Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Ann Rheum Dis ; 78(3): 421-428, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30610061

RESUMEN

OBJECTIVE: Osteoarthritis (OA) appears to be associated with various metabolic disorders, but the potential contribution of amino acid metabolism to OA pathogenesis has not been clearly elucidated. Here, we explored whether alterations in the amino acid metabolism of chondrocytes could regulate OA pathogenesis. METHODS: Expression profiles of amino acid metabolism-regulating genes in primary-culture passage 0 mouse chondrocytes were examined by microarray analysis, and selected genes were further characterised in mouse OA chondrocytes and OA cartilage of human and mouse models. Experimental OA in mice was induced by destabilisation of the medial meniscus (DMM) or intra-articular (IA) injection of adenoviruses expressing catabolic regulators. The functional consequences of arginase II (Arg-II) were examined in Arg2-/- mice and those subjected to IA injection of an adenovirus encoding Arg-II (Ad-Arg-II). RESULTS: The gene encoding Arg-II, an arginine-metabolising enzyme, was specifically upregulated in chondrocytes under various pathological conditions and in OA cartilage from human patients with OA and various mouse models. Adenovirus-mediated overexpression of Arg-II in mouse joint tissues caused OA pathogenesis, whereas genetic ablation of Arg2 in mice (Arg2-/-) abolished all manifestations of DMM-induced OA. Mechanistically, Arg-II appears to cause OA cartilage destruction at least partly by upregulating the expression of matrix-degrading enzymes (matrix metalloproteinase 3 [MMP3] and MMP13) in chondrocytes via the nuclear factor (NF)-κB pathway. CONCLUSIONS: Our results indicate that Arg-II is a crucial regulator of OA pathogenesis in mice. Although chondrocytes of human and mouse do not identically, but similarly, respond to Arg-II, our results suggest that Arg-II could be a therapeutic target of OA pathogenesis.


Asunto(s)
Arginasa/fisiología , Artritis Experimental/enzimología , Cartílago Articular/enzimología , Condrocitos/enzimología , Osteoartritis/enzimología , Animales , Artritis Experimental/inducido químicamente , Modelos Animales de Enfermedad , Humanos , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Ratones , Osteoartritis/inducido químicamente , Regulación hacia Arriba
2.
Biochem Biophys Res Commun ; 451(2): 179-83, 2014 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-25078625

RESUMEN

Arginase may play a major role in the regulation of vascular function in various cardiovascular disorders by impairing nitric oxide (NO) production. In the current study, we investigated whether supplementation of the arginase inhibitor N(ω)-hydroxy-nor-l-arginine (nor-NOHA) could restore endothelial function in an animal model of diet-induced obesity. Arginase 1 expression was significantly lower in the aorta of C57BL/6J mice fed a high-fat diet (HFD) supplemented with nor-NOHA (40mgkg(-1)/day) than in mice fed HFD without nor-NOHA. Arginase inhibition led to considerable increases in eNOS expression and NO levels and significant decreases in the levels of circulating ICAM-1. These findings were further confirmed by the results of siRNA-mediated knockdown of Arg in human umbilical vein endothelial cells. In conclusion, arginase inhibition can help restore dysregulated endothelial function by increasing the eNOS-dependent NO production in the endothelium, indicating that arginase could be a therapeutic target for correcting obesity-induced vascular endothelial dysfunction.


Asunto(s)
Arginasa/antagonistas & inhibidores , Endotelio Vascular/enzimología , Endotelio Vascular/fisiopatología , Obesidad/enzimología , Obesidad/fisiopatología , Animales , Arginasa/genética , Arginasa/fisiología , Arginina/análogos & derivados , Arginina/farmacología , Dieta Alta en Grasa/efectos adversos , Endotelio Vascular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Expresión Génica , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Molécula 1 de Adhesión Intercelular/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/fisiología , Nitritos/metabolismo , Obesidad/etiología , ARN Interferente Pequeño/genética
3.
Neonatology ; 101(2): 106-15, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21952491

RESUMEN

BACKGROUND: Prolonged exposure of immature lungs to hyperoxia contributes to neonatal lung injury and airway hyperreactivity. We have previously demonstrated that neonatal exposure of rat pups to ≥95% O2 impairs airway relaxation due to disruption of nitric oxide (NO)-cyclic guanosine monophosphate (cGMP) signaling. OBJECTIVE: We now hypothesize that these impaired relaxation responses are secondary to hyperoxia-induced upregulation of arginase, which competes with NO synthase for L-arginine. METHODS: Rat pups were exposed to moderate neonatal hyperoxia (50% O2) or room air for 7 days from birth. In additional hyperoxic and room air groups, exogenous L-arginine (300 mg/kg/day i.p.) or arginase inhibitor (Nω-hydroxy-nor-arginine, 30 mg/kg/day i.p.) were administered daily. After 7 days, animals were anesthetized and sacrificed either for preparation of lung parenchymal strips or lung perfusion. RESULTS: In response to electrical field stimulation (EFS), bethanechol-preconstricted lung parenchymal strips from hyperoxic pups exhibited significantly reduced relaxation compared to room air controls. Supplementation of L-arginine or arginase blockade restored hyperoxia-induced impairment of relaxation. Expression of arginase I in airway epithelium was increased in response to hyperoxia but reduced by arginase blockade. Arginase activity was also significantly increased in hyperoxic lungs as compared to room air controls and reduced following arginase blockade. EFS-induced production of NO was decreased in hyperoxia-exposed airway smooth muscle and restored by arginase blockade. CONCLUSION: These data suggest that NO-cGMP signaling is disrupted in neonatal rat pups exposed to even moderate hyperoxia due to increased arginase activity and consequent decreased bioavailability of the substrate L-arginine. We speculate that supplementation of arginine and/or inhibition of arginase may be a useful therapeutic tool to prevent or treat neonatal lung injury.


Asunto(s)
Arginasa/fisiología , Hiperoxia/fisiopatología , Pulmón/enzimología , Pulmón/fisiología , Relajación Muscular/fisiología , Animales , Animales Recién Nacidos , Arginasa/antagonistas & inhibidores , Arginasa/biosíntesis , Arginina/análogos & derivados , Arginina/farmacología , Betanecol/farmacología , Estimulación Eléctrica , Hiperoxia/metabolismo , Pulmón/citología , Músculo Liso/efectos de los fármacos , Músculo Liso/fisiopatología , Óxido Nítrico/biosíntesis , Parasimpaticomiméticos/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba
4.
Clin Cancer Res ; 17(22): 6992-7002, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21948231

RESUMEN

PURPOSE: The source of glioblastoma (GBM)-associated immunosuppression remains multifactorial. We sought to clarify and therapeutically target myeloid cell-derived peripheral immunosuppression in patients with GBM. EXPERIMENTAL DESIGN: Direct ex vivo T-cell function, serum Arginase I (ArgI) levels, and circulating myeloid lineage populations were compared between patients with GBM and normal donors or patients with other intracranial tumors. Immunofunctional assays were conducted using bulk and sorted cell populations to explore the potential transfer of myeloid cell-mediated immunosuppression and to identify a potential mechanism for these effects. ArgI-mediated immunosuppression was therapeutically targeted in vitro through pharmacologic inhibition or arginine supplementation. RESULTS: We identified a significantly expanded population of circulating, degranulated neutrophils associated with elevated levels of serum ArgI and decreased T-cell CD3ζ expression within peripheral blood from patients with GBM. Sorted CD11b(+) cells from patients with GBM were found to markedly suppress normal donor T-cell function in coculture, and media harvested from mitogen-stimulated GBM peripheral blood mononuclear cell (PBMC) or GBM-associated mixed lymphoid reactions showed ArgI levels that were significantly higher than controls. Critically, T-cell suppression in both settings could be completely reversed through pharmacologic ArgI inhibition or with arginine supplementation. CONCLUSIONS: These data indicate that peripheral cellular immunosuppression in patients with GBM is associated with neutrophil degranulation and elevated levels of circulating ArgI, and that T-cell function can be restored in these individuals by targeting ArgI. These data identify a novel pathway of GBM-mediated suppression of cellular immunity and offer a potential therapeutic window for improving antitumor immunity in affected patients.


Asunto(s)
Arginasa/fisiología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/inmunología , Degranulación de la Célula , Glioblastoma/tratamiento farmacológico , Glioblastoma/inmunología , Tolerancia Inmunológica , Terapia Molecular Dirigida/métodos , Neutrófilos/inmunología , Arginasa/antagonistas & inhibidores , Arginina/uso terapéutico , Neoplasias Encefálicas/enzimología , Células Cultivadas , Glioblastoma/enzimología , Humanos , Inmunidad Celular , Linfocitos T/inmunología
5.
Pulm Pharmacol Ther ; 24(4): 407-13, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21281730

RESUMEN

Pulmonary embolism (PE) causes pulmonary hypertension by mechanical obstruction and constriction of non-obstructed vasculature. We tested if experimental PE impairs pulmonary vascular endothelium-dependent dilation via activation of arginase II. Experimental PE was induced in male Sprague-Dawley rats by infusing 25 µm microspheres in the right jugular vein, producing moderate pulmonary hypertension. Shams received vehicle injection. Pulmonary arterial rings were isolated after 18 h and isometric tensions were determined. Dilations were induced with acetylcholine, calcium ionophore A23187 or nitroglycerin (NTG) in pre-contracted rings (phenylephrine). Protein expression was assessed by Western blot and immunohistochemistry. Arginase activity was inhibited by intravenous infusion of N(w)-hydroxy-nor-l-arginine (nor-NOHA). l-Arginine supplementation was also given. Endothelium-dependent dilation responses were significantly reduced in PE vs. vehicle-treated animals (ACh: 50 ± 9% vs. 93 ± 3%; A23187: 19 ± 7% vs. 85 ± 7%, p < 0.05), while endothelium-independent dilations (NTG) were unchanged. Endothelial nitric oxide synthase (eNOS) protein content was unchanged by PE. Expression of arginase II increased 4.5-fold and immunohistochemistry revealed increased arginase II staining. Nor-NOHA treatment and l-arginine supplementation significantly improved pulmonary artery ring endothelium-dependent dilation in PE (ACh: 58 ± 6% PE, 88 ± 6% PE + nor-NOHA, 84 ± 4% PE + l-arginine). Experimental PE impairs endothelium-dependent pulmonary artery dilation, while endothelium-independent dilation remains unchanged. The data support the conclusion that up-regulation of arginase II protein expression contributes to pulmonary artery endothelial dysfunction in this model of experimental PE.


Asunto(s)
Arginasa/fisiología , Células Endoteliales/fisiología , Arteria Pulmonar/fisiopatología , Embolia Pulmonar/fisiopatología , Animales , Arginina/farmacología , Masculino , Óxido Nítrico Sintasa de Tipo III/análisis , Embolia Pulmonar/enzimología , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba , Vasodilatación
6.
Cardiovasc Res ; 85(1): 194-203, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19684035

RESUMEN

AIMS: Preeclampsia is a hypertensive disorder characterized by vascular oxidative stress. Decreased availability of the vasodilator nitric oxide (NO) has been postulated to be involved in the pathophysiology of this disorder. Arginase, an enzyme that competes with nitric oxide synthase (NOS) for l-arginine, not only reduces NO formation but also increases superoxide production by NOS. In placenta of preeclamptic women, arginase upregulation has been shown to be increased and contributes to superoxide formation via uncoupling of NOS. However, the role of arginase in the maternal vasculature is not clear. We hypothesized that arginase would be upregulated in the maternal vasculature of women with preeclampsia and contribute to oxidative stress within the endothelium. METHODS AND RESULTS: We observed increased arginase expression in the maternal vasculature of women with preeclampsia compared with normotensive pregnant women. Furthermore, human umbilical vein endothelial cells treated with 2% plasma from preeclamptic women show increased arginase II expression and activity that was reduced by a peroxynitrite scavenger. Also, both 3-morpholino sydnonimine and exogenous peroxynitrite increased arginase expression and activity. Preeclamptic plasma treatment increased superoxide and peroxynitrite levels. Superoxide levels were significantly reduced after arginase and NOS inhibition with [(S)-(2-boronoethyl)-l-cysteine] and N(omega)-nitro-l-arginine methyl ester, respectively, but peroxynitrite levels were in fact increased after arginase inhibition. Moreover, in the presence of preeclamptic plasma, l-arginine supplementation increased peroxynitrite formation during arginase inhibition. CONCLUSION: Increased arginase expression in preeclampsia can induce uncoupling of NOS as a source of superoxide in the maternal vasculature in preeclampsia. However, l-arginine supplementation in the face of oxidative stress could lead to a further increase in peroxynitrite.


Asunto(s)
Arginasa/fisiología , Células Endoteliales/metabolismo , Estrés Oxidativo , Preeclampsia/metabolismo , Arginina/farmacología , Biopterinas/análogos & derivados , Biopterinas/farmacología , Femenino , Humanos , Molsidomina/análogos & derivados , Molsidomina/farmacología , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/fisiología , FN-kappa B/fisiología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/metabolismo , Ácido Peroxinitroso/farmacología , Embarazo
7.
Bratisl Lek Listy ; 110(10): 592-7, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20017447

RESUMEN

BACKGROUND: The interest of arginase action is increasing because limitation of L-arginine bioavailability by arginase for NO synthesis via constitutive NOS can contribute to airway hyperreactivity. OBJECTIVES: We investigated the effect of intervention in the arginase activity in guinea pig model of experimental ovalbumin-induced airway hyperreactivity. METHODS: We analysed the response of tracheal and lung tissue smooth muscle strips to histamine or acetylcholine after in vitro administration of arginase in a dose of 75 UI or after administration of the non-selective inhibitor of arginase N(omega)-hydroxy-L-arginine (NOHA) in a dose of 5 and 10 micromol. We used as well as the incubation of strips with the precursor of NO synthesis L-arginine in a dose of 10(-4) mol/l together with NOHA. RESULTS: We did not find any significant differences in the reactivity of tracheal and lung tissue smooth muscle if we applied arginase in a dose of 75 UI in vitro. NOHA in a dose of 5 a 10 micromol induced the decrease of tracheal and lung tissue smooth muscle reactivity overall. The decrease of the contraction amplitude was dose-dependent. The supplementation of NO synthesis precursor L-arginine in a dose of 10(-4) mol/l together with NOHA intensified the decrease of the airways reactivity induced by an arginase inhibition. CONCLUSION: The results suggest that arginase is involved in the control of airways bronchomotoric tone and therefore modulation of arginase activity could be a useful tool for airway smooth muscle tone control in clinical conditions (Fig. 7, Ref. 33). Full Text (Free, PDF) www.bmj.sk.


Asunto(s)
Arginasa/fisiología , Hiperreactividad Bronquial/fisiopatología , Acetilcolina/farmacología , Animales , Arginasa/antagonistas & inhibidores , Arginasa/farmacología , Cobayas , Técnicas In Vitro , Pulmón/efectos de los fármacos , Masculino , Contracción Muscular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , NG-Nitroarginina Metil Éster/farmacología , Tráquea/efectos de los fármacos
8.
J Immunol ; 183(12): 8068-76, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19923451

RESUMEN

The balance between the products of L-arginine metabolism in macrophages regulates the outcome of Leishmania major infection. L-arginine can be oxidized by host inducible NO synthase to produce NO, which contributes to parasite killing. In contrast, L-arginine hydrolysis by host arginase blocks NO generation and provides polyamines, which can support parasite proliferation. Additionally, Leishmania encode their own arginase which has considerable potential to modulate infectivity and disease pathogenesis. In this study, we compared the infectivity and impact on host cellular immune response in vitro and in vivo of wild-type (WT) L. major with that of a parasite arginase null mutant (arg(-)) L. major. We found that arg(-) L. major are impaired in their macrophage infectivity in vitro independent of host inducible NO synthase activities. As with in vitro results, the proliferation of arg(-) L. major in animal infections was also significantly impaired in vivo, resulting in delayed onset of lesion development, attenuated pathology, and low parasite burden. Despite this attenuated pathology, the production of cytokines by cells from the draining lymph node of mice infected with WT and arg(-) L. major was similar at all times tested. Interestingly, in vitro and in vivo arginase levels were significantly lower in arg(-) than in WT-infected cases and were directly correlated with parasite numbers inside infected cells. These results suggest that Leishmania-encoded arginase enhances disease pathogenesis by augmenting host cellular arginase activities and that contrary to previous in vitro studies, the host cytokine response does not influence host arginase activity.


Asunto(s)
Arginasa/metabolismo , Citocinas/fisiología , Hiperargininemia/inmunología , Hiperargininemia/parasitología , Leishmania major/enzimología , Leishmania major/crecimiento & desarrollo , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/parasitología , Adyuvantes Inmunológicos/fisiología , Animales , Arginasa/genética , Arginasa/fisiología , Células de la Médula Ósea/enzimología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/patología , Proliferación Celular , Células Cultivadas , Activación Enzimática/genética , Activación Enzimática/inmunología , Femenino , Hiperargininemia/enzimología , Leishmania major/genética , Leishmaniasis Cutánea/enzimología , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/patología , Ratones , Ratones Endogámicos BALB C
9.
Arch Biochem Biophys ; 461(1): 146-50, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17291445

RESUMEN

A rat brain cDNA encoding for a novel protein with agmatinase activity was cloned and functionally expressed. The protein was expressed as a histidine-tagged fusion product with a molecular weight of about 63 kDa. Agmatine hydrolysis was strictly dependent on Mn(2+); K(m) and k(cat) values were 2.5+/-0.2 mM and 0.8+/-0.2 s(-1), respectively. The product putrescine was a linear competitive inhibitor (K(i)=5+/-0.5 mM). The substrate specificity, metal ion requirement and pH optimum (9.5) coincide with those reported for Escherichia coli agmatinase, the best characterized of the agmatinases. However, as indicated by the k(cat)/K(m) (320 M(-1)s(-1)), the recombinant protein was about 290-fold less efficient than the bacterial enzyme. The deduced amino sequence revealed great differences with all known agmatinases, thus excluding the protein from the arginase family. It was, however, highly identical (>85%) to the predicted sequences for fragments of hypothetical or unnamed LIM domain-containing proteins. As a suggestion, the agmatinase activity is adscribed to a protein with an active site that promiscuously catalyze a reaction other than the one it evolved to catalyze.


Asunto(s)
Arginasa/química , Arginasa/genética , Encéfalo/enzimología , Clonación Molecular , ADN Complementario/genética , Familia de Multigenes , Ureohidrolasas/química , Ureohidrolasas/genética , Secuencia de Aminoácidos , Animales , Arginasa/fisiología , Sitios de Unión/genética , Catálisis , ADN Complementario/biosíntesis , ADN Complementario/fisiología , Humanos , Datos de Secuencia Molecular , Ratas , Ureohidrolasas/biosíntesis
10.
J Immunol ; 177(8): 5595-603, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17015747

RESUMEN

Increased arginase I activity is associated with allergic disorders such as asthma. How arginase I contributes to and is regulated by allergic inflammatory processes remains unknown. CD4+ Th2 lymphocytes (Th2 cells) and IL-13 are two crucial immune regulators that use STAT6-dependent pathways to induce allergic airways inflammation and enhanced airways responsiveness to spasmogens (airways hyperresponsiveness (AHR)). This pathway is also used to activate arginase I in isolated cells and in hepatic infection with helminths. In the present study, we show that arginase I expression is also regulated in the lung in a STAT6-dependent manner by Th2-induced allergic inflammation or by IL-13 alone. IL-13-induced expression of arginase I correlated directly with increased synthesis of urea and with reduced synthesis of NO. Expression of arginase I, but not eosinophilia or mucus hypersecretion, temporally correlated with the development, persistence, and resolution of IL-13-induced AHR. Pharmacological supplementation with l-arginine or with NO donors amplified or attenuated IL-13-induced AHR, respectively. Moreover, inducing loss of function of arginase I specifically in the lung by using RNA interference abrogated the development of IL-13-induced AHR. These data suggest an important role for metabolism of l-arginine by arginase I in the modulation of IL-13-induced AHR and identify a potential pathway distal to cytokine receptor interactions for the control of IL-13-mediated bronchoconstriction in asthma.


Asunto(s)
Arginasa/antagonistas & inhibidores , Arginasa/fisiología , Hiperreactividad Bronquial/etiología , Interleucina-13/fisiología , Interferencia de ARN , Animales , Arginasa/genética , Arginasa/metabolismo , Arginina/metabolismo , Asma/etiología , Asma/metabolismo , Regulación Enzimológica de la Expresión Génica , Pulmón/enzimología , Pulmón/patología , Ratones , Ratones Noqueados , Donantes de Óxido Nítrico/metabolismo , Factor de Transcripción STAT6 , Células Th2
11.
J Immunol ; 173(3): 2109-17, 2004 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15265947

RESUMEN

Arginase is the endogenous inhibitor of inducible NO synthase (iNOS), because both enzymes use the same substrate, l-arginine (Arg). Importantly, arginase synthesizes ornithine, which is metabolized by the enzyme ornithine decarboxylase (ODC) to produce polyamines. We investigated the role of these enzymes in the Citrobacter rodentium model of colitis. Arginase I, iNOS, and ODC were induced in the colon during the infection, while arginase II was not up-regulated. l-Arg supplementation of wild-type mice or iNOS deletion significantly improved colitis, and l-Arg treatment of iNOS(-/-) mice led to an additive improvement. There was a significant induction of IFN-gamma, IL-1, and TNF-alpha mRNA expression in colitis tissues that was markedly attenuated with l-Arg treatment or iNOS deletion. Treatment with the arginase inhibitor S-(2-boronoethyl)-l-cysteine worsened colitis in both wild-type and iNOS(-/-) mice. Polyamine levels were increased in colitis tissues, and were further increased by l-Arg. In addition, in vivo inhibition of ODC with alpha-difluoromethylornithine also exacerbated the colitis. Taken together, these data indicate that arginase is protective in C. rodentium colitis by enhancing the generation of polyamines in addition to competitive inhibition of iNOS. Modulation of the balance of iNOS and arginase, and of the arginase-ODC metabolic pathway may represent a new strategy for regulating intestinal inflammation.


Asunto(s)
Arginasa/fisiología , Colitis/enzimología , Infecciones por Enterobacteriaceae/enzimología , Óxido Nítrico Sintasa/biosíntesis , Ornitina Descarboxilasa/fisiología , Animales , Arginasa/antagonistas & inhibidores , Arginasa/biosíntesis , Arginasa/genética , Arginina/metabolismo , Arginina/uso terapéutico , Ácidos Borónicos/farmacología , Ácidos Borónicos/toxicidad , Citrobacter rodentium , Colitis/tratamiento farmacológico , Colitis/microbiología , Colitis/patología , Eflornitina/farmacología , Eflornitina/toxicidad , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Inducción Enzimática , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Ornitina/metabolismo , Ornitina Descarboxilasa/biosíntesis , Ornitina Descarboxilasa/genética , Poliaminas/metabolismo
12.
FASEB J ; 17(15): 2328-30, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14563685

RESUMEN

A reduction in L-arginine availability has been implicated in the impairment of endothelium-dependent nitric oxide (NO)-mediated vasodilation by ischemia-reperfusion (I/R). However, the mechanisms contributing to dysregulation of the L-arginine pool remain unknown. Because endothelial cells can metabolize L-arginine via two major enzymes, that is, NO synthase (NOS) and arginase, we hypothesized that up-regulation of arginase during I/R reduces L-arginine availability to NOS and thus impairs NO-mediated vasodilation. To test this hypothesis, a local I/R was produced in the porcine heart by occlusion of a small branch of left anterior descending artery for 30 min, followed by reperfusion for 90 min. Arterioles (60-110 microm) isolated from non-ischemic and ischemic regions of subepicardium were cannulated and pressurized without flow for in vitro study. Vessels from both regions developed similar levels of basal tone. Although the dilation of I/R vessels to endothelium-independent agonist sodium nitroprusside was not altered, the endothelium-dependent NO-mediated dilations to adenosine and serotonin were attenuated. I/R not only inhibited arteriolar production of NO but also increased arteriolar arginase activity. Arginase inhibitor alpha-difluoromethylornithine enhanced NO production/dilation in normal vessels and also restored the NO-mediated function in I/R vessels. Treating I/R vessels with L-arginine also restored vasodilations. Immunohistochemical data revealed that I/R up-regulated arginase but down-regulated NOS expression in the arteriolar endothelium. Pretreating the animals with protein synthesis inhibitor cycloheximide prevented I/R-induced arginase up-regulation and also preserved NO-mediated vascular function. These results suggest that one mechanism by which I/R inhibits NO-mediated arteriolar dilation is through increased arginase activity, which limits the availability of L-arginine to NOS for NO production. In addition, the inability of arginase blockade or L-arginine supplementation to completely restore vasodilatory function may be attributable to the down-regulation of endothelial NOS expression.


Asunto(s)
Arginasa/fisiología , Vasos Coronarios/fisiopatología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/fisiopatología , Óxido Nítrico/metabolismo , Vasodilatación , Adenosina/farmacología , Animales , Arginasa/antagonistas & inhibidores , Arginina/farmacología , Arteriolas/efectos de los fármacos , Arteriolas/enzimología , Arteriolas/fisiopatología , Vasos Coronarios/enzimología , Técnicas de Cultivo , Eflornitina/farmacología , Inhibidores Enzimáticos/farmacología , Modelos Biológicos , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo III , Serotonina/farmacología , Porcinos , Vasodilatadores/farmacología
13.
J Nutr ; 131(10): 2732-40, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11584097

RESUMEN

Arginine is an intermediate of the ornithine cycle and serves as a precursor for the synthesis of nitric oxide, creatine, agmatine and proteins. It is considered to be a conditionally essential amino acid because endogenous synthesis only barely meets daily requirements. In rapidly growing suckling neonates, endogenous arginine biosynthesis is crucial to compensate for the insufficient supply of arginine via the milk. Evidence is accumulating that the intestine rather than the kidney plays a major role in arginine synthesis in this period. Accordingly, ectopic expression of hepatic arginase in murine enterocytes by genetic modification induces a selective arginine deficiency. The ensuing phenotype, whose severity correlates with the level of transgene expression in the enterocytes, could be reversed with arginine supplementation. We analyzed the effect of arginine deficiency on guanidine metabolism and neuromotor behavior. Arginine-deficient transgenic mice continued to suffer from an arginine deficiency after the arginine biosynthetic enzymes had disappeared from the enterocytes. Postweaning catch-up growth in arginine-deficient mice was characterized by increased levels of all measured amino acids except arginine. Furthermore, plasma total amino acid concentration, including arginine, was significantly lower in adult male than in adult female transgenic mice. Decreases in the concentration of plasma and tissue arginine led to significant decreases in most metabolites of arginine. However, the accumulation of the toxic guanidino compounds, guanidinosuccinic acid and methylguanidine, corresponded inversely with circulating arginine concentration, possibly reflecting a higher oxidative stress under hypoargininemic conditions. In addition, hypoargininemia was associated with disturbed neuromotor behavior, although brain levels of toxic guanidino compounds and ammonia were normal.


Asunto(s)
Aminoácidos/sangre , Arginasa/fisiología , Arginina/deficiencia , Guanidinas/metabolismo , Análisis de Varianza , Animales , Arginasa/metabolismo , Arginina/metabolismo , Conducta Animal , Intestinos/enzimología , Ratones , Ratones Transgénicos
14.
Cell ; 87(5): 845-55, 1996 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-8945512

RESUMEN

Viral envelope (Env)-receptor interactions have been implicated in the cell death associated with infection by subgroups B and D avian leukosis-sarcoma viruses (ALVs). A chicken protein, CAR1, was identified that permitted infection of mammalian cells by these viral subgroups. CAR1 bound to a viral Env fusion protein, comprising an ALV-B surface Env protein and the Fc region of an immunoglobulin, indicating that it is a specific viral receptor. CAR1 contains two extracellular cysteine-rich domains characteristic of the TNFR family and a cytoplasmic region strikingly similar to the death domain of TNFR1 and Fas, implicating this receptor in cell killing. Chicken embryo fibroblasts susceptible to ALV-B infection and transfected quail QT6 cells expressing CAR1 underwent apoptosis in response to the Env-Ig fusion protein, demonstrating that this cytopathic ALV receptor can mediate cell death.


Asunto(s)
Alpharetrovirus/química , Apoptosis/fisiología , Arginasa/fisiología , Proteínas Fúngicas/fisiología , Proteínas de la Membrana/fisiología , Células 3T3/química , Células 3T3/citología , Células 3T3/virología , Alpharetrovirus/genética , Alpharetrovirus/metabolismo , Animales , Secuencia de Bases , Células COS/química , Células COS/citología , Células COS/virología , Clonación Molecular , ADN Complementario/aislamiento & purificación , Genes Virales/fisiología , Genoma , Humanos , Ratones , Datos de Secuencia Molecular , Unión Proteica/fisiología , Codorniz , Receptores del Factor de Necrosis Tumoral/fisiología , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA