Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(16)2021 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-34445556

RESUMEN

Neutrophil extracellular traps (NETs), built from mitochondrial or nuclear DNA, proteinases, and histones, entrap and eliminate pathogens in the course of bacterial or viral infections. Neutrophils' activation and the formation of NETs have been described as major risk factors for acute lung injury, multi-organ damage, and mortality in COVID-19 disease. NETs-related lung injury involves both epithelial and endothelial cells, as well as the alveolar-capillary barrier. The markers for NETs formation, such as circulating DNA, neutrophil elastase (NE) activity, or myeloperoxidase-DNA complexes, were found in lung specimens of COVID-19 victims, as well as in sera and tracheal aspirates obtained from COVID-19 patients. DNA threads form large conglomerates causing local obstruction of the small bronchi and together with NE are responsible for overproduction of mucin by epithelial cells. Various components of NETs are involved in the pathogenesis of cytokine storm in SARS-CoV-2 pulmonary disease. NETs are responsible for the interplay between inflammation and thrombosis in the affected lungs. The immunothrombosis, stimulated by NETs, has a poor prognostic significance. Better understanding of the role of NETs in the course of COVID-19 can help to develop novel approaches to the therapeutic interventions in this condition.


Asunto(s)
COVID-19/inmunología , Trampas Extracelulares/virología , Pulmón/inmunología , Neutrófilos/inmunología , SARS-CoV-2/inmunología , COVID-19/patología , COVID-19/virología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/virología , Células Endoteliales/patología , Células Epiteliales/patología , Trampas Extracelulares/inmunología , Histonas/inmunología , Humanos , Elastasa de Leucocito/deficiencia , Elastasa de Leucocito/inmunología , Pulmón/patología , Pulmón/virología , Activación Neutrófila , Neutrófilos/virología , Peroxidasa/inmunología
2.
Front Immunol ; 12: 653932, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33968054

RESUMEN

Severe congenital neutropenia (SCN) is a rare hematological condition with heterogenous genetic background. Neutrophil elastase (NE) encoded by ELANE gene is mutated in over half of the SCN cases. The role of NE defects in myelocytes maturation arrest in bone marrow is widely investigated; however, the mechanism underlying this phenomenon has still remained unclear. In this review, we sum up the studies exploring mechanisms of neutrophil deficiency, biological role of NE in neutrophil and the effects of ELANE mutation and neutropenia pathogenesis. We also explain the hypotheses presented so far and summarize options of neutropenia therapy.


Asunto(s)
Síndromes Congénitos de Insuficiencia de la Médula Ósea/diagnóstico , Síndromes Congénitos de Insuficiencia de la Médula Ósea/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Elastasa de Leucocito/deficiencia , Neutropenia/congénito , Neutrófilos/enzimología , Regulación de la Expresión Génica , Humanos , Elastasa de Leucocito/química , Elastasa de Leucocito/genética , Elastasa de Leucocito/metabolismo , Mutación , Neutropenia/diagnóstico , Neutropenia/genética , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Transducción de Señal , Relación Estructura-Actividad
3.
Int J Mol Sci ; 22(2)2021 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-33450865

RESUMEN

Neutrophils are recruited into the heart at an early stage following a myocardial infarction (MI). These secrete several proteases, one of them being neutrophil elastase (NE), which promotes inflammatory responses in several disease models. It has been shown that there is an increase in NE activity in patients with MI; however, the role of NE in MI remains unclear. Therefore, the present study aimed to investigate the role of NE in the pathogenesis of MI in mice. NE expression peaked on day 1 in the infarcted hearts. In addition, NE deficiency improved survival and cardiac function post-MI, limiting fibrosis in the noninfarcted myocardium. Sivelestat, an NE inhibitor, also improved survival and cardiac function post-MI. Flow cytometric analysis showed that the numbers of heart-infiltrating neutrophils and inflammatory macrophages (CD11b+F4/80+CD206low cells) were significantly lower in NE-deficient mice than in wild-type (WT) mice. At the border zone between intact and necrotic areas, the number of terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive apoptotic cells was lower in NE-deficient mice than in WT mice. Western blot analyses revealed that the expression levels of insulin receptor substrate 1 and phosphorylation of Akt were significantly upregulated in NE-knockout mouse hearts, indicating that NE deficiency might improve cardiac survival by upregulating insulin/Akt signaling post-MI. Thus, NE may enhance myocardial injury by inducing an excessive inflammatory response and suppressing Akt signaling in cardiomyocytes. Inhibition of NE might serve as a novel therapeutic target in the treatment of MI.


Asunto(s)
Elastasa de Leucocito/deficiencia , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Neutrófilos/enzimología , Animales , Apoptosis/genética , Biomarcadores , Biopsia , Modelos Animales de Enfermedad , Pruebas de Función Cardíaca , Insulinas/metabolismo , Elastasa de Leucocito/metabolismo , Ratones , Ratones Noqueados , Infarto del Miocardio/mortalidad , Infarto del Miocardio/patología , Miocardio/patología , Miocitos Cardíacos/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Remodelación Ventricular/genética
5.
Inflammation ; 43(3): 1054-1064, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32002713

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease. Inflammatory pathways contribute to disease pathogenesis; however, regulation of the underlying mechanism is not completely understood. IL-1ß, a pro-inflammatory cytokine, participates in the development and progression of NAFLD. To become bioactive, IL-1ß requires enzymatic processing. Mechanisms that activate IL-1ß include the classical NLRP3 inflammasome-caspase-1 and the neutrophil serine proteases, neutrophil elastase, and proteinase-3. Several studies have shown that both caspase-1 and the neutrophil serine proteases are important for NAFLD development. However, it is unknown whether these pathways interact and if they have a synergistic effect in promoting NAFLD. In the present study, we developed a novel and unique mouse model by intercrossing caspase-1/11 knockout mice with neutrophil elastase/proteinase-3 double knockout mice. Subsequently, these mice were examined regarding the development of high-fat diet-induced NAFLD. Our results show that mice deficient in caspase-1, neutrophil elastase, and proteinase-3 were protected from developing diet-induced weigh gain, liver steatosis, and adipose tissue inflammation when compared with controls. We conclude that pathways that process pro-IL-1ß to bioactive IL-1ß play an important role in promoting the development of NAFLD and obesity-induced inflammation. Targeting these pathways could have a therapeutic potential in patients with NAFLD.


Asunto(s)
Caspasa 1/deficiencia , Interleucina-1beta/antagonistas & inhibidores , Elastasa de Leucocito/deficiencia , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo , Serina Endopeptidasas/deficiencia , Animales , Caspasa 1/genética , Dieta Alta en Grasa/efectos adversos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Elastasa de Leucocito/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Obesidad/genética , Obesidad/prevención & control , Serina Endopeptidasas/genética
6.
mSphere ; 4(5)2019 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-31484737

RESUMEN

Disseminated candidiasis is a life-threatening disease and remains the most common bloodstream infection in hospitalized patients in the United States. Despite the availability of modern antifungal therapy, crude mortality in the last decade has remained unacceptably high. In particular, Candida auris is a multidrug-resistant, health care-associated fungal pathogen and has recently emerged as the first fungal pathogen to cause a global public health threat. A reliable animal model for disseminated C. auris candidiasis is therefore needed to study the unique aspects of this little-known host-pathogen interaction. In this study, we established an inbred A/J intravenous model as an appropriate model for human disseminated C. auris infection. We found that C5 deficiency in A/J mice results in a complex phenotype characterized by rapid fungal proliferation in target organs and the development of a unique and rapidly fatal response. In contrast, C57BL/6J mice and mice deficient in neutrophil elastase (NE-/-) survived high-dose C. auris intravenous challenge, even with cyclophosphamide (CY)-induced immunosuppression. Our study is the first to provide insight into the role of C5 in the host responses to C. auris invasive infection and establishes an inbred A/J mouse model of systemic C. auris infection without CY-induced immunosuppression.IMPORTANCE In the last decade, Candida auris has emerged globally as a multidrug-resistant fungal pathogen. Although C. auris was initially isolated from the external ear canal, it can cause outbreaks of invasive infections with very high mortality and comorbidities. Recent reports highlight the ongoing challenges due to organism misidentification, high rates of multifungal drug resistance, and unacceptably high patient mortality. The assessment of C. auris virulence in a specific genetic deficiency mouse model of invasive C. auris infection in this study contributes to the little knowledge of host defense to C. auris infection, which holds promise as a model for investigating the pathogenesis of C. auris invasive infection, exploring the immune responses elicited by the fungus, evaluating the possible induction of immunity to the infection, and targeting candidates for an antifungal vaccine.


Asunto(s)
Candida/patogenicidad , Candidiasis/microbiología , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Animales , Candida/inmunología , Candidiasis/inmunología , Complemento C5/deficiencia , Humanos , Elastasa de Leucocito/deficiencia , Ratones , Ratones Endogámicos A , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Datos Preliminares , Virulencia
7.
FASEB J ; 33(10): 10794-10807, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31284755

RESUMEN

Visceral leishmaniasis is a deadly illness caused by Leishmania donovani that provokes liver and spleen inflammation and tissue destruction. In cutaneous leishmaniasis, the protein of L. major, named inhibitor of serine peptidases (ISP) 2, inactivates neutrophil elastase (NE) present at the macrophage surface, resulting in blockade of TLR4 activation, prevention of TNF-α and IFN-ß production, and parasite survival. We report poor intracellular growth of L. donovani in macrophages from knockout mice for NE (ela-/-), TLR4, or TLR2. NE and TLR4 colocalized with the parasite in the parasitophorous vacuole. Parasite load in the liver and spleen of ela-/- mice were reduced and accompanied by increased NO and decreased TGF-ß production. Expression of ISP2 was not detected in L. donovani, and a transgenic line constitutively expressing ISP2, displayed poor intracellular growth in macrophages and decreased burden in mice. Infected ela-/- macrophages displayed significantly lower IFN-ß mRNA than background mice macrophages, and the intracellular growth was fully restored by exogenous IFN-ß. We propose that L. donovani utilizes the host NE-TLR machinery to induce IFN-ß necessary for parasite survival and growth during early infection. Low or absent expression of parasite ISP2 in L. donovani is necessary to preserve the activation of the NE-TLR pathway.-Dias, B. T., Dias-Teixeira, K. L., Godinho, J. P., Faria, M. S., Calegari-Silva, T., Mukhtar, M. M., Lopes, U. G., Mottram, J. C., Lima, A. P. C. A. Neutrophil elastase promotes Leishmania donovani infection via interferon-ß.


Asunto(s)
Interferón beta/metabolismo , Leishmania donovani/patogenicidad , Leishmaniasis Visceral/etiología , Elastasa de Leucocito/metabolismo , Animales , Animales Modificados Genéticamente , Leishmania donovani/genética , Leishmania donovani/fisiología , Leishmaniasis Visceral/metabolismo , Leishmaniasis Visceral/parasitología , Elastasa de Leucocito/deficiencia , Elastasa de Leucocito/genética , Macrófagos/metabolismo , Macrófagos/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Protozoarias/genética , Proteínas Protozoarias/fisiología , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
8.
J Pathol ; 248(1): 88-102, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30632166

RESUMEN

Ischemia/reperfusion (I/R) injury is a severe inflammatory insult associated with numerous pathologies, such as myocardial infarction, stroke and acute kidney injury. I/R injury is characterized by a rapid influx of activated neutrophils secreting toxic free radical species and degrading enzymes that can irreversibly damage the tissue, thus impairing organ functions. Significant efforts have been invested in identifying therapeutic targets to suppress neutrophil recruitment and activation post-I/R injury. In this context, pharmacological targeting of neutrophil elastase (NE) has shown promising anti-inflammatory efficacy in a number of experimental and clinical settings of I/R injury and is considered a plausible clinical strategy for organ care. However, the mechanisms of action of NE, and hence its inhibitors, in this process are not fully understood. Here we conducted a comprehensive analysis of the impact of NE genetic deletion on neutrophil infiltration in four murine models of I/R injury as induced in the heart, kidneys, intestine and cremaster muscle. In all models, neutrophil migration into ischemic regions was significantly suppressed in NE-/- mice as compared with wild-type controls. Analysis of inflamed cremaster muscle and mesenteric microvessels by intravital and confocal microscopy revealed a selective entrapment of neutrophils within venular walls, most notably at the level of the venular basement membrane (BM) following NE deletion/pharmacological blockade. This effect was associated with the suppression of NE-mediated remodeling of the low matrix protein expressing regions within the venular BM used by transmigrating neutrophils as exit portals. Furthermore, whilst NE deficiency led to reduced neutrophil activation and vascular leakage, levels of monocytes and prohealing M2 macrophages were reduced in tissues of NE-/- mice subjected to I/R. Collectively our results identify a vital and non-redundant role for NE in supporting neutrophil breaching of the venular BM post-I/R injury but also suggest a protective role for NE in promoting tissue repair. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Elastasa de Leucocito/fisiología , Neutrófilos/fisiología , Daño por Reperfusión/enzimología , Migración Transendotelial y Transepitelial/fisiología , Remodelación Vascular/fisiología , Animales , Membrana Basal/enzimología , Membrana Basal/patología , Membrana Basal/fisiopatología , Modelos Animales de Enfermedad , Eliminación de Gen , Riñón/irrigación sanguínea , Riñón/patología , Elastasa de Leucocito/deficiencia , Elastasa de Leucocito/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Infiltración Neutrófila/fisiología , Neutrófilos/enzimología , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología , Vénulas/enzimología , Vénulas/patología , Vénulas/fisiopatología
9.
J Am Heart Assoc ; 7(4)2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29437605

RESUMEN

BACKGROUND: To investigate whether neutrophil elastase (NE) plays a causal role in atherosclerosis, and the molecular mechanisms involved. METHODS AND RESULTS: NE genetic-deficient mice (Apolipoprotein E-/-/NE-/- mice), bone marrow transplantation, and a specific NE inhibitor (GW311616A) were employed in this study to establish the causal role of NE in atherosclerosis. Aortic expression of NE mRNA and plasma NE activity was significantly increased in high-fat diet (HFD)-fed wild-type (WT) (Apolipoprotein E-/-) mice but, as expected, not in NE-deficient mice. Selective NE knockout markedly reduced HFD-induced atherosclerosis and significantly increased indicators of atherosclerotic plaque stability. While plasma lipid profiles were not affected by NE deficiency, decreased levels of circulating proinflammatory cytokines and inflammatory monocytes (Ly6Chi/CD11b+) were observed in NE-deficient mice fed with an HFD for 12 weeks as compared with WT. Bone marrow reconstitution of WT mice with NE-/- bone marrow cells significantly reduced HFD-induced atherosclerosis, while bone marrow reconstitution of NE-/- mice with WT bone marrow cells restored the pathological features of atherosclerotic plaques induced by HFD in NE-deficient mice. In line with these findings, pharmacological inhibition of NE in WT mice through oral administration of NE inhibitor GW311616A also significantly reduced atherosclerosis. Mechanistically, we demonstrated that NE promotes foam cell formation by increasing ATP-binding cassette transporter ABCA1 protein degradation and inhibiting macrophage cholesterol efflux. CONCLUSIONS: We outlined a pathogenic role for NE in foam cell formation and atherosclerosis development. Consequently, inhibition of NE may represent a potential therapeutic approach to treating cardiovascular disease.


Asunto(s)
Aorta/efectos de los fármacos , Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Elastasa de Leucocito/antagonistas & inhibidores , Elastasa de Leucocito/deficiencia , Neutrófilos/efectos de los fármacos , Piperidinas/farmacología , Inhibidores de Serina Proteinasa/farmacología , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Aorta/enzimología , Aorta/patología , Enfermedades de la Aorta/enzimología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/enzimología , Aterosclerosis/genética , Aterosclerosis/patología , Células Cultivadas , Colesterol/sangre , Citocinas/sangre , Modelos Animales de Enfermedad , Células Espumosas/efectos de los fármacos , Células Espumosas/metabolismo , Células Espumosas/patología , Mediadores de Inflamación/sangre , Elastasa de Leucocito/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Neutrófilos/enzimología , Placa Aterosclerótica , Proteolisis
10.
PLoS One ; 11(12): e0168055, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27942017

RESUMEN

Severe congenital neutropenia (SCN) is characterised by a differentiation block in the bone marrow and low neutrophil numbers in the peripheral blood, which correlates with increased risk of bacterial infections. Several underlying gene defects have been identified in SCN patients. Mutations in the neutrophil elastase (ELANE) gene are frequently found in SCN and cyclic neutropenia. Both mislocalization and misfolding of mutant neutrophil elastase protein resulting in ER stress and subsequent induction of the unfolded protein response (UPR) have been proposed to be responsible for neutrophil survival and maturation defects. However, the detailed molecular mechanisms still remain unclear, in part due to the lack of appropriate in vitro and in vivo models. Here we used a system of neutrophil differentiation from immortalised progenitor lines by conditional expression of Hoxb8, permitting the generation of mature near-primary neutrophils in vitro and in vivo. NE-deficient Hoxb8 progenitors were reconstituted with murine and human forms of typical NE mutants representative of SCN and cyclic neutropenia, and differentiation of the cells was analysed in vitro and in vivo. ER stress induction by NE mutations could be recapitulated during neutrophil differentiation in all NE mutant-reconstituted Hoxb8 cells. Despite ER stress induction, no change in survival, maturation or function of differentiating cells expressing either murine or human NE mutants was observed. Further analysis of in vivo differentiation of Hoxb8 cells in a murine model of adoptive transfer did not reveal any defects in survival or differentiation in the mouse. Although the Hoxb8 system has been found to be useful for dissection of defects in neutrophil development, our findings indicate that the use of murine systems for analysis of NE-mutation-associated pathogenesis is complicated by differences between humans and mice in the physiology of granulopoiesis, which may go beyond possible differences in expression and activity of neutrophil elastase itself.


Asunto(s)
Elastasa de Leucocito/genética , Leucopoyesis , Neutropenia/genética , Neutrófilos/citología , Animales , Supervivencia Celular , Modelos Animales de Enfermedad , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Elastasa de Leucocito/deficiencia , Ratones , Ratones Endogámicos C57BL , Mutación , Neutropenia/enzimología , Neutrófilos/enzimología , Especificidad de la Especie
11.
Respir Res ; 17(1): 129, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27751187

RESUMEN

BACKGROUND: In acutely injured lungs, massively recruited polymorphonuclear neutrophils (PMNs) secrete abnormally neutrophil elastase (NE). Active NE creates a localized proteolytic environment where various host molecules are degraded leading to impairment of tissue homeostasis. Among the hallmarks of neutrophil-rich pathologies is a disrupted epithelium characterized by the loss of cell-cell adhesion and integrity. Epithelial-cadherin (E-cad) represents one of the most important intercellular junction proteins. E-cad exhibits various functions including its role in maintenance of tissue integrity. While much interest has focused on the expression and role of E-cad in different physio- and physiopathological states, proteolytic degradation of this structural molecule and ensuing potential consequences on host lung tissue injury are not completely understood. METHODS: NE capacity to cleave E-cad was determined in cell-free and lung epithelial cell culture systems. The impact of such cleavage on epithelial monolayer integrity was then investigated. Using mice deficient in NE in a clinically relevant experimental model of acute pneumonia, we examined whether degraded E-cad is associated with lung inflammation and injury and whether NE contributes to E-cad cleavage. Finally, we checked for the presence of both degraded E-cad and NE in bronchoalveolar lavage samples obtained from patients with exacerbated COPD, a clinical manifestation characterised by a neutrophilic inflammatory response. RESULTS: We show that NE is capable of degrading E-cad in vitro and in cultured cells. NE-mediated degradation of E-cad was accompanied with loss of epithelial monolayer integrity. Our in vivo findings provide evidence that NE contributes to E-cad cleavage that is concomitant with lung inflammation and injury. Importantly, we observed that the presence of degraded E-cad coincided with the detection of NE in diseased human lungs. CONCLUSIONS: Active NE has the capacity to cleave E-cad and interfere with its cell-cell adhesion function. These data suggest a mechanism by which unchecked NE participates potentially to the pathogenesis of neutrophil-rich lung inflammatory and tissue-destructive diseases.


Asunto(s)
Lesión Pulmonar Aguda/enzimología , Cadherinas/metabolismo , Células Epiteliales/enzimología , Elastasa de Leucocito/metabolismo , Pulmón/enzimología , Neutrófilos/enzimología , Neumonía Bacteriana/enzimología , Enfermedad Pulmonar Obstructiva Crónica/enzimología , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Antígenos CD , Líquido del Lavado Bronquioalveolar/química , Línea Celular , Modelos Animales de Enfermedad , Células Epiteliales/patología , Elastasa de Leucocito/deficiencia , Elastasa de Leucocito/genética , Pulmón/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía Bacteriana/genética , Neumonía Bacteriana/patología , Proteolisis
12.
Arterioscler Thromb Vasc Biol ; 36(8): 1660-1669, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27283739

RESUMEN

OBJECTIVE: We previously established that neutrophil-derived dipeptidyl peptidase I (DPPI) is essential for experimental abdominal aortic aneurysm (AAA) development. Because DPPI activates several neutrophil serine proteases, it remains to be determined whether the AAA-promoting effect of DPPI is mediated by neutrophil serine proteases. APPROACH AND RESULTS: Using an elastase-induced AAA model, we demonstrate that the absence of 2 neutrophil serine proteases, neutrophil elastase and proteinase-3, recapitulates the AAA-resistant phenotype of DPPI-deficient mice. DPPI and neutrophil serine proteases direct the in vitro and in vivo release of extracellular structures termed neutrophil extracellular traps (NETs). Administration of DNase1, which dismantles NETs, suppresses elastase-induced AAA in wild-type animals and in DPPI-deficient mice reconstituted with wild-type neutrophils. NETs also contain the cathelicidin-related antimicrobial peptide that complexes with self-DNA in recruiting plasmacytoid dendritic cells (pDCs), inducing type I interferons (IFNs) and promoting AAA in DPPI-deficient mice. Conversely, depletion of pDCs or blockade of type I IFNs suppresses experimental AAA. Moreover, we find an abundance of human cathelicidin peptide, a 37 amino acid sequence starting with 2 leucines and the human orthologue of cathelicidin-related antimicrobial peptide, in the vicinity of pDCs in human AAA tissues. Increased type I IFN mRNA expression is observed in human AAA tissues and circulating IFN-α is detected in ≈50% of the AAA sera examined. CONCLUSIONS: These results suggest that neutrophil protease-mediated NET release contributes to elastase-induced AAA through pDC activation and type I IFN production. These findings increase our understanding of the pathways underlying AAA inflammatory responses and suggest that limiting NET, pDC, and type I IFN activities may suppress aneurysm progression.


Asunto(s)
Aorta Abdominal/enzimología , Aneurisma de la Aorta Abdominal/enzimología , Catepsina C/metabolismo , Células Dendríticas/enzimología , Trampas Extracelulares/enzimología , Elastasa de Leucocito/metabolismo , Mieloblastina/metabolismo , Neutrófilos/enzimología , Animales , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/prevención & control , Catepsina C/genética , Células Cultivadas , Modelos Animales de Enfermedad , Genotipo , Humanos , Interferón Tipo I/metabolismo , Elastasa de Leucocito/deficiencia , Elastasa de Leucocito/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mieloblastina/deficiencia , Mieloblastina/genética , Neutrófilos/patología , Fenotipo , Transducción de Señal
13.
J Thromb Haemost ; 14(3): 551-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26712312

RESUMEN

UNLABELLED: ESSENTIALS: Neutrophil elastase (NE) plays a role in extracellular trap formation (NETosis) triggered by microbes. The contribution of NE was evaluated in mouse NETosis models of sterile inflammation and thrombosis. NE is not required for mouse neutrophil NET production in vitro with non-infectious stimuli. NE deficiency had no significant effect on thrombosis in the inferior vena cava stenosis model. BACKGROUND: Neutrophil serine proteases have been implicated in coagulation and neutrophil extracellular trap (NET) formation. In human neutrophils, neutrophil elastase (NE) translocates to the nucleus during NETosis and cleaves histones, thus aiding in chromatin decondensation. NE(-/-) mice were shown not to release NETs in response to microbes. However, mouse studies evaluating the role of NE in NET formation in sterile inflammation and thrombosis are lacking. OBJECTIVE: We wished to establish if neutrophils from NE(-/-) mice have a defect in NETosis, similar to peptidylarginine deiminase 4 (PAD4(-/-)) mice, and how this might have an impact on venous thrombosis, a model where NETs are produced and are crucial to thrombus development. METHODS: We performed in vitro NET assays using neutrophils from wild-type (WT), NE(-/-), SerpinB1 (SB1)(-/-) and NE(-/-) SB1(-/-) mice. We compared WT and NE(-/-) animals using the inferior vena cava stenosis model of deep vein thrombosis (DVT). RESULTS: Neutrophil elastase deficiency resulted in a small reduction in ionomycin-induced NET formation in vitro without affecting histone citrullination. However, NET production in response to phorbol 12-myristate 13-acetate or platelet activating factor was normal in neutrophils from two independent NE-deficient mouse lines, and in NE(-/-) SB1(-/-) as compared with SB1(-/-) neutrophils. NE deficiency or inhibition did not prevent NETosis in vivo or DVT outcome. CONCLUSIONS: Neutrophil elastase is not required for NET formation in mice. NE(-/-) mice, which form pathological venous thrombi containing NETs, do not phenocopy PAD4(-/-) mice in in vitro NETosis assays or experimental venous thrombosis. Our study suggests that NET-targeted therapies need to be highly effective to have an impact on DVT.


Asunto(s)
Trampas Extracelulares/metabolismo , Elastasa de Leucocito/deficiencia , Neutrófilos/enzimología , Trombosis de la Vena/enzimología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Genotipo , Ionomicina/farmacología , Elastasa de Leucocito/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Activación Neutrófila , Neutrófilos/efectos de los fármacos , Fenotipo , Acetato de Tetradecanoilforbol/farmacología , Trombosis de la Vena/sangre , Trombosis de la Vena/genética
15.
Proc Natl Acad Sci U S A ; 110(37): 15007-12, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-23975926

RESUMEN

The rationale of α1-antitrypsin (AAT) augmentation therapy to treat progressive emphysema in AAT-deficient patients is based on inhibition of neutrophil elastase; however, the benefit of this treatment remains unclear. Here we show that clinical grade AAT (with elastase inhibitory activity) and a recombinant form of AAT (rAAT) without anti-elastase activity reduces lung inflammatory responses to LPS in elastase-deficient mice. WT and elastase-deficient mice treated with either native AAT or rAAT exhibited significant reductions in infiltrating neutrophils (23% and 68%), lavage fluid levels of TNF-α (70% and 80%), and the neutrophil chemokine KC (CXCL1) (64% and 90%), respectively. Lung parenchyma TNF-α, DNA damage-inducible transcript 3 and X-box binding protein-1 mRNA levels were reduced in both mouse strains treated with AAT; significantly lower levels of these genes, as well as IL-1ß gene expression, were observed in lungs of AAT-deficient patients treated with AAT therapy compared with untreated patients. In vitro, LPS-induced cytokines from WT and elastase-deficient mouse neutrophils, as well as neutrophils of healthy humans, were similarly reduced by AAT or rAAT; human neutrophils adhering to endothelial cells were decreased by 60-80% (P < 0.001) with either AAT or rAAT. In mouse pancreatic islet macrophages, LPS-induced surface expression of MHC II, Toll-like receptor-2 and -4 were markedly lower (80%, P < 0.001) when exposed to either AAT or rAAT. Consistently, in vivo and in vitro, rAAT reduced inflammatory responses at concentrations 40- to 100-fold lower than native plasma-derived AAT. These data provide evidence that the anti-inflammatory and immunomodulatory properties of AAT can be independent of elastase inhibition.


Asunto(s)
Antiinflamatorios/farmacología , Factores Inmunológicos/farmacología , Elastasa de Leucocito/metabolismo , Enfisema Pulmonar/tratamiento farmacológico , alfa 1-Antitripsina/farmacología , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/inmunología , Adulto , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Elastasa de Leucocito/antagonistas & inhibidores , Elastasa de Leucocito/deficiencia , Lipopolisacáridos/toxicidad , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Enfisema Pulmonar/genética , Enfisema Pulmonar/inmunología , Proteínas Recombinantes/farmacología , Deficiencia de alfa 1-Antitripsina/tratamiento farmacológico , Deficiencia de alfa 1-Antitripsina/genética , Deficiencia de alfa 1-Antitripsina/inmunología
16.
Pediatr Endocrinol Rev ; 11 Suppl 1: 59-63, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24380123

RESUMEN

Lysosomal storage diseases (LSDs), which are inborn errors of metabolism, encompass around 50 different inherited syndromes. Together, they have an incidence of 1/7000 newborns. LSDs are caused by deficiencies in lysosomal enzymes or transporters, resulting in intra-lysosomal buildup of under graded metabolites. Common features of LSDs include bone disease, organomegaly and central and peripheral nervous system degeneration. These diseases were first described in the 1880s. Despite more than an hundred years of study of the genetic and molecular bases of LSDs, little is known about the events that lead from intra-lysosomal accumulation to the distinctive cell dysfunction and pathology that is characteristic of each disease. This review focuses on the main historical discoveries in LSD biology, from the original descriptions of their phenotypes, to animal models, including therapeutic strategies and challenges to treat this family of devastating diseases.


Asunto(s)
Predisposición Genética a la Enfermedad , Terapia Genética/métodos , Elastasa de Leucocito/deficiencia , Enfermedades por Almacenamiento Lisosomal , Animales , Salud Global , Humanos , Incidencia , Enfermedades por Almacenamiento Lisosomal/epidemiología , Enfermedades por Almacenamiento Lisosomal/genética , Enfermedades por Almacenamiento Lisosomal/terapia , Fenotipo
17.
Arterioscler Thromb Vasc Biol ; 32(12): 2901-9, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23042818

RESUMEN

OBJECTIVE: The purpose of this study was to evaluate potential mechanisms promoting abdominal aortic aneurysm development with tobacco smoke (TS) exposure. METHODS AND RESULTS: Experiments used the elastase perfusion model of abdominal aortic aneurysms with smoke-free controls. The effect of TS exposure was evaluated in C57/Bl6 mice, after broad-spectrum matrix metalloproteinase inhibition with doxycycline and in mice deficient in matrix metalloproteinase-9, matrix metalloproteinase-12, Cathepsin-S, and Neutrophil Elastase. Preparations of washed marrow, spleen, and peripheral blood leukocytes were transferred to smoke-free mice from 6-week TS-exposed mice or smoke-free mice. All mice were euthanized 14 days after elastase perfusion, and the percentage of change in aortic diameter (%Δ aortic diameter) was calculated. Electron microscopy of aortic tissue from animals exposed to TS without elastase exposure did not demonstrate any ultrastructural changes. Neither doxycycline nor any specific elastase deficiency was effective at preventing an increase in %Δ aortic diameter in TS-exposed animals. Smoke exposure for 6 weeks increased the %Δ aortic diameter after a smoke-free interval of up to 6 weeks before elastase perfusion. Leukocyte preparations from TS-exposed mice localized to abdominal aortic aneurysms and increased the %Δ aortic diameter in smoke-free mice. CONCLUSIONS: The effect of TS on the development of abdominal aortic aneurysms is not dependent on the activity of elastolytic enzymes and persists for long periods despite cessation of TS. Alterations in leukocyte response to aortic injury appear to mediate this effect.


Asunto(s)
Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/fisiopatología , Leucocitos Mononucleares/patología , Leucocitos Mononucleares/fisiología , Fumar/efectos adversos , Animales , Aorta Abdominal/patología , Aorta Abdominal/fisiopatología , Aorta Abdominal/ultraestructura , Aneurisma de la Aorta Abdominal/patología , Catepsinas/deficiencia , Catepsinas/genética , Catepsinas/fisiología , Recuento de Células , Modelos Animales de Enfermedad , Doxiciclina/farmacología , Elastasa de Leucocito/deficiencia , Elastasa de Leucocito/genética , Elastasa de Leucocito/fisiología , Masculino , Metaloproteinasa 12 de la Matriz/deficiencia , Metaloproteinasa 12 de la Matriz/genética , Metaloproteinasa 12 de la Matriz/fisiología , Metaloproteinasa 9 de la Matriz/deficiencia , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/fisiología , Metaloproteinasas de la Matriz/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
18.
J Cell Biol ; 191(3): 677-91, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20974816

RESUMEN

Neutrophils release decondensed chromatin termed neutrophil extracellular traps (NETs) to trap and kill pathogens extracellularly. Reactive oxygen species are required to initiate NET formation but the downstream molecular mechanism is unknown. We show that upon activation, neutrophil elastase (NE) escapes from azurophilic granules and translocates to the nucleus, where it partially degrades specific histones, promoting chromatin decondensation. Subsequently, myeloperoxidase synergizes with NE in driving chromatin decondensation independent of its enzymatic activity. Accordingly, NE knockout mice do not form NETs in a pulmonary model of Klebsiella pneumoniae infection, which suggests that this defect may contribute to the immune deficiency of these mice. This mechanism provides for a novel function for serine proteases and highly charged granular proteins in the regulation of chromatin density, and reveals that the oxidative burst induces a selective release of granular proteins into the cytoplasm through an unknown mechanism.


Asunto(s)
Espacio Extracelular/metabolismo , Elastasa de Leucocito/metabolismo , Neutrófilos/citología , Neutrófilos/enzimología , Peroxidasa/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae , Elastasa de Leucocito/deficiencia , Ratones , Ratones Noqueados , Neutrófilos/inmunología , Valores de Referencia
19.
Respir Physiol Neurobiol ; 173(2): 164-70, 2010 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-20696285

RESUMEN

Previous studies have suggested that in vitro modulation of neutrophil chemokines and inflammatory cytokines by neutrophil elastase (NE) does not translate to the in vivo setting. We aimed to test the role of NE in the recruitment of neutrophils, cytokine production and lung function responses to respiratory viral infection. To address this, we inoculated neutrophil elastase (NE(-/-)) deficient and wild-type (WT) 129Sv mice with 50µL of 10(4.5)pfu Influenza A/Mem71 (H3N1) or a control preparation. Mice were subjected to methacholine (MCh) challenge at 3-4 days post-infection during the peak of cellular inflammation. Inflammation, protein content and cytokines (TNF-α and MIP-2) were assessed in bronchoalveolar lavage fluid. Influenza-infected mice had a heightened responsiveness to MCh, increased cellular inflammation, increased protein leak and altered cytokine production, none of which were influenced by the absence of NE. These data demonstrate that NE does not modulate neutrophil recruitment, cytokine production, epithelial permeability or responsiveness to bronchoconstricting agents during acute influenza infection in mice.


Asunto(s)
Hiperreactividad Bronquial/etiología , Hiperreactividad Bronquial/inmunología , Citocinas/metabolismo , Elastasa de Leucocito/metabolismo , Pulmón/fisiopatología , Infecciones por Orthomyxoviridae/complicaciones , Resistencia de las Vías Respiratorias/efectos de los fármacos , Resistencia de las Vías Respiratorias/genética , Animales , Hiperreactividad Bronquial/genética , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Broncoconstrictores/administración & dosificación , Quimiocina CXCL2/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Elastasa de Leucocito/deficiencia , Pulmón/efectos de los fármacos , Pulmón/virología , Cloruro de Metacolina/administración & dosificación , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Estadísticas no Paramétricas , Factor de Necrosis Tumoral alfa/metabolismo
20.
Artículo en Inglés | MEDLINE | ID: mdl-20008220

RESUMEN

Congenital neutropenia comprises a variety of genetically heterogeneous phenotypic traits. Molecular elucidation of the underlying genetic defects has yielded important insights into the physiology of neutrophil differentiation and function. Non-syndromic variants of congenital neutropenia are caused by mutations in ELA2, HAX1, GFI1, or WAS. Syndromic variants of congenital neutropenia may be due to mutations in genes controlling glucose metabolism (SLC37A4, G6PC3) or lysosomal function (LYST, RAB27A, ROBLD3/p14, AP3B1, VPS13B). Furthermore, defects in genes encoding ribosomal proteins (SBDS, RMRP) and mitochondrial proteins (AK2, TAZ) are associated with congenital neutropenia syndromes. Despite remarkable progress in the field, many patients with congenital neutropenia cannot yet definitively be classified by genetic terms. This review addresses diagnostic and therapeutic aspects of congenital neutropenia and covers recent molecular and pathophysiological insights of selected congenital neutropenia syndromes.


Asunto(s)
Neutropenia/congénito , Proteínas Adaptadoras Transductoras de Señales , Adenilato Quinasa/deficiencia , Adenilato Quinasa/genética , Metabolismo de los Hidratos de Carbono/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Heterogeneidad Genética , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Humanos , Elastasa de Leucocito/deficiencia , Elastasa de Leucocito/genética , Lisosomas , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Neutropenia/tratamiento farmacológico , Neutropenia/genética , Neutropenia/fisiopatología , Neutropenia/cirugía , Proteínas/genética , Proteínas Ribosómicas/deficiencia , Proteínas Ribosómicas/genética , Síndrome , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Trasplante Homólogo , Proteína del Síndrome de Wiskott-Aldrich/deficiencia , Proteína del Síndrome de Wiskott-Aldrich/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...