Then, cell cultures were harvested and analyzed. RESULTS: Lipopolysaccharide significantly induced Nrf2 activation and HO-1 expression. Lipopolysaccharide also significantly induced NF-kappaB activation, CAT-2 expression, and L-arginine transport. In a dose-dependent manner, hemin enhanced the lipopolysaccharide-induced Nrf2 activation and HO-1 expression. In contrast, hemin, also in a dose-dependent manner, significantly attenuated the lipopolysaccharide-induced NF-kappaB activation, CAT-2 expression, and L-arginine transport. Furthermore, the effects of hemin were significantly reversed by both tin protoporphyrin and hemoglobin. CONCLUSIONS: HO-1 induction significantly inhibited CAT-2 expression and L-arginine transport in lipopolysaccharide-stimulated macrophages, possibly through mechanisms involved activation of Nrf2 and inhibition of NF-kappaB. In addition, carbon monoxide mediated, at least in part, the effects of HO-1 induction on CAT-2 expression and L-arginine transport.


Asunto(s)
Arginina/metabolismo , Transportador de Aminoácidos Catiônicos 2/antagonistas & inhibidores , Transportador de Aminoácidos Catiônicos 2/biosíntesis , Hemo-Oxigenasa 1/fisiología , Hemina/farmacología , Macrófagos/metabolismo , Factor 2 Relacionado con NF-E2/fisiología , FN-kappa B/fisiología , Animales , Transporte Biológico Activo/efectos de los fármacos , Monóxido de Carbono/metabolismo , Núcleo Celular/química , Células Cultivadas , Citosol/química , Hemoglobinas/farmacología , Immunoblotting , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Metaloporfirinas/farmacología , Ratones , Protoporfirinas/farmacología
4.
J Immunol ; 171(3): 1232-9, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12874210

RESUMEN

L-Arginine plays a central role in the normal function of several organs including the immune system. It is metabolized in macrophages by inducible nitric oxide synthase to produce nitric oxide, important in the cytotoxic mechanisms, and by arginase I (ASE I) and arginase II (ASE II) to synthesize L-ornithine and urea, the first being the precursor for the production of polyamines needed for cell proliferation. L-Arginine availability can modulate T cell function. Human T cells stimulated and cultured in the absence of L-arginine lose the expression of the TCR zeta-chain (CD3zeta) and have an impaired proliferation and a decreased cytokine production. The aim of this work was to test whether activated macrophages could modulate extracellular levels of L-arginine and alter T cell function, and to determine which metabolic pathway was responsible for this event. The results show that macrophages stimulated with IL-4 + IL-13 up-regulate ASE I and cationic amino acid transporter 2B, causing a rapid reduction of extracellular levels of L-arginine and inducing decreased expression of CD3zeta and diminished proliferation in normal T lymphocytes. Competitive inhibitors of ASE I or the addition of excess L-arginine lead to the re-expression of CD3zeta and recovery of T cell proliferation. In contrast, inducible nitric oxide synthase or ASE II failed to significantly reduce the extracellular levels of L-arginine and modulate CD3zeta expression. These results may provide new insights into the mechanisms leading to T cell dysfunction and the down-regulation of CD3zeta in cancer and chronic infectious diseases.


Asunto(s)
Arginina/metabolismo , Complejo CD3/biosíntesis , Macrófagos Peritoneales/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Arginasa/fisiología , Arginina/antagonistas & inhibidores , Arginina/fisiología , Complejo CD3/metabolismo , Transportador de Aminoácidos Catiônicos 2/biosíntesis , Transportador de Aminoácidos Catiônicos 2/genética , Transportador de Aminoácidos Catiônicos 2/fisiología , División Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Regulación hacia Abajo/inmunología , Espacio Extracelular/enzimología , Espacio Extracelular/inmunología , Espacio Extracelular/metabolismo , Femenino , Humanos , Interleucina-13/farmacología , Interleucina-4/farmacología , Células Jurkat , Activación de Macrófagos/inmunología , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/fisiología , Ratones , Ratones Endogámicos C57BL , Linfocitos T/enzimología , Regulación hacia Arriba/inmunología
5.
Am J Pathol ; 162(1): 203-11, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12507903

RESUMEN

Abnormal proliferation of keratinocytes in the skin appears crucial to the pathogenesis of psoriasis, but the underlying mechanisms remain unknown. Nitric oxide (NO), released from keratinocytes at high concentrations, is considered a key inhibitor of cellular proliferation and inducer of differentiation in vitro. Although high-output NO synthesis is suggested by the expression of inducible NO synthase (iNOS) mRNA and protein in psoriasis lesions, the pronounced hyperproliferation of psoriatic keratinocytes may indicate that iNOS activity is too low to effectively deliver anti-proliferative NO concentrations. Here we show that arginase 1 (ARG1), which substantially participates in the regulation of iNOS activity by competing for the common substrate L-arginine, is highly overexpressed in the hyperproliferative psoriatic epidermis and is co-expressed with iNOS. Expression of L-arginine transporter molecules is found to be normal. Treatment of primary cultured keratinocytes with Th1-cytokines, as present in a psoriatic environment, leads to de novo expression of iNOS but concomitantly a significant down-regulation of ARG1. Persistent ARG1 overexpression in psoriasis lesions, therefore, may represent a disease-associated deviation from normal expression patterns. Furthermore, the culturing of activated keratinocytes in the presence of an ARG inhibitor results in a twofold increase in nitrite accumulation providing evidence for an L-arginine substrate competition in human keratinocytes. High-output NO synthesis is indeed associated with a significant decrease in cellular proliferation as shown by down-regulation of Ki67 expression in cultured keratinocytes but also in short-term organ cultures of normal human skin. In summary, our data demonstrate for the first time a link between a human inflammatory skin disease, limited iNOS activity, and ARG1 overexpression. This link may have substantial implications for the pathophysiology of psoriasis and the development of new treatment strategies.


Asunto(s)
Arginasa/biosíntesis , Carcinoma Basocelular/enzimología , Queratinocitos/metabolismo , Óxido Nítrico Sintasa/metabolismo , Psoriasis/enzimología , Neoplasias Cutáneas/enzimología , Sistemas de Transporte de Aminoácidos Básicos , Arginasa/antagonistas & inhibidores , Arginasa/genética , Biopsia , Carcinoma Basocelular/patología , Transportador de Aminoácidos Catiônicos 2/biosíntesis , Transportador de Aminoácidos Catiônicos 2/genética , División Celular/efectos de los fármacos , Células Cultivadas , Citocinas/farmacología , Regulación hacia Abajo/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Técnicas In Vitro , Queratinocitos/efectos de los fármacos , Queratinocitos/patología , Antígeno Ki-67/biosíntesis , Antígeno Ki-67/genética , Óxido Nítrico/biosíntesis , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Nitritos/metabolismo , Psoriasis/patología , ARN Mensajero/biosíntesis , Valores de Referencia , Piel/citología , Piel/enzimología , Neoplasias Cutáneas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA