Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 23(6): 927-939, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35624205

RESUMEN

Hypoxemia is a defining feature of acute respiratory distress syndrome (ARDS), an often-fatal complication of pulmonary or systemic inflammation, yet the resulting tissue hypoxia, and its impact on immune responses, is often neglected. In the present study, we have shown that ARDS patients were hypoxemic and monocytopenic within the first 48 h of ventilation. Monocytopenia was also observed in mouse models of hypoxic acute lung injury, in which hypoxemia drove the suppression of type I interferon signaling in the bone marrow. This impaired monopoiesis resulted in reduced accumulation of monocyte-derived macrophages and enhanced neutrophil-mediated inflammation in the lung. Administration of colony-stimulating factor 1 in mice with hypoxic lung injury rescued the monocytopenia, altered the phenotype of circulating monocytes, increased monocyte-derived macrophages in the lung and limited injury. Thus, tissue hypoxia altered the dynamics of the immune response to the detriment of the host and interventions to address the aberrant response offer new therapeutic strategies for ARDS.


Asunto(s)
Lesión Pulmonar , Síndrome de Dificultad Respiratoria , Animales , Humanos , Hipoxia/etiología , Inflamación/complicaciones , Pulmón , Lesión Pulmonar/complicaciones , Ratones
3.
Immunity ; 48(5): 846-848, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29768171

RESUMEN

Neutrophil recruitment in response to pathogen invasion is mediated through "self" tissue damage signals (DAMPs) and pathogen associated signals (PAMPs). In this issue of Immunity, Huang and Niethammer, (2018) demonstrate that DAMP signaling is a prerequisite for neutrophil recruitment.


Asunto(s)
Inmunidad Innata , Pez Cebra , Animales , Infiltración Neutrófila , Transducción de Señal
4.
Immunol Rev ; 314(1): 427-441, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36326284

RESUMEN

Neutrophils are a critical element of host defense and are rapidly recruited to inflammatory sites. Such sites are frequently limited in oxygen and/or nutrient availability, presenting a metabolic challenge for infiltrating cells. Long believed to be uniquely dependent on glycolysis, it is now clear that neutrophils possess far greater metabolic plasticity than previously thought, with the capacity to generate energy stores and utilize extracellular proteins to fuel central carbon metabolism and biosynthetic activity. Out-with cellular energetics, metabolic programs have also been implicated in the production of neutrophils and their progenitors in the bone marrow compartment, activation of neutrophil antimicrobial responses, inflammatory and cell survival signaling cascades, and training of the innate immune response. Thus, understanding the mechanisms by which metabolic processes sustain changes in neutrophil effector functions and how these are subverted in disease states provides exciting new avenues for the treatment of dysfunctional neutrophilic inflammation which are lacking in clinical practice to date.


Asunto(s)
Inmunidad Innata , Inflamación , Humanos , Neutrófilos
5.
Blood ; 139(2): 281-286, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34411229

RESUMEN

Neutrophils are predominantly glycolytic cells that derive little ATP from oxidative phosphorylation; however, they possess an extensive mitochondrial network and maintain a mitochondrial membrane potential. Although studies have shown neutrophils need their mitochondria to undergo apoptosis and regulate NETosis, the metabolic role of the respiratory chain in these highly glycolytic cells is still unclear. Recent studies have expanded on the role of reactive oxygen species (ROS) released from the mitochondria as intracellular signaling molecules. Our study shows that neutrophils can use their mitochondria to generate ROS and that mitochondrial ROS release is increased in hypoxic conditions. This is needed for the stabilization of a high level of the critical hypoxic response factor and pro-survival protein HIF-1α in hypoxia. Further, we demonstrate that neutrophils use the glycerol 3-phosphate pathway as a way of directly regulating mitochondrial function through glycolysis, specifically to maintain polarized mitochondria and produce ROS. This illustrates an additional pathway by which neutrophils can regulate HIF-1α stability and will therefore be an important consideration when looking for treatments of inflammatory conditions in which HIF-1α activation and neutrophil persistence at the site of inflammation are linked to disease severity.


Asunto(s)
Glicerofosfatos/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Mitocondrias/metabolismo , Neutrófilos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Hipoxia de la Célula , Células Cultivadas , Humanos , Estabilidad Proteica
6.
Am J Respir Crit Care Med ; 207(8): 998-1011, 2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36724365

RESUMEN

Rationale: Chronic obstructive pulmonary disease (COPD) is a disease characterized by persistent airway inflammation and disordered macrophage function. The extent to which alterations in macrophage bioenergetics contribute to impaired antioxidant responses and disease pathogenesis has yet to be fully delineated. Objectives: Through the study of COPD alveolar macrophages (AMs) and peripheral monocyte-derived macrophages (MDMs), we sought to establish if intrinsic defects in core metabolic processes drive macrophage dysfunction and redox imbalance. Methods: AMs and MDMs from donors with COPD and healthy donors underwent functional, metabolic, and transcriptional profiling. Measurements and Main Results: We observed that AMs and MDMs from donors with COPD display a critical depletion in glycolytic- and mitochondrial respiration-derived energy reserves and an overreliance on glycolysis as a source for ATP, resulting in reduced energy status. Defects in oxidative metabolism extend to an impaired redox balance associated with defective expression of the NADPH-generating enzyme, ME1 (malic enzyme 1), a known target of the antioxidant transcription factor NRF2 (nuclear factor erythroid 2-related factor 2). Consequently, selective activation of NRF2 resets the COPD transcriptome, resulting in increased generation of TCA cycle intermediaries, improved energetic status, favorable redox balance, and recovery of macrophage function. Conclusions: In COPD, an inherent loss of metabolic plasticity leads to metabolic exhaustion and reduced redox capacity, which can be rescued by activation of the NRF2 pathway. Targeting these defects, via NRF2 augmentation, may therefore present an attractive therapeutic strategy for the treatment of the aberrant airway inflammation described in COPD.


Asunto(s)
Factor 2 Relacionado con NF-E2 , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Macrófagos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Malato Deshidrogenasa/metabolismo
7.
EMBO Rep ; 20(5)2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30872317

RESUMEN

Inflamed and infected tissue sites are characterised by oxygen and nutrient deprivation. The cellular adaptations to insufficient oxygenation, hypoxia, are mainly regulated by a family of transcription factors known as hypoxia-inducible factors (HIFs). The protein members of the HIF signalling pathway are critical regulators of both the innate and adaptive immune responses, and there is an increasing body of evidence to suggest that the elicited changes occur through cellular metabolic reprogramming. Here, we review the literature on innate immunometabolism to date and discuss the role of hypoxia in innate cell metabolic reprogramming, and how this determines immune responses.


Asunto(s)
Inflamación/metabolismo , Células Mieloides/metabolismo , Inmunidad Adaptativa/inmunología , Animales , Hipoxia de la Célula/inmunología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunidad Innata/inmunología , Inflamación/inmunología , Células Mieloides/inmunología , Oxígeno/metabolismo , Transducción de Señal/inmunología
8.
Nature ; 573(7772): 41-42, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31481766
9.
Am J Respir Crit Care Med ; 200(2): 235-246, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-30849228

RESUMEN

Rationale: Acute respiratory distress syndrome is defined by the presence of systemic hypoxia and consequent on disordered neutrophilic inflammation. Local mechanisms limiting the duration and magnitude of this neutrophilic response remain poorly understood. Objectives: To test the hypothesis that during acute lung inflammation tissue production of proresolution type 2 cytokines (IL-4 and IL-13) dampens the proinflammatory effects of hypoxia through suppression of HIF-1α (hypoxia-inducible factor-1α)-mediated neutrophil adaptation, resulting in resolution of lung injury. Methods: Neutrophil activation of IL4Ra (IL-4 receptor α) signaling pathways was explored ex vivo in human acute respiratory distress syndrome patient samples, in vitro after the culture of human peripheral blood neutrophils with recombinant IL-4 under conditions of hypoxia, and in vivo through the study of IL4Ra-deficient neutrophils in competitive chimera models and wild-type mice treated with IL-4. Measurements and Main Results: IL-4 was elevated in human BAL from patients with acute respiratory distress syndrome, and its receptor was identified on patient blood neutrophils. Treatment of human neutrophils with IL-4 suppressed HIF-1α-dependent hypoxic survival and limited proinflammatory transcriptional responses. Increased neutrophil apoptosis in hypoxia, also observed with IL-13, required active STAT signaling, and was dependent on expression of the oxygen-sensing prolyl hydroxylase PHD2. In vivo, IL-4Ra-deficient neutrophils had a survival advantage within a hypoxic inflamed niche; in contrast, inflamed lung treatment with IL-4 accelerated resolution through increased neutrophil apoptosis. Conclusions: We describe an important interaction whereby IL4Rα-dependent type 2 cytokine signaling can directly inhibit hypoxic neutrophil survival in tissues and promote resolution of neutrophil-mediated acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Subunidad alfa del Receptor de Interleucina-4/inmunología , Interleucina-4/inmunología , Neutrófilos/inmunología , Receptores de Superficie Celular/inmunología , Síndrome de Dificultad Respiratoria/inmunología , Lesión Pulmonar Aguda/metabolismo , Animales , Apoptosis/efectos de los fármacos , Hipoxia de la Célula/inmunología , Supervivencia Celular/efectos de los fármacos , Regulación de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-4/metabolismo , Interleucina-4/farmacología , Subunidad alfa del Receptor de Interleucina-4/genética , Subunidad alfa del Receptor de Interleucina-4/metabolismo , Ratones , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Receptores de Superficie Celular/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal
10.
Am J Respir Crit Care Med ; 198(6): 739-750, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29547002

RESUMEN

RATIONALE: Previous studies have identified defects in bacterial phagocytosis by alveolar macrophages (AMs) in patients with chronic obstructive pulmonary disease (COPD), but the mechanisms and clinical consequences remain incompletely defined. OBJECTIVES: To examine the effect of COPD on AM phagocytic responses and identify the mechanisms, clinical consequences, and potential for therapeutic manipulation of these defects. METHODS: We isolated AMs and monocyte-derived macrophages (MDMs) from a cohort of patients with COPD and control subjects within the Medical Research Council COPDMAP consortium and measured phagocytosis of bacteria in relation to opsonic conditions and clinical features. MEASUREMENTS AND MAIN RESULTS: COPD AMs and MDMs have impaired phagocytosis of Streptococcus pneumoniae. COPD AMs have a selective defect in uptake of opsonized bacteria, despite the presence of antipneumococcal antibodies in BAL, not observed in MDMs or healthy donor AMs. AM defects in phagocytosis in COPD are significantly associated with exacerbation frequency, isolation of pathogenic bacteria, and health-related quality-of-life scores. Bacterial binding and initial intracellular killing of opsonized bacteria in COPD AMs was not reduced. COPD AMs have reduced transcriptional responses to opsonized bacteria, such as cellular stress responses that include transcriptional modules involving antioxidant defenses and Nrf2 (nuclear factor erythroid 2-related factor 2)-regulated genes. Agonists of the cytoprotective transcription factor Nrf2 (sulforaphane and compound 7) reverse defects in phagocytosis of S. pneumoniae and nontypeable Haemophilus influenzae by COPD AMs. CONCLUSIONS: Patients with COPD have clinically relevant defects in opsonic phagocytosis by AMs, associated with impaired transcriptional responses to cellular stress, which are reversed by therapeutic targeting with Nrf2 agonists.


Asunto(s)
Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Fagocitosis/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Humanos , Isotiocianatos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/fisiología , Masculino , Persona de Mediana Edad , Fagocitosis/fisiología , Streptococcus pneumoniae , Sulfóxidos
11.
Br Med Bull ; 128(1): 5-14, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30137312

RESUMEN

Introduction: It has been known for some time that neutrophils are present in the tumour microenvironment, but only recently have their roles been explored. Sources of data: Comprehensive literature search of neutrophils and cancer (PubMed, Google Scholar and CrossRef) for key articles (systematic reviews, meta-analyses, primary research). References from these articles cross-checked for additional relevant studies. Areas of agreement: Neutrophils are a heterogeneous population with both pro- and antitumour roles, and display plasticity. Several neutrophil subpopulations have been identified, defined by a combination of features (density, maturity, surface markers, morphology and anatomical site). Areas of controversy: Limitations in translating murine tumour models to human pathology and paucity of human data. Consensus in defining human neutrophil subpopulations. Growing points: Neutrophils as therapeutic targets and as possible playmakers in the biological response to newer targeted cancer drugs. Areas timely for developing research: Understanding the metabolic programming of neutrophils in the tumour microenvironment.


Asunto(s)
Neoplasias/inmunología , Neutrófilos/patología , Microambiente Tumoral/inmunología , Humanos , Inmunidad Celular/fisiología , Neoplasias/patología , Neutrófilos/inmunología , Transducción de Señal
12.
Blood ; 123(18): 2753-4, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24786455

RESUMEN

In this issue of Blood, Jun et al, through the study of neutrophils deficient in the glucose-6-phosphate transporter, describe a novel role for the peroxisome proliferator-activated receptor-γ (PPARG) pathway in the regulation of key neutrophil functions and link this to concomitant hypoxia-inducible factor (HIF) 1α stabilization.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/metabolismo , Neutrófilos/metabolismo , Humanos
13.
Blood ; 123(3): 366-76, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24196071

RESUMEN

Neutrophil lifespan and function are regulated by hypoxia via components of the hypoxia inducible factor (HIF)/von Hippel Lindau/hydroxylase pathway, including specific roles for HIF-1α and prolyl hydroxylase-3. HIF-2α has both distinct and overlapping biological roles with HIF-1α and has not previously been studied in the context of neutrophil biology. We investigated the role of HIF-2α in regulating key neutrophil functions. Human and murine peripheral blood neutrophils expressed HIF-2α, with expression up-regulated by acute and chronic inflammatory stimuli and in disease-associated inflammatory neutrophil. HIF2A gain-of-function mutations resulted in a reduction in neutrophil apoptosis both ex vivo, through the study of patient cells, and in vivo in a zebrafish tail injury model. In contrast, HIF-2α-deficient murine inflammatory neutrophils displayed increased sensitivity to nitrosative stress induced apoptosis ex vivo and increased neutrophil apoptosis in vivo, resulting in a reduction in neutrophilic inflammation and reduced tissue injury. Expression of HIF-2α was temporally dissociated from HIF-1α in vivo and predominated in the resolution phase of inflammation. These data support a critical and selective role for HIF-2α in persistence of neutrophilic inflammation and provide a platform to dissect the therapeutic utility of targeting HIF-2α in chronic inflammatory diseases.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación de la Expresión Génica , Inflamación , Neutrófilos/metabolismo , Animales , Apoptosis , Hipoxia de la Célula , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Muramidasa , Neutrófilos/citología , Fagocitosis , Fenotipo , ARN/metabolismo , Estallido Respiratorio , Pez Cebra
14.
PLoS Pathog ; 9(12): e1003789, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24367256

RESUMEN

Tuberculosis is a current major world-health problem, exacerbated by the causative pathogen, Mycobacterium tuberculosis (Mtb), becoming increasingly resistant to conventional antibiotic treatment. Mtb is able to counteract the bactericidal mechanisms of leukocytes to survive intracellularly and develop a niche permissive for proliferation and dissemination. Understanding of the pathogenesis of mycobacterial infections such as tuberculosis (TB) remains limited, especially for early infection and for reactivation of latent infection. Signaling via hypoxia inducible factor α (HIF-α) transcription factors has previously been implicated in leukocyte activation and host defence. We have previously shown that hypoxic signaling via stabilization of Hif-1α prolongs the functionality of leukocytes in the innate immune response to injury. We sought to manipulate Hif-α signaling in a well-established Mycobacterium marinum (Mm) zebrafish model of TB to investigate effects on the host's ability to combat mycobacterial infection. Stabilization of host Hif-1α, both pharmacologically and genetically, at early stages of Mm infection was able to reduce the bacterial burden of infected larvae. Increasing Hif-1α signaling enhanced levels of reactive nitrogen species (RNS) in neutrophils prior to infection and was able to reduce larval mycobacterial burden. Conversely, decreasing Hif-2α signaling enhanced RNS levels and reduced bacterial burden, demonstrating that Hif-1α and Hif-2α have opposing effects on host susceptibility to mycobacterial infection. The antimicrobial effect of Hif-1α stabilization, and Hif-2α reduction, were demonstrated to be dependent on inducible nitric oxide synthase (iNOS) signaling at early stages of infection. Our findings indicate that induction of leukocyte iNOS by stabilizing Hif-1α, or reducing Hif-2α, aids the host during early stages of Mm infection. Stabilization of Hif-1α therefore represents a potential target for therapeutic intervention against tuberculosis.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Infecciones por Mycobacterium no Tuberculosas/inmunología , Mycobacterium marinum , Óxido Nítrico/metabolismo , Animales , Animales Modificados Genéticamente , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Infecciones por Mycobacterium no Tuberculosas/genética , Infecciones por Mycobacterium no Tuberculosas/microbiología , Neutrófilos/metabolismo , Óxido Nítrico Sintasa de Tipo II/fisiología , Nitrosación , Transducción de Señal/genética , Pez Cebra
15.
Circ Res ; 112(12): 1583-91, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23564640

RESUMEN

RATIONALE: Hypoxia followed by reoxygenation promotes inflammation by activating nuclear factor κB transcription factors in endothelial cells (ECs). This process involves modification of the signaling intermediary tumor necrosis factor receptor-associated factor 6 with polyubiquitin chains. Thus, cellular mechanisms that suppress tumor necrosis factor receptor-associated factor 6 ubiquitination are potential therapeutic targets to reduce inflammation in hypoxic tissues. OBJECTIVE: In this study, we tested the hypothesis that endothelial activation in response to hypoxia-reoxygenation can be influenced by Cezanne, a deubiquitinating enzyme that cleaves ubiquitin from specific modified proteins. METHODS AND RESULTS: Studies of cultured ECs demonstrated that hypoxia (1% oxygen) induced Cezanne via p38 mitogen-activated protein kinase-dependent transcriptional and post-transcriptional mechanisms. Hypoxia-reoxygenation had minimal effects on proinflammatory signaling in unmanipulated ECs but significantly enhanced Lys63 polyubiquitination of tumor necrosis factor receptor-associated factor 6, activation of nuclear factor κB, and expression of inflammatory genes after silencing of Cezanne. Thus, although hypoxia primed cells for inflammatory activation, it simultaneously induced Cezanne, which impeded signaling to nuclear factor κB by suppressing tumor necrosis factor receptor-associated factor 6 ubiquitination. Similarly, ischemia induced Cezanne in the murine kidney in vascular ECs, glomerular ECs, podocytes, and epithelial cells, and genetic deletion of Cezanne enhanced renal inflammation and injury in murine kidneys exposed to ischemia followed by reperfusion. CONCLUSIONS: We conclude that inflammatory responses to ischemia are controlled by a balance between ubiquitination and deubiquitination, and that Cezanne is a key regulator of this process. Our observations have important implications for therapeutic targeting of inflammation and injury during ischemia-reperfusion.


Asunto(s)
Endopeptidasas/metabolismo , Células Endoteliales/enzimología , Inflamación/prevención & control , Riñón/irrigación sanguínea , Daño por Reperfusión/enzimología , Factor 6 Asociado a Receptor de TNF/metabolismo , Animales , Hipoxia de la Célula , Células Cultivadas , Modelos Animales de Enfermedad , Endopeptidasas/deficiencia , Endopeptidasas/genética , Células Endoteliales/inmunología , Humanos , Inflamación/enzimología , Inflamación/genética , Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Oxígeno/metabolismo , Interferencia de ARN , Ratas , Ratas Endogámicas F344 , Daño por Reperfusión/genética , Daño por Reperfusión/inmunología , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/genética , Factores de Tiempo , Transcripción Genética , Transfección , Ubiquitinación , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
16.
J Immunol ; 189(4): 1955-65, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22786772

RESUMEN

Hypoxia and HIFs (HIF-1α and HIF-2α) modulate innate immune responses in the setting of systemic inflammatory responses and sepsis. The HIF prolyl hydroxylase enzymes PHD1, PHD2 and PHD3 regulate the mammalian adaptive response to hypoxia; however, their significance in the innate immune response has not been elucidated. We demonstrate in this study that deficiency of PHD3 (PHD3(-/-)) specifically shortens the survival of mice subjected to various models of abdominal sepsis because of an overwhelming innate immune response, leading to premature organ dysfunction. By contrast, this phenotype was absent in mice deficient for PHD1 (PHD1(-/-)) or PHD2 (PHD2(+/-)). In vivo, plasma levels of proinflammatory cytokines were enhanced, and recruitment of macrophages to internal organs was increased in septic PHD3-deficient mice. Reciprocal bone marrow transplantation in sublethally irradiated mice revealed that enhanced susceptibility of PHD3-deficient mice to sepsis-related lethality was specifically caused by loss of PHD3 in myeloid cells. Several in vitro assays revealed enhanced cytokine production, migration, phagocytic capacity, and proinflammatory activation of PHD3-deficient macrophages. Increased proinflammatory activity of PHD3-deficient macrophages occurred concomitantly with enhanced HIF-1α protein stabilization and increased NF-κB activity, and interference with the expression of HIF-1α or the canonical NF-κB pathway blunted their proinflammatory phenotype. It is concluded that impairment of PHD3 enzyme function aggravates the clinical course of abdominal sepsis via HIF-1α- and NF-κB-mediated enhancement of the innate immune response.


Asunto(s)
Inmunidad Innata/inmunología , Macrófagos/inmunología , Procolágeno-Prolina Dioxigenasa/inmunología , Sepsis/inmunología , Transducción de Señal/inmunología , Animales , Western Blotting , Quimiotaxis de Leucocito/inmunología , Citocinas/biosíntesis , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunohistoquímica , Macrófagos/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/inmunología , FN-kappa B/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Sepsis/metabolismo
17.
Biol Chem ; 394(4): 471-7, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23324383

RESUMEN

Many inflammatory diseases are characterised by persistent and inappropriate neutrophil activation, systemic or localised hypoxia, and bacterial colonisation. Hypoxia represents an important regulator of inflammatory responses because it inhibits neutrophil apoptosis, a process central to the timely resolution of inflammation. Progress in understanding how cells respond to hypoxia has led to the identification of hypoxia-inducible transcription factors (HIFs) and their hydroxylation by the prolyl hydroxylase enzymes. There is now a significant body of data to support a critical role for this HIF pathway in regulating neutrophil function. Moreover, manipulations of specific components of this pathway have very divergent effects on myeloid cell function. In this review, we will discuss the role individual members of the HIF pathway play in regulating key neutrophil functions and the implications this has for the development of effective therapeutic strategies that selectively target inappropriate neutrophil persistence while maintaining a fully competent immune response.


Asunto(s)
Hipoxia/metabolismo , Oxígeno/metabolismo , Animales , Humanos , Inmunidad Innata/fisiología , Células Mieloides/metabolismo , Procolágeno-Prolina Dioxigenasa/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología
18.
Blood ; 118(3): 712-22, 2011 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-21555741

RESUMEN

The oxygen-sensing transcription factor hypoxia-inducible factor-1α (HIF-1α) plays a critical role in the regulation of myeloid cell function. The mechanisms of regulation are not well understood, nor are the phenotypic consequences of HIF modulation in the context of neutrophilic inflammation. Species conservation across higher metazoans enables the use of the genetically tractable and transparent zebrafish (Danio rerio) embryo to study in vivo resolution of the inflammatory response. Using both a pharmacologic approach known to lead to stabilization of HIF-1α, and selective genetic manipulation of zebrafish HIF-1α homologs, we sought to determine the roles of HIF-1α in inflammation resolution. Both approaches reveal that activated Hif-1α delays resolution of inflammation after tail transection in zebrafish larvae. This delay can be replicated by neutrophil-specific Hif activation and is a consequence of both reduced neutrophil apoptosis and increased retention of neutrophils at the site of tissue injury. Hif-activated neutrophils continue to patrol the injury site during the resolution phase, when neutrophils would normally migrate away. Site-directed mutagenesis of Hif in vivo reveals that hydroxylation of Hif-1α by prolyl hydroxylases critically regulates the Hif pathway in zebrafish neutrophils. Our data demonstrate that Hif-1α regulates neutrophil function in complex ways during inflammation resolution in vivo.


Asunto(s)
Apoptosis/inmunología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/inmunología , Inflamación/inmunología , Neutrófilos/inmunología , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/inmunología , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Movimiento Celular/inmunología , Modelos Animales de Enfermedad , Embrión no Mamífero/inmunología , Femenino , Variación Genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/inmunología , Proteínas de Pez Cebra/metabolismo
20.
J Immunol ; 186(1): 453-463, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21135168

RESUMEN

Neutrophils play a central role in the innate immune response and a critical role in bacterial killing. Most studies of neutrophil function have been conducted under conditions of ambient oxygen, but inflamed sites where neutrophils operate may be extremely hypoxic. Previous studies indicate that neutrophils sense and respond to hypoxia via the ubiquitous prolyl hydroxylase/hypoxia-inducible factor pathway and that this can signal for enhanced survival. In the current study, human neutrophils were shown to upregulate hypoxia-inducible factor (HIF)-1α-dependent gene expression under hypoxic incubation conditions (3 kPa), with a consequent substantial delay in the onset of apoptosis. Despite this, polarization and chemotactic responsiveness to IL-8 and fMLP were entirely unaffected by hypoxia. Similarly, hypoxia did not diminish the ability of neutrophils to phagocytose serum-opsonized heat-killed streptococci. Of the secretory functions examined, IL-8 generation was preserved and elastase release was enhanced by hypoxia. Hypoxia did, however, cause a major reduction in respiratory burst activity induced both by the soluble agonist fMLP and by ingestion of opsonized zymosan, without affecting expression of the NADPH oxidase subunits. Critically, this reduction in respiratory burst activity under hypoxia was associated with a significant defect in the killing of Staphylococcus aureus. In contrast, killing of Escherichia coli, which is predominantly oxidase independent, was fully preserved under hypoxia. In conclusion, these studies suggest that although the NADPH oxidase-dependent bacterial killing mechanism may be compromised by hypoxia, neutrophils overall appear extremely well adapted to operate successfully under severely hypoxic conditions.


Asunto(s)
Actividad Bactericida de la Sangre/inmunología , Neutrófilos/inmunología , Neutrófilos/microbiología , Estallido Respiratorio/inmunología , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/inmunología , Degranulación de la Célula/inmunología , Hipoxia de la Célula/inmunología , Células Cultivadas , Quimiotaxis de Leucocito/inmunología , Humanos , Elastasa de Leucocito/metabolismo , N-Formilmetionina Leucil-Fenilalanina/farmacología , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/sangre , NADPH Oxidasas/fisiología , Neutrófilos/enzimología , Neutrófilos/metabolismo , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/sangre , Receptores Inmunológicos/sangre , Receptores Inmunológicos/metabolismo , Staphylococcus aureus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA