Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 42(12): 113544, 2023 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-38060381

RESUMEN

Dysregulated iron or Ca2+ homeostasis has been reported in Parkinson's disease (PD) models. Here, we discover a connection between these two metals at the mitochondria. Elevation of iron levels causes inward mitochondrial Ca2+ overflow, through an interaction of Fe2+ with mitochondrial calcium uniporter (MCU). In PD neurons, iron accumulation-triggered Ca2+ influx across the mitochondrial surface leads to spatially confined Ca2+ elevation at the outer mitochondrial membrane, which is subsequently sensed by Miro1, a Ca2+-binding protein. A Miro1 blood test distinguishes PD patients from controls and responds to drug treatment. Miro1-based drug screens in PD cells discover Food and Drug Administration-approved T-type Ca2+-channel blockers. Human genetic analysis reveals enrichment of rare variants in T-type Ca2+-channel subtypes associated with PD status. Our results identify a molecular mechanism in PD pathophysiology and drug targets and candidates coupled with a convenient stratification method.


Asunto(s)
Calcio , Enfermedad de Parkinson , Humanos , Calcio/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Preparaciones Farmacéuticas/metabolismo , Hierro/metabolismo , Mitocondrias/metabolismo
2.
Front Cell Dev Biol ; 9: 765408, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34805172

RESUMEN

Mutations in MAPT gene cause multiple neurological disorders, including frontal temporal lobar degeneration and parkinsonism. Increasing evidence indicates impaired mitochondrial homeostasis and mitophagy in patients and disease models of pathogenic MAPT. Here, using MAPT patients' fibroblasts as a model, we report that disease-causing MAPT mutations compromise early events of mitophagy. By employing biochemical and mitochondrial assays we discover that upon mitochondrial depolarization, the recruitment of LRRK2 and Parkin to mitochondria and degradation of the outer mitochondrial membrane protein Miro1 are disrupted. Using high resolution electron microscopy, we reveal that the contact of mitochondrial membranes with ER and cytoskeleton tracks is dissociated following mitochondrial damage. This membrane dissociation is blocked by a pathogenic MAPT mutation. Furthermore, we provide evidence showing that tau protein, which is encoded by MAPT gene, interacts with Miro1 protein, and this interaction is abolished by pathogenic MAPT mutations. Lastly, treating fibroblasts of a MAPT patient with a small molecule promotes Miro1 degradation following depolarization. Altogether, our results show molecular defects in a peripheral tissue of patients and suggest that targeting mitochondrial quality control may have a broad application for future therapeutic intervention.

3.
Nat Metab ; 3(9): 1242-1258, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34504353

RESUMEN

Mitochondria are the main site for generating reactive oxygen species, which are key players in diverse biological processes. However, the molecular pathways of redox signal transduction from the matrix to the cytosol are poorly defined. Here we report an inside-out redox signal of mitochondria. Cysteine oxidation of MIC60, an inner mitochondrial membrane protein, triggers the formation of disulfide bonds and the physical association of MIC60 with Miro, an outer mitochondrial membrane protein. The oxidative structural change of this membrane-crossing complex ultimately elicits cellular responses that delay mitophagy, impair cellular respiration and cause oxidative stress. Blocking the MIC60-Miro interaction or reducing either protein, genetically or pharmacologically, extends lifespan and health-span of healthy fruit flies, and benefits multiple models of Parkinson's disease and Friedreich's ataxia. Our discovery provides a molecular basis for common treatment strategies against oxidative stress.


Asunto(s)
Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Transducción de Señal , Animales , Humanos , Oxidación-Reducción , Conformación Proteica , Especies Reactivas de Oxígeno/metabolismo
4.
Nat Commun ; 12(1): 83, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33397950

RESUMEN

Trafficking of mitochondria into dendrites and axons plays an important role in the physiology and pathophysiology of neurons. Mitochondrial outer membrane protein Miro and adaptor proteins TRAKs/Milton link mitochondria to molecular motors. Here we show that metaxins MTX-1 and MTX-2 contribute to mitochondrial transport into both dendrites and axons of C. elegans neurons. MTX1/2 bind to MIRO-1 and kinesin light chain KLC-1, forming a complex to mediate kinesin-1-based movement of mitochondria, in which MTX-1/2 are essential and MIRO-1 plays an accessory role. We find that MTX-2, MIRO-1, and TRAK-1 form another distinct adaptor complex to mediate dynein-based transport. Additionally, we show that failure of mitochondrial trafficking in dendrites causes age-dependent dendrite degeneration. We propose that MTX-2 and MIRO-1 form the adaptor core for both motors, while MTX-1 and TRAK-1 specify each complex for kinesin-1 and dynein, respectively. MTX-1 and MTX-2 are also required for mitochondrial transport in human neurons, indicative of their evolutionarily conserved function.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/metabolismo , Animales , Animales Modificados Genéticamente , Axones/metabolismo , Dendritas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Cinesinas , Unión Proteica , Transporte de Proteínas
5.
Cell Metab ; 30(6): 1131-1140.e7, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31564441

RESUMEN

The identification of molecular targets and pharmacodynamic markers for Parkinson's disease (PD) will empower more effective clinical management and experimental therapies. Miro1 is localized on the mitochondrial surface and mediates mitochondrial motility. Miro1 is removed from depolarized mitochondria to facilitate their clearance via mitophagy. Here, we explore the clinical utility of Miro1 for detecting PD and for gauging potential treatments. We measure the Miro1 response to mitochondrial depolarization using biochemical assays in skin fibroblasts from a broad spectrum of PD patients and discover that more than 94% of the patients' fibroblast cell lines fail to remove Miro1 following depolarization. We identify a small molecule that can repair this defect of Miro1 in PD fibroblasts. Treating patient-derived neurons and fly models with this compound rescues the locomotor deficits and dopaminergic neurodegeneration. Our results indicate that tracking this Miro1 marker and engaging in Miro1-based therapies could open new avenues to personalized medicine.


Asunto(s)
Antiparkinsonianos/farmacología , Proteínas de Drosophila , Proteínas Mitocondriales , Degeneración Nerviosa/tratamiento farmacológico , Neuronas/efectos de los fármacos , Enfermedad de Parkinson , Proteínas de Unión al GTP rho , Adulto , Anciano , Animales , Antiparkinsonianos/uso terapéutico , Biomarcadores/metabolismo , Drosophila , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/metabolismo , Femenino , Fibroblastos , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/metabolismo , Neuronas/patología , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Proteínas de Unión al GTP rho/metabolismo
6.
Acta Neuropathol ; 136(4): 607-620, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29923074

RESUMEN

Alpha-synuclein is a component of Lewy bodies, the pathological hallmark of Parkinson's disease (PD), and is also mutated in familial PD. Here, by extensively analyzing PD patient brains and neurons, and fly models, we show that alpha-synuclein accumulation results in upregulation of Miro protein levels. Miro is a motor/adaptor on the outer mitochondrial membrane that mediates mitochondrial motility, and is removed from damaged mitochondria to facilitate mitochondrial clearance via mitophagy. PD patient neurons abnormally accumulate Miro on the mitochondrial surface leading to delayed mitophagy. Partial reduction of Miro rescues mitophagy phenotypes and neurodegeneration in human neurons and flies. Upregulation of Miro by alpha-synuclein requires an interaction via the N-terminus of alpha-synuclein. Our results highlight the importance of mitochondria-associated alpha-synuclein in human disease, and present Miro as a novel therapeutic target.


Asunto(s)
Proteínas de Drosophila/genética , Mitofagia/genética , Neuronas/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , alfa-Sinucleína/metabolismo , Proteínas de Unión al GTP rho/genética , Animales , Conducta Animal , Encéfalo/patología , Diferenciación Celular/genética , Drosophila melanogaster , Humanos , Células Madre Pluripotentes Inducidas/patología , Mitocondrias/genética , Mitocondrias/metabolismo , Trastornos del Movimiento/genética , Trastornos del Movimiento/psicología , Mutación/genética , Degeneración Nerviosa/genética , Transducción de Señal/genética , Regulación hacia Arriba , alfa-Sinucleína/genética
7.
Mol Biol Cell ; 29(10): 1219-1227, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29563254

RESUMEN

PTEN-induced putative kinase 1 (PINK1) is a mitochondria-targeted kinase whose mutations are a cause of Parkinson's disease. We set out to better understand PINK1's effects on mitochondrial proteins in vivo. Using an unbiased phosphoproteomic screen in Drosophila, we found that PINK1 mediates the phosphorylation of MCAD, a mitochondrial matrix protein critical to fatty acid metabolism. By mimicking phosphorylation of this protein in a PINK1 null background, we restored PINK1 null's climbing, flight, thorax, and wing deficiencies. Owing to MCAD's role in fatty acid metabolism, we examined the metabolic profile of PINK1 null flies, where we uncovered significant disruptions in both acylcarnitines and amino acids. Some of these disruptions were rescued by phosphorylation of MCAD, consistent with MCAD's rescue of PINK1 null's organismal phenotypes. Our work validates and extends the current knowledge of PINK1, identifies a novel function of MCAD, and illuminates the need for and effectiveness of metabolic profiling in models of neurodegenerative disease.


Asunto(s)
Acil-CoA Deshidrogenasa/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Acil-CoA Deshidrogenasa/genética , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Aminoácidos/deficiencia , Aminoácidos/metabolismo , Animales , Animales Modificados Genéticamente , Carnitina/análogos & derivados , Carnitina/metabolismo , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Oxidación-Reducción , Fenotipo , Fosforilación , Fosfoserina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
8.
Mol Cell ; 69(5): 744-756.e6, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29456190

RESUMEN

Mitochondrial crista structure partitions vital cellular reactions and is precisely regulated by diverse cellular signals. Here, we show that, in Drosophila, mitochondrial cristae undergo dynamic remodeling among distinct subcellular regions and the Parkinson's disease (PD)-linked Ser/Thr kinase PINK1 participates in their regulation. Mitochondria increase crista junctions and numbers in selective subcellular areas, and this remodeling requires PINK1 to phosphorylate the inner mitochondrial membrane protein MIC60/mitofilin, which stabilizes MIC60 oligomerization. Expression of MIC60 restores crista structure and ATP levels of PINK1-null flies and remarkably rescues their behavioral defects and dopaminergic neurodegeneration. In an extension to human relevance, we discover that the PINK1-MIC60 pathway is conserved in human neurons, and expression of several MIC60 coding variants in the mitochondrial targeting sequence found in PD patients in Drosophila impairs crista junction formation and causes locomotion deficits. These findings highlight the importance of maintenance and plasticity of crista junctions to cellular homeostasis in vivo.


Asunto(s)
Proteínas de Drosophila/metabolismo , Membranas Mitocondriales/metabolismo , Neuronas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Proteínas de Drosophila/genética , Drosophila melanogaster , Humanos , Membranas Mitocondriales/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Neuronas/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Fosforilación/genética , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética
9.
Mol Biol Cell ; 28(24): 3471-3479, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-28904209

RESUMEN

MIC60/mitofilin constitutes a hetero-oligomeric complex on the inner mitochondrial membranes to maintain crista structure. However, little is known about its physiological functions. Here, by characterizing Drosophila MIC60 mutants, we define its roles in vivo. We discover that MIC60 performs dual functions to maintain mitochondrial homeostasis. In addition to its canonical role in crista membrane structure, MIC60 regulates mitochondrial motility, likely by influencing protein levels of the outer mitochondrial membrane protein Miro that anchors mitochondria to the microtubule motors. Loss of MIC60 causes loss of Miro and mitochondrial arrest. At a cellular level, loss of MIC60 disrupts synaptic structure and function at the neuromuscular junctions. The dual roles of MIC60 in both mitochondrial crista structure and motility position it as a crucial player for cellular integrity and survival.


Asunto(s)
Proteínas de Drosophila/metabolismo , Mitocondrias/fisiología , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas Musculares/metabolismo , Animales , Movimiento Celular/fisiología , Drosophila , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Membranas Mitocondriales/fisiología , Unión Neuromuscular/metabolismo , Unión Neuromuscular/fisiología
10.
Neuromethods ; 123: 49-66, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29977105

RESUMEN

Mitochondria are among a cell's most vital organelles. They not only produce the majority of the cell's ATP but also play a key role in Ca2+ buffering and apoptotic signaling. While proper allocation of mitochondria is critical to all cells, it is particularly important for the highly polarized neurons. Because mitochondria are mainly synthesized in the soma, they must be transported long distances to be distributed to the far-flung reaches of the neuron-up to 1 m in the case of some human motor neurons. Furthermore, damaged mitochondria can be detrimental to neuronal health, causing oxidative stress and even cell death, therefore the retrograde transport of damaged mitochondria back to the soma for proper disposal, as well as the anterograde transport of fresh mitochondria from the soma to repair damage, are equally critical. Intriguingly, errors in mitochondrial transport have been increasingly implicated in neurological disorders. Here, we describe how to investigate mitochondrial transport in three complementary neuronal systems: cultured induced pluripotent stem cell-derived neurons, cultured rat hippocampal and cortical neurons, and Drosophila larval neurons in vivo. These models allow us to uncover the molecular and cellular mechanisms underlying transport issues that may occur under physiological or pathological conditions.

11.
J Nat Sci ; 2(10)2016.
Artículo en Inglés | MEDLINE | ID: mdl-27747291

RESUMEN

Chronic Fatigue Syndrome (CFS) is a debilitating disease characterized by physical and mental exhaustion. The underlying pathogenesis is unknown, but impairments in certain mitochondrial functions have been found in some CFS patients. To thoroughly reveal mitochondrial deficiencies in CFS patients, here we examine the key aspects of mitochondrial function in blood cells from a paired CFS patient-control series. Surprisingly, we discover that in patients the ATP levels are higher and mitochondrial cristae are more condensed compared to their paired controls, while the mitochondrial crista length, mitochondrial size, shape, density, membrane potential, and enzymatic activities of the complexes in the electron transport chain remain intact. We further show that the increased ATP largely comes from non-mitochondrial sources. Our results indicate that the fatigue symptom in this cohort of patients is unlikely caused by lack of ATP and severe mitochondrial malfunction. On the contrary, it might be linked to a pathological mechanism by which more ATP is produced by non-mitochondrial sources.

12.
Cell Stem Cell ; 19(6): 709-724, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27618216

RESUMEN

Mitochondrial movements are tightly controlled to maintain energy homeostasis and prevent oxidative stress. Miro is an outer mitochondrial membrane protein that anchors mitochondria to microtubule motors and is removed to stop mitochondrial motility as an early step in the clearance of dysfunctional mitochondria. Here, using human induced pluripotent stem cell (iPSC)-derived neurons and other complementary models, we build on a previous connection of Parkinson's disease (PD)-linked PINK1 and Parkin to Miro by showing that a third PD-related protein, LRRK2, promotes Miro removal by forming a complex with Miro. Pathogenic LRRK2G2019S disrupts this function, delaying the arrest of damaged mitochondria and consequently slowing the initiation of mitophagy. Remarkably, partial reduction of Miro levels in LRRK2G2019S human neuron and Drosophila PD models rescues neurodegeneration. Miro degradation and mitochondrial motility are also impaired in sporadic PD patients. We reveal that prolonged retention of Miro, and the downstream consequences that ensue, may constitute a central component of PD pathogenesis.


Asunto(s)
Proteínas Mitocondriales/metabolismo , Mitofagia , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Proteolisis , Proteínas de Unión al GTP rho/metabolismo , Animales , Axones/metabolismo , Línea Celular , Neuronas Dopaminérgicas/metabolismo , Drosophila melanogaster/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/genética , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Mitocondrias/metabolismo , Actividad Motora , Mutación/genética , Degeneración Nerviosa/complicaciones , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Neuroprotección , Enfermedad de Parkinson/complicaciones , Unión Proteica , Proteínas Quinasas/metabolismo , Interferencia de ARN , Transducción de Señal , Estrés Fisiológico , Ubiquitina-Proteína Ligasas/metabolismo
13.
PLoS Pathog ; 11(8): e1005116, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26305877

RESUMEN

The small GTPase Rab27a has been shown to control membrane trafficking and microvesicle transport pathways, in particular the secretion of exosomes. In the liver, high expression of Rab27a correlates with the development of hepatocellular carcinoma. We discovered that low abundance of Rab27a resulted in decreased hepatitis C virus (HCV) RNA and protein abundances in virus-infected cells. Curiously, both cell-associated and extracellular virus yield decreased in Rab27a depleted cells, suggesting that reduced exosome secretion did not cause the observed effect. Instead, Rab27a enhanced viral RNA replication by a mechanism that involves the liver-specific microRNA miR-122. Rab27a surrounded lipid droplets and was enriched in membrane fractions that harbor viral replication proteins, suggesting a supporting role for Rab27a in viral gene expression. Curiously, Rab27a depletion decreased the abundance of miR-122, whereas overexpression of miR-122 in Rab27a-depleted cells rescued HCV RNA abundance. Because intracellular HCV RNA abundance is enhanced by the binding of two miR-122 molecules to the extreme 5' end of the HCV RNA genome, the diminished amounts of miR-122 in Rab27a-depleted cells could have caused destabilization of HCV RNA. However, the abundance of HCV RNA carrying mutations on both miR-122-binding sites and whose stability was supported by ectopically expressed miR-122 mimetics with compensatory mutations also decreased in Rab27a-depleted cells. This result indicates that the effect of Rab27a depletion on HCV RNA abundance does not depend on the formation of 5' terminal HCV/miR-122 RNA complexes, but that miR-122 has a Rab27a-dependent function in the HCV lifecycle, likely the downregulation of a cellular inhibitor of HCV gene expression. These findings suggest that the absence of miR-122 results in a vulnerability not only to exoribonucleases that attack the viral genome, but also to upregulation of one more cellular factor that inhibit viral gene expression.


Asunto(s)
Regulación Viral de la Expresión Génica/fisiología , Hepacivirus/fisiología , MicroARNs/metabolismo , Replicación Viral/fisiología , Proteínas de Unión al GTP rab/metabolismo , Northern Blotting , Western Blotting , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/virología , Línea Celular Tumoral , Electroporación , Exosomas/metabolismo , Exosomas/virología , Técnica del Anticuerpo Fluorescente , Interacciones Huésped-Patógeno/fisiología , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/virología , ARN Interferente Pequeño , ARN Viral/biosíntesis , Transfección , Proteínas rab27 de Unión a GTP
14.
Sci Rep ; 4: 6962, 2014 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-25376463

RESUMEN

Mutations in the mitochondrial Ser/Thr kinase PINK1 cause Parkinson's disease. One of the substrates of PINK1 is the outer mitochondrial membrane protein Miro, which regulates mitochondrial transport. In this study, we uncovered novel physiological functions of PINK1-mediated phosphorylation of Miro, using Drosophila as a model. We replaced endogenous Drosophila Miro (DMiro) with transgenically expressed wildtype, or mutant DMiro predicted to resist PINK1-mediated phosphorylation. We found that the expression of phospho-resistant DMiro in a DMiro null mutant background phenocopied a subset of phenotypes of PINK1 null. Specifically, phospho-resistant DMiro increased mitochondrial movement and synaptic growth at larval neuromuscular junctions, and decreased the number of dopaminergic neurons in adult brains. Therefore, PINK1 may inhibit synaptic growth and protect dopaminergic neurons by phosphorylating DMiro. Furthermore, muscle degeneration, swollen mitochondria and locomotor defects found in PINK1 null flies were not observed in phospho-resistant DMiro flies. Thus, our study established an in vivo platform to define functional consequences of PINK1-mediated phosphorylation of its substrates.


Asunto(s)
Encéfalo/metabolismo , Neuronas Dopaminérgicas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Mitocondrias/metabolismo , Unión Neuromuscular/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Unión al GTP rho/metabolismo , Animales , Animales Modificados Genéticamente , Encéfalo/patología , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/patología , Proteínas de Drosophila/deficiencia , Drosophila melanogaster/genética , Regulación de la Expresión Génica , Humanos , Larva/genética , Larva/metabolismo , Locomoción/genética , Mitocondrias/genética , Mitocondrias/patología , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Músculos/metabolismo , Músculos/patología , Mutación , Unión Neuromuscular/genética , Unión Neuromuscular/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Fenotipo , Fosforilación , Proteínas Serina-Treonina Quinasas/deficiencia , Transducción de Señal , Sinapsis/metabolismo , Sinapsis/patología , Proteínas de Unión al GTP rho/genética
15.
Inflamm Allergy Drug Targets ; 11(4): 251-61, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22452605

RESUMEN

AIMS: This study determines whether KMUP-1 inhalation suppresses ovalbumine (OVA)-sensitized and - challenged peri-bronchial vascular inflammation and remodeling in mice. METHODS AND RESULTS: After short-term KMUP-1 (1-5 mM, 30 min)-nebulization and L-NAME (12 mM, 15 min)- pretreatment, endothelial nitric oxide synthase (eNOS) and matrix metalloproteinases-9 (MMP-9) expression in lung were measured by Western blotting analysis. In 28-days experiment, mice were sensitized with intraperitoneal OVA on day 1 and day 8, challenged with OVA nebulization and treated with KMUP-1 nebulization (5 mM, 30 mins) on day 21-27. Expression of eNOS, inducible nitric oxide synthase (iNOS), soluble guanylyl cyclase (sGC), protein kinase G (PKG), MMP-9, VCAM-1 and ICAM-1 were measured by Western blotting analysis. eNOS- and MMP-9-immunostaining were used for peri-vascular or peri-bronchial localization. Hematoxylin and eosin staining was used to show the vascular and bronchial wall thickness and infiltration of inflammatory cells. Cell counting and measurement of NOmetabolite (NOx) in bronchoalveolar lavage fluid (BALF) were used to examine the NO production. KMUP-1 increased eNOS and decreased MMP-9 expression. L-NAME-pretreatment reversed these changes. KMUP-1 reduced OVA-sensitized vascular and bronchial wall thickening, eNOS-immunostaining at the alveolar septa, MMP-9-immunostaining in the bronchioles and infiltrated inflammatory cells in the peri-vascular and peri-bronchiolar regions. The OVA-sensitized decrease of sGC and PKG and increase of iNOS, ICAM-1/VCAM-1 and plasma cytokines IL-5/IL-13 were reversed; cell count, NOx and MMP-9-activity in BALF were decreased by KMUP-1. CONCLUSIONS: Inhaled KMUP-1, preventing allergic pulmonary vascular inflammation and remodeling, would be useful for the treatment of asthma and respiratory obstruction disease.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Hipersensibilidad/tratamiento farmacológico , Metaloproteinasa 9 de la Matriz/metabolismo , Óxido Nítrico/metabolismo , Piperidinas/administración & dosificación , Neumonía/tratamiento farmacológico , Xantinas/administración & dosificación , Animales , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Femenino , Guanilato Ciclasa/metabolismo , Hipersensibilidad/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Endogámicos BALB C , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Neumonía/metabolismo , Neumonía/prevención & control , Receptores Citoplasmáticos y Nucleares/metabolismo , Guanilil Ciclasa Soluble , Molécula 1 de Adhesión Celular Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...