Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Genet Metab ; 143(1-2): 108531, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-39053125

RESUMEN

PMM2-CDG is the most common congenital disorder of glycosylation (CDG). Patients with this disease often carry compound heterozygous mutations of the gene encoding the phosphomannomutase 2 (PMM2) enzyme. PMM2 converts mannose-6-phosphate (M6P) to mannose-1-phosphate (M1P), which is a critical upstream metabolite for proper protein N-glycosylation. Therapeutic options for PMM2-CDG patients are limited to management of the disease symptoms, as no drug is currently approved to treat this disease. GLM101 is a M1P-loaded liposomal formulation being developed as a candidate drug to treat PMM2-CDG. This report describes the effect of GLM101 treatment on protein N-glycosylation of PMM2-CDG patient-derived fibroblasts. This treatment normalized intracellular GDP-mannose, increased the relative glycoprotein mannosylation content and TNFα-induced ICAM-1 expression. Moreover, glycomics profiling revealed that GLM101 treatment of PMM2-CDG fibroblasts resulted in normalization of most high mannose glycans and partial correction of multiple complex and hybrid glycans. In vivo characterization of GLM101 revealed its favorable pharmacokinetics, liver-targeted biodistribution, and tolerability profile with achieved systemic concentrations significantly greater than its effective in vitro potency. Taken as a whole, the results described in this report support further exploration of GLM101's safety, tolerability, and efficacy in PMM2-CDG patients.

2.
J Inherit Metab Dis ; 46(6): 1089-1103, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37494004

RESUMEN

Maple syrup urine disease (MSUD) is an inborn error of branched-chain amino acid metabolism affecting several thousand individuals worldwide. MSUD patients have elevated levels of plasma leucine and its metabolic product α-ketoisocaproate (KIC), which can lead to severe neurotoxicity, coma, and death. Patients must maintain a strict diet of protein restriction and medical formula, and periods of noncompliance or illness can lead to acute metabolic decompensation or cumulative neurological impairment. Given the lack of therapeutic options for MSUD patients, we sought to develop an oral enzyme therapy that can degrade leucine within the gastrointestinal tract prior to its systemic absorption and thus enable patients to maintain acceptable plasma leucine levels while broadening their access to natural protein. We identified a highly active leucine decarboxylase enzyme from Planctomycetaceae bacterium and used directed evolution to engineer the enzyme for stability to gastric and intestinal conditions. Following high-throughput screening of over 12 000 enzyme variants over 9 iterative rounds of evolution, we identified a lead variant, LDCv10, which retains activity following simulated gastric or intestinal conditions in vitro. In intermediate MSUD mice or healthy nonhuman primates given a whey protein meal, oral treatment with LDCv10 suppressed the spike in plasma leucine and KIC and reduced the leucine area under the curve in a dose-dependent manner. Reduction in plasma leucine correlated with decreased brain leucine levels following oral LDCv10 treatment. Collectively, these data support further development of LDCv10 as a potential new therapy for MSUD patients.


Asunto(s)
Enfermedad de la Orina de Jarabe de Arce , Humanos , Ratones , Animales , Leucina , Aminoácidos de Cadena Ramificada , Proteínas , Terapia Enzimática , Primates/metabolismo
3.
Mol Genet Metab ; 139(4): 107653, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37463544

RESUMEN

Classical homocystinuria (HCU) is a rare inborn error of amino acid metabolism characterized by accumulation of homocysteine, an intermediate product of methionine metabolism, leading to significant systemic toxicities, particularly within the vascular, skeletal, and ocular systems. Most patients require lifelong dietary therapy with severe restriction of natural protein to minimize methionine intake, and many patients still struggle to maintain healthy homocysteine levels. Since eliminating methionine from the diet reduces homocysteine levels, we hypothesized that an enzyme that can degrade methionine within the gastrointestinal (GI) tract could help HCU patients maintain healthy levels while easing natural protein restrictions. We describe the preclinical development of CDX-6512, a methionine gamma lyase (MGL) enzyme that was engineered for stability and activity within the GI tract for oral administration to locally degrade methionine. CDX-6512 is stable to low pH and intestinal proteases, enabling it to survive the harsh GI environment without enteric coating and to degrade methionine freed from dietary protein within the small intestine. Administering CDX-6512 to healthy non-human primates following a high protein meal led to a dose-dependent suppression of plasma methionine. In Tg-I278T Cbs-/- mice, an animal model that recapitulates aspects of HCU disease including highly elevated serum homocysteine levels, oral dosing of CDX-6512 after a high protein meal led to suppression in serum levels of both methionine and homocysteine. When animals received a daily dose of CDX-6512 with a high protein meal for two weeks, the Tg-I278T Cbs-/- mice maintained baseline homocysteine levels, whereas homocysteine levels in untreated animals increased by 39%. These preclinical data demonstrate the potential of CDX-6512 as an oral enzyme therapy for HCU.


Asunto(s)
Homocistinuria , Humanos , Ratones , Animales , Homocistinuria/tratamiento farmacológico , Homocistinuria/genética , Metionina/metabolismo , Homocisteína , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Racemetionina , Tracto Gastrointestinal/metabolismo
4.
Sci Rep ; 13(1): 4748, 2023 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-36959353

RESUMEN

Fabry disease is caused by a deficiency of α-galactosidase A (GLA) leading to the lysosomal accumulation of globotriaosylceramide (Gb3) and other glycosphingolipids. Fabry patients experience significant damage to the heart, kidney, and blood vessels that can be fatal. Here we apply directed evolution to generate more stable GLA variants as potential next generation treatments for Fabry disease. GLAv05 and GLAv09 were identified after screening more than 12,000 GLA variants through 8 rounds of directed evolution. Both GLAv05 and GLAv09 exhibit increased stability at both lysosomal and blood pH, stability to serum, and elevated enzyme activity in treated Fabry fibroblasts (19-fold) and GLA-/- podocytes (10-fold). GLAv05 and GLAv09 show improved pharmacokinetics in mouse and non-human primates. In a Fabry mouse model, the optimized variants showed prolonged half-lives in serum and relevant tissues, and a decrease of accumulated Gb3 in heart and kidney. To explore the possibility of diminishing the immunogenic potential of rhGLA, amino acid residues in sequences predicted to bind MHC II were targeted in late rounds of GLAv09 directed evolution. An MHC II-associated peptide proteomics assay confirmed a reduction in displayed peptides for GLAv09. Collectively, our findings highlight the promise of using directed evolution to generate enzyme variants for more effective treatment of lysosomal storage diseases.


Asunto(s)
Enfermedad de Fabry , Humanos , Ratones , Animales , Enfermedad de Fabry/tratamiento farmacológico , Enfermedad de Fabry/genética , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo , Riñón/metabolismo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo
7.
J Biol Chem ; 282(44): 31882-90, 2007 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-17766241

RESUMEN

The liver X receptors (LXRalpha/beta) are part of the nuclear receptor family and are believed to regulate cholesterol and lipid homeostasis. It has also been suggested that LXR agonists possess anti-inflammatory properties. The aim of this work was to determine the effect of LXR agonists on the innate immune response in human primary lung macrophages and a pre-clinical rodent model of lung inflammation. Before profiling the impact of the agonist, we established that both the human macrophages and the rodent lungs expressed LXRalpha/beta. We then used two structurally distinct LXR agonists to demonstrate that activation of this transcription factor reduces cytokine production in THP-1 cells and lung macrophages. Then, using the expression profile of ATP binding cassettes A1 (ABCA-1; a gene directly linked to LXR activation) as a biomarker for lung exposure of the compound, we demonstrated an LXR-dependent reduction in lung neutrophilia rodents in vivo. This inhibition was not associated with a suppression of c-Fos/c-Jun mRNA expression or NF-kappaB/AP-1 DNA binding, suggesting that any anti-inflammatory activity of LXR agonists is not via inhibition of NF-kappaB/AP-1 transcriptional activity. These data do not completely rule out an impact of these agonists on these two prominent transcription factors. In summary, this study is the first to demonstrate anti-inflammatory actions of LXRs in the lung. Chronic innate inflammatory responses observed in some airway diseases is thought to be central to disease pathogenesis. Therefore, data suggest that LXR ligands have utility in the treatment of lung diseases that involves chronic inflammation mediated by macrophages and neutrophils.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Macrófagos Alveolares/inmunología , Neumonía/inmunología , Receptores Citoplasmáticos y Nucleares/inmunología , Animales , Línea Celular , Proteínas de Unión al ADN/agonistas , Proteínas de Unión al ADN/genética , Endotoxinas/inmunología , Humanos , Receptores X del Hígado , Masculino , Receptores Nucleares Huérfanos , Ratas , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/genética
8.
J Pharmacol Exp Ther ; 319(1): 468-76, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16861399

RESUMEN

Phosphodiesterase type 4 (PDE(4)) inhibitors are currently being evaluated as potential therapies for inflammatory airway diseases. However, this class of compounds has been shown to cause an arteritis/vasculitis of unknown etiology in rats and cynomolgus monkeys. Studies in rodents have demonstrated the anti-inflammatory effects of PDE(4) inhibitors on lipopolysaccharide (LPS)-induced airway inflammation. The aim of this work was to assess the direct effects of PDE(4) inhibitors on inflammatory cells and cytokine levels in the lung in relation to therapeutic effects. The effects of the PDE(4) inhibitors 3-cyclo-propylmethoxy-4-difluoromethoxy-N-[3,5-di-chloropyrid-4-yl]-benzamide (roflumilast) and 3-(cyclopentyloxy)-N-(3,5-dichloro-4-pyridyl)-4-methoxybenzamide (piclamilast) were assessed in vivo, using BALB/c mice, and in vitro, in unstimulated human endothelial and epithelial cell lines. In BALB/c mice, LPS challenge caused an increase in neutrophils in bronchoalveolar lavage (BAL) and lung tissue and BAL tumor necrosis factor-alpha levels, which were inhibited by treatment with either roflumilast or piclamilast (30-100 mg/kg subcutaneously). However, roflumilast and piclamilast alone (100 mg/kg) caused a significant increase in plasma and lung tissue keratinocyte-derived chemokine (KC) levels, and lung tissue neutrophils. In vitro, both piclamilast and roflumilast caused an increase in interleukin (IL)-8 release from human umbilical vein endothelial cells but not BEAS-2B cells, suggesting that one source of the increased KC may be endothelial cells. At doses that antagonized an LPS-induced inflammatory response, the PDE(4) inhibitors possessed proinflammatory activities in the lung that may limit their therapeutic potential. The proinflammatory cytokines KC and IL-8 therefore may provide surrogate biomarkers, both in preclinical animal models and in the clinic, to assess potential proinflammatory effects of this class of compounds.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , Inflamación/inducido químicamente , Inhibidores de Fosfodiesterasa/toxicidad , Aminopiridinas/farmacología , Animales , Benzamidas/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Ciclopropanos/farmacología , Citocinas/biosíntesis , Humanos , Interleucina-8/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Piridinas/farmacología
9.
Mol Pharmacol ; 69(6): 1791-800, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16517756

RESUMEN

Nuclear factor kappaB (NF-kappaB) is a transcription factor believed to be central in the expression of numerous inflammatory genes and the pathogenesis of many respiratory diseases. We have previously demonstrated increased NF-kappaB pathway activation in a steroid-sensitive animal model of lipopolysaccharide (LPS)-driven airway inflammation. It is noteworthy that this phenomenon was not observed in a steroid-insensitive model of elastase-induced inflammation in the rat. The aim of this study was to gather further evidence to suggest that these similar profiles of neutrophilic inflammation can be NF-kappaB-dependent or -independent by determining the impact of an IkappaB kinase-2 (IKK-2) inhibitor, 2-[(aminocarbonyl)amino]-5-(4-fluorophenyl)-3-thiophenecarboxamide (TPCA-1). In the LPS model, TPCA-1 blocked the increase in NF-kappaB DNA binding, a marker of NF-kappaB pathway activation. This inhibition was associated with a reduction in inflammatory mediator release [tumor necrosis factor alpha (TNFalpha)/interleukin-1beta (IL-1beta)/matrix metalloproteinase-9 (MMP-9)] and lung inflammatory cell burden (neutrophilia/eosinophilia). These data were paralleled with a steroid and in human cell based assays. In the elastase-driven inflammation model, in which our group has previously failed to measure an increase in NF-kappaB DNA binding, neither TPCA-1 nor the steroid, affected mediator release (IL-1beta/MMP-9) or cellular burden (neutrophilia/lymphomononuclear cells). This is the first study to examine the effect of an IKK-2 inhibitor in well validated models that mimic aspects of the inflammatory lesion evident in diseases such as COPD. In conclusion, we have demonstrated that animal models with similar profiles of airway inflammation can be IKK-2 inhibitor/steroid-sensitive or -insensitive. If both profiles of inflammation exist in the clinic, then this finding is extremely exciting and may lead to greater understanding of disease pathology and the discovery of novel anti-inflammatory targets.


Asunto(s)
Amidas/farmacología , Antiinflamatorios no Esteroideos/farmacología , Inhibidores Enzimáticos/farmacología , Quinasa I-kappa B/antagonistas & inhibidores , Enfermedades Pulmonares/enzimología , Tiofenos/farmacología , Animales , ADN/metabolismo , Modelos Animales de Enfermedad , Humanos , Quinasa I-kappa B/metabolismo , Interleucina-1/metabolismo , Lipopolisacáridos/toxicidad , Enfermedades Pulmonares/inducido químicamente , Enfermedades Pulmonares/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Elastasa Pancreática/toxicidad , Ratas , Factor de Necrosis Tumoral alfa/metabolismo
10.
J Pharmacol Exp Ther ; 316(3): 1318-27, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16368902

RESUMEN

The exact role of p38 mitogen-activated protein kinase (MAPK) in the expression of inflammatory cytokines is not clear; it may regulate transcriptionally, post-transcriptionally, translationally, or post-translationally. The involvement of one or more of these mechanisms has been suggested to depend on the particular cytokine, the cell type studied, and the specific stimulus used. Interpretation of some of the published data is further complicated by the use of inhibitors such as 4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)-1H-imidazole (SB 203580) used at single, high concentrations. The aim of this study was to determine the impact of two second-generation p38 MAPK inhibitors on the expression of a range of inflammatory cytokines at the gene and protein levels in human cultured cells. Similar assessment of the impact of these compounds on inflammatory cytokine expression in a preclinical in vivo model of airway inflammation was performed. The results in THP-1 cells and primary airway macrophages clearly show that protein expression is inhibited at much lower concentrations of inhibitor than are needed to impact on gene expression. In the rodent model, both compounds, at doses that cause maximal inhibition of cellular recruitment, inhibit tumor necrosis factor alpha (TNFalpha) protein production without impacting on nuclear factor kappaB pathway activation or TNFalpha gene expression. In summary, the data shown here demonstrate that, although at high compound concentrations there is some level of transcriptional regulation, the predominant role of p38 MAPK in cytokine production is at the translational level. These data question whether the effect of p38 inhibitors on gene transcription is related to their potential therapeutic role as anti-inflammatory compounds.


Asunto(s)
Antiinflamatorios/farmacología , Citocinas/biosíntesis , Mediadores de Inflamación/metabolismo , Pulmón/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Animales , Dexametasona/farmacología , Humanos , Imidazoles/farmacología , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Masculino , Monocitos/metabolismo , Pirimidinas/farmacología , Ratas , Transcripción Genética/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
11.
Am J Respir Crit Care Med ; 172(8): 962-71, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16002568

RESUMEN

RATIONALE: Nuclear factor (NF)-kappaB is a transcription factor known to regulate the expression of many inflammatory genes, including cytokines, chemokines, and adhesion molecules. NF-kappaB is held inactive in the cytoplasm, bound to I-kappaB. The removal of I-kappaB, via the actions of inhibitor of kappaB (I-kappaB) kinase-2 (IKK-2), allows NF-kappaB to enter the nucleus. OBJECTIVES: To determine the impact of inhibiting IKK-2 on in vitro and in vivo models of airway inflammation. METHODS: The effect of inhibiting IKK-2 was assessed in stimulated, cultured, primary human airway smooth muscle cells and an antigen-driven rat model of lung inflammation. MEASUREMENTS: The release of cytokines from cultured cells and inflammatory cytokine expression and cellular burden in the lung were determined. MAIN RESULTS: Two structurally distinct molecules and dominant negative technology demonstrated that inhibition of IKK-2 activity completely blocked cytokine release from cultured cells, whereas the two glucocorticoid comparators had limited impact on granulocyte colony-stimulating factor, interleukin 8, and eotaxin release. In addition, in an in vivo antigen-driven model of airway inflammation, the IKK-2 inhibitor blocked NF-kappaB nuclear translocation, which was associated with a reduction in inflammatory cytokine gene and protein expression, airway eosinophilia, and late asthmatic reaction, similar in magnitude to that obtained with budesonide. CONCLUSION: This study demonstrates that inhibiting IKK-2 results in a general reduction of the inflammatory response in vitro and in vivo. Compounds of this class could have therapeutic utility in the treatment of asthma and may, in certain respects, possess a beneficial efficacy profile compared with that of a steroid.


Asunto(s)
Amidas/uso terapéutico , Asma/tratamiento farmacológico , Modelos Animales de Enfermedad , Quinasa I-kappa B/antagonistas & inhibidores , Músculo Liso/efectos de los fármacos , Sistema Respiratorio/efectos de los fármacos , Tiofenos/uso terapéutico , Amidas/inmunología , Animales , Antiinflamatorios/inmunología , Antiinflamatorios/uso terapéutico , Asma/inmunología , Asma/fisiopatología , Budesonida/inmunología , Budesonida/uso terapéutico , Células Cultivadas/efectos de los fármacos , Células Cultivadas/inmunología , Quimiocina CCL11 , Quimiocinas CC/inmunología , Dexametasona/inmunología , Dexametasona/uso terapéutico , Evaluación Preclínica de Medicamentos , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Factor Estimulante de Colonias de Granulocitos/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/inmunología , Humanos , Quinasa I-kappa B/inmunología , Inflamación , Interleucina-8/inmunología , Músculo Liso/citología , Músculo Liso/inmunología , Músculo Liso/fisiopatología , FN-kappa B/efectos de los fármacos , FN-kappa B/inmunología , Ratas , Sistema Respiratorio/citología , Sistema Respiratorio/inmunología , Sistema Respiratorio/fisiopatología , Tiofenos/inmunología
12.
Am J Respir Crit Care Med ; 172(1): 74-84, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15805185

RESUMEN

RATIONALE: Emphysema is one component of chronic obstructive pulmonary disease (COPD), a respiratory disease currently increasing in prevalence worldwide. The mainstay therapy adopted to treat patients with COPD is glucocorticoids; unfortunately, this treatment has limited impact on disease symptoms or underlying airway inflammation. OBJECTIVE: There is an urgent need to develop therapies that modify both the underlying inflammation, thought to be involved in disease progression, and the structural changes in the emphysematous lung. METHODS: We have characterized an elastase-driven model of experimental emphysema in the rat that demonstrates COPD-like airway inflammation and determined the impact of a clinically relevant glucocorticoid. MEASUREMENTS AND MAIN RESULTS: We observed an increase in lung neutrophils, lymphomononuclear cells, mucus production, and inflammatory cytokines. Also present were increases in average air space area, which are associated with emphysema-like changes in lung function, such as increased residual volume and decreased flow; these increases in area were maintained for up to 10 weeks. In addition, we observed that elastase-induced airway neutrophilia is steroid resistant. Interestingly, the inflammation observed after elastase administration was found to be temporally associated with a lack of nuclear factor-kappaB pathway activation. This apparent nuclear factor-kappaB-independent inflammation may explain why treatment with a glucocorticoid was ineffective in this preclinical model and could suggest parallels in the steroid-resistant human disease. CONCLUSION: We believe that this model, in addition to its suitability for testing therapies that may modify existing emphysema, could be useful in the search for new therapies to reduce the steroid-resistant airway inflammation evident in COPD.


Asunto(s)
Antiinflamatorios/farmacología , Budesonida/farmacología , FN-kappa B/metabolismo , Neumonía/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Transducción de Señal , Algoritmos , Animales , Antiinflamatorios/uso terapéutico , Budesonida/uso terapéutico , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Resistencia a Medicamentos , Enfisema/metabolismo , Masculino , Elastasa Pancreática , Neumonía/inducido químicamente , Neumonía/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/patología , Ratas , Ratas Sprague-Dawley
13.
J Pharmacol Exp Ther ; 311(2): 625-33, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15226380

RESUMEN

Lipopolysaccharide (LPS) is known to generate nitric oxide (NO) in the airway through the activation of nitric-oxide synthase (NOS). The functional consequences of this on the inflammatory response are not clear, with conflicting data published. In the clinic, exhaled NO (ex-NO) is used as a noninvasive biomarker to assess the extent of airway inflammation. It is proposed that monitoring levels of ex-NO could be a useful guide to determining the effectiveness of disease modifying therapies. The aim was, using pharmacological tools, to determine the role of NO in an aerosolized LPS-driven animal model of airway inflammation by assessment of ex-NO, neutrophilia, and inflammatory biomarkers, using a nonselective NOS inhibitor, N(G)-nitro-l-arginine methyl ester (l-NAME), and a selective inducible NOS (iNOS) inhibitor, N-3 (aminomethyl)benzyl)acetamidine (1400W). Real-time mRNA analysis of the lung tissue indicated an increased gene expression of iNOS following LPS challenge with minimal impact on constitutive NOS isoforms. LPS induced an increase in ex-NO, which appeared to correlate with the increase in iNOS gene expression and airway neutrophilia. Treatment with l-NAME and 1400W resulted in comparable reductions in ex-NO, a reduction in airway neutrophilia, but had little impact on a range of inflammatory biomarkers. This study indicates that the LPS-induced rise in ex-NO is due to enhanced iNOS activity and that NO has a role in airway neutrophilia. Additionally, it appears using ex-NO as a guide to monitoring airway inflammation may have some use, but data should be interpreted with caution when assessing therapies that may directly impact on NO formation.


Asunto(s)
Enfermedades Bronquiales/diagnóstico , Lipopolisacáridos , Neutrófilos/patología , Óxido Nítrico/análisis , Amidinas/farmacología , Animales , Bencilaminas/farmacología , Biomarcadores , Enfermedades Bronquiales/inducido químicamente , Enfermedades Bronquiales/patología , Dexametasona/farmacología , Interacciones Farmacológicas , Expresión Génica/efectos de los fármacos , Inflamación/inducido químicamente , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo
14.
Respir Res ; 4: 3, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12657158

RESUMEN

BACKGROUND: The link between eosinophils and the development of airway hyperresponsiveness (AHR) in asthma is still controversial. This question was assessed in a murine model of asthma in which we performed a dose ranging study to establish whether the dose of steroid needed to inhibit the eosinophil infiltration correlated with that needed to block AHR. METHODS: The sensitised BALB/c mice were dosed with vehicle or dexamethasone (0.01-3 mg/kg) 2 hours before and 6 hours after each challenge (once daily for 6 days) and 2 hours before AHR determination by whole-body plethysmography. At 30 minutes after the AHR to aerosolised methacholine the mice were lavaged and differential white cell counts were determined. Challenging with antigen caused a significant increase in eosinophils in the bronchoalveolar lavage (BAL) fluid and lung tissue, and increased AHR. RESULTS: Dexamethasone reduced BAL and lung tissue eosinophilia (ED50 values of 0.06 and 0.08 mg/kg, respectively), whereas a higher dose was needed to block AHR (ED50 of 0.32 mg/kg at 3 mg/ml methacholine. Dissociation was observed between the dose of steroid needed to affect AHR in comparison with eosinophilia and suggests that AHR is not a direct consequence of eosinophilia. CONCLUSION: This novel pharmacological approach has revealed a clear dissociation between eosinophilia and AHR by using steroids that are the mainstay of asthma therapy. These data suggest that eosinophilia is not associated with AHR and questions the rationale that many pharmaceutical companies are adopting in developing low-molecular-mass compounds that target eosinophil activation/recruitment for the treatment of asthma.


Asunto(s)
Antiinflamatorios/administración & dosificación , Asma/tratamiento farmacológico , Hiperreactividad Bronquial/prevención & control , Dexametasona/administración & dosificación , Eosinofilia/prevención & control , Administración por Inhalación , Animales , Antiinflamatorios/uso terapéutico , Asma/inmunología , Asma/patología , Hiperreactividad Bronquial/diagnóstico , Líquido del Lavado Bronquioalveolar/citología , Broncoconstrictores/administración & dosificación , Dexametasona/uso terapéutico , Relación Dosis-Respuesta a Droga , Eosinofilia/inmunología , Eosinofilia/patología , Inmunización , Pulmón/patología , Masculino , Cloruro de Metacolina/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/inmunología
15.
J Pharmacol Exp Ther ; 304(3): 1285-91, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12604708

RESUMEN

Excessive local production of nitric oxide (NO) has been suggested to play a role in rodent models of airway inflammation and in pulmonary diseases such as asthma. However, even given the plethora of data available including gene expression data, pharmacological data, and gene deletion studies in animal models, it is still not clear which nitric-oxide synthase (NOS) isoform is involved in eosinophilic airway inflammation. In this rat study, the nonselective NOS inhibitor L-NAME (N(G)-nitro-L-arginine methyl ester), but not a selective inducible NOS (iNOS) inhibitor 1400W (N-3-(aminomethyl)benzyl)acetamidine), impacted on Sephadex-induced inflammation by significantly inhibiting lung edema, eosinophil infiltration, tumor necrosis factor alpha, interleukin-13, and eotaxin levels in the lung tissue. Furthermore, iNOS gene expression was not induced following Sephadex administration, which confirms that iNOS does not play a role in this model. To demonstrate that this phenomenon was not restricted to this model of asthma, L-NAME, but not 1400W, was shown to reduce eosinophilia in an antigen-induced model. However, in contrast to the Sephadex model, there was an induction of iNOS gene expression after antigen challenge. In a model of aerosolized lipopolysaccharide-induced inflammation, where iNOS gene expression is increased, 1400W inhibited the increased neutrophilia. These data suggest that the compound has been administered using an appropriate dosing regimen for iNOS inhibition in the rat lung. In conclusion, it appears that constitutive, not inducible, NOS isoforms are important in NO production in models of allergic inflammation, which questions whether there is a role for iNOS inhibitors as therapy for the treatment of asthma.


Asunto(s)
Dextranos/farmacología , Eosinófilos/patología , Inflamación/enzimología , Óxido Nítrico Sintasa/fisiología , Amidinas/farmacología , Animales , Bencilaminas/farmacología , Bronquios/patología , Citocinas/metabolismo , Dexametasona/uso terapéutico , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Eosinofilia/tratamiento farmacológico , Eosinófilos/efectos de los fármacos , Inflamación/inducido químicamente , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Neutrófilos/efectos de los fármacos , Óxido Nítrico Sintasa/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Wistar
16.
Br J Pharmacol ; 137(2): 263-75, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12208784

RESUMEN

1. The antigen-induced inflammatory response in the Brown Norway rat is a model commonly used to assess the impact of novel compounds on airway eosinophilia. A detailed functional, cellular and molecular characterization of this model has not yet been performed within a single study. This information together with the temporal changes in this phenomenon should be known before this model can be used, with confidence, to elucidate the mechanisms of action of novel anti-inflammatory drugs. 2. Antigen challenge caused an accumulation of eosinophils in lung tissue 24 h after challenge. Accumulation of CD2(+) T cells was not apparent until after 72 h. 3. Interestingly, mRNA for the Th2 type cytokines interleukin (IL)-4, IL-5 and IL-13 and eotaxin were elevated in lung tissue after challenge and the expression of IL-13 and eotaxin protein increased at around 8-12 h. The temporal changes in both the biomarker production and the functional responses are important factors to consider in protocol design prior to initiating a compound screening program. 4. A neutralising antibody (R73) against alphabeta-TCR caused a significant reduction in T cell numbers accompanied by a significant suppression of eosinophil accumulation. 5. Airway hyperreactivity (AHR) was not apparent in this specific Brown Norway model in sensitized animals after a single or multiple challenges although eosinophil influx was seen in the same animals. 6. In conclusion, this is a convenient pre-clinical model (incorporating the measurement of biomarkers and functional responses) for screening novel small molecule inhibitors and/or biotherapeutics targeted against T cell/eosinophil infiltration/activation.


Asunto(s)
Modelos Animales de Enfermedad , Eosinofilia/etiología , Hipersensibilidad/etiología , Inflamación/etiología , Enfermedades Pulmonares/etiología , Animales , Biomarcadores , Hiperreactividad Bronquial , Quimiocina CCL11 , Quimiocinas CC/genética , Citocinas/genética , Eosinófilos/fisiología , Pulmón/inmunología , Pulmón/patología , Masculino , ARN Mensajero/análisis , Ratas , Ratas Endogámicas BN , Receptores de Antígenos de Linfocitos T alfa-beta/fisiología , Linfocitos T
17.
J Immunol ; 169(2): 974-82, 2002 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12097404

RESUMEN

We postulated that the seleno-organic compound ebselen would attenuate neutrophil recruitment and activation after aerosolized challenge with endotoxin (LPS) through its effect as an antioxidant and inhibitor of gene activation. Rats were given ebselen (1-100 mg/kg i.p.) followed by aerosolized LPS exposure (0.3 mg/ml for 30 min). Airway inflammatory indices were measured 4 h postchallenge. Bronchoalveolar lavage (BAL) fluid cellularity and myeloperoxidase activity were used as a measure of neutrophil recruitment and activation. RT-PCR analysis was performed in lung tissue to assess gene expression of TNF-alpha, cytokine-induced neutrophil chemoattractant-1 (CINC-1), macrophage-inflammatory protein-2 (MIP-2), ICAM-1, IL-10, and inducible NO synthase. Protein levels in lung and BAL were also determined by ELISA. Ebselen pretreatment inhibited neutrophil influx and activation as assessed by BAL fluid cellularity and myeloperoxidase activity in cell-free BAL and BAL cell homogenates. This protective effect was accompanied by a significant reduction in lung and BAL fluid TNF-alpha and IL-1 beta protein and/or mRNA levels. Ebselen pretreatment also prevented lung ICAM-1 mRNA up-regulation in response to airway challenge with LPS. This was not a global effect of ebselen on LPS-induced gene expression, because the rise in lung and BAL CINC-1 and MIP-2 protein levels were unaffected as were lung mRNA expressions for CINC-1, MIP-2, IL-10, and inducible NO synthase. These data suggest that the anti-inflammatory properties of ebselen are achieved through an inhibition of lung ICAM-1 expression possibly through an inhibition of TNF-alpha and IL-1 beta, which are potent neutrophil recruiting mediators and effective inducers of ICAM-1 expression.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Azoles/administración & dosificación , Quimiocinas CXC/biosíntesis , Péptidos y Proteínas de Señalización Intercelular , Lipopolisacáridos/toxicidad , Pulmón/efectos de los fármacos , Pulmón/patología , Infiltración Neutrófila/efectos de los fármacos , Infiltración Neutrófila/inmunología , Compuestos de Organoselenio/administración & dosificación , Aerosoles , Animales , Azoles/uso terapéutico , Bronquios/efectos de los fármacos , Bronquios/inmunología , Bronquios/metabolismo , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Quimiocina CXCL1 , Quimiocina CXCL2 , Quimiocinas CXC/fisiología , Factores Quimiotácticos/biosíntesis , Modelos Animales de Enfermedad , Sustancias de Crecimiento/biosíntesis , Inflamación/inmunología , Inyecciones Intraperitoneales , Molécula 1 de Adhesión Intercelular/biosíntesis , Interleucina-1/antagonistas & inhibidores , Interleucina-1/biosíntesis , Interleucina-1/genética , Isoindoles , Pulmón/inmunología , Masculino , Monocinas/biosíntesis , Compuestos de Organoselenio/uso terapéutico , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/biosíntesis , Ratas , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
18.
J Immunol ; 168(6): 3004-16, 2002 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11884473

RESUMEN

Intratracheal instillation of Sephadex particles is a convenient model for assessing the impact of potential anti-inflammatory compounds on lung eosinophilia thought to be a key feature in asthma pathophysiology. However, the underlying cellular and molecular mechanisms involved are poorly understood. We have studied the time course of Sephadex-induced lung eosinophilia, changes in pulmonary T cell numbers, and gene and protein expression as well as the immunological and pharmacological modulation of these inflammatory indices in the Sprague Dawley rat. Sephadex increased T cell numbers (including CD4(+) T cells) and evoked a pulmonary eosinophilia that was associated with an increase in gene/protein expression of the Th2-type cytokines IL-4, IL-5, and IL-13 and eotaxin in lung tissue. Sephadex instillation also induced airway hyperreactivity to acetylcholine and bradykinin. A neutralizing Ab (R73) against the alphabeta-TCR caused 54% depletion of total (CD2(+)) pulmonary T cells accompanied by a significant inhibition of IL-4, IL-13 and eotaxin gene expression together with suppression (65% inhibition) of eosinophils in lung tissue 24 h after Sephadex treatment. Sephadex-induced eosinophilia and Th2 cytokine gene and/or protein expression were sensitive to cyclosporin A and budesonide, compounds that inhibit T cell function, suggesting a pivotal role for T cells in orchestrating Sephadex-induced inflammation in this model.


Asunto(s)
Dextranos/toxicidad , Pulmón/inmunología , Pulmón/patología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Acetilcolina/administración & dosificación , Animales , Anticuerpos Monoclonales/administración & dosificación , Biomarcadores/análisis , Bradiquinina/administración & dosificación , Hiperreactividad Bronquial/inducido químicamente , Budesonida/uso terapéutico , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Células Cultivadas/efectos de los fármacos , Células Cultivadas/inmunología , Células Cultivadas/metabolismo , Ciclosporina/uso terapéutico , Citocinas/biosíntesis , Citocinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-2/biosíntesis , Intubación Intratraqueal , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Depleción Linfocítica , Masculino , Mastocitos/efectos de los fármacos , Mastocitos/inmunología , Eosinofilia Pulmonar/inducido químicamente , Eosinofilia Pulmonar/inmunología , Ratas , Ratas Sprague-Dawley , Subgrupos de Linfocitos T/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA