Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 23(6): 927-939, 2022 06.
Article in English | MEDLINE | ID: mdl-35624205

ABSTRACT

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.


Subject(s)
Lung Injury , Respiratory Distress Syndrome , Animals , Humans , Hypoxia/etiology , Inflammation/complications , Lung , Lung Injury/complications , Mice
3.
Immunity ; 48(5): 846-848, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29768171

ABSTRACT

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.


Subject(s)
Immunity, Innate , Zebrafish , Animals , Neutrophil Infiltration , Signal Transduction
4.
Immunol Rev ; 314(1): 427-441, 2023 03.
Article in English | MEDLINE | ID: mdl-36326284

ABSTRACT

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.


Subject(s)
Immunity, Innate , Inflammation , Humans , Neutrophils
5.
J Hepatol ; 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38879173

ABSTRACT

BACKGROUND & AIMS: Biliary complications are a major cause of morbidity and mortality in liver transplantation. Up to 25% of patients that develop biliary complications require additional surgical procedures, re-transplantation or die in the absence of a suitable regraft. Here, we investigate the role of the primary cilium, a highly-specialised sensory organelle, in biliary injury leading to post-transplant biliary complications. METHODS: Human biopsies were used to study the structure and function of primary cilia in liver transplant recipients that develop biliary complications (N=7) in comparison with recipients without biliary complications (N=12). To study the biological effects of the primary cilia during transplantation, we generated murine models that recapitulate liver procurement and cold storage, and assessed the elimination of the primary cilia in biliary epithelial cells in the K19CreERTKif3aflox/flox mouse model. To explore the molecular mechanisms responsible for the observed phenotypes we used in vitro models of ischemia, cellular senescence and primary cilia ablation. Finally, we used pharmacological and genetic approaches to target cellular senescence and the primary cilia, both in mouse models and discarded human donor livers. RESULTS: Prolonged ischemic periods before transplantation result in ciliary shortening and cellular senescence, an irreversible cell cycle arrest that blocks regeneration. Our results indicate that primary cilia damage results in biliary injury and a loss of regenerative potential. Senescence negatively impacts primary cilia structure and triggers a negative feedback loop that further impairs regeneration. Finally, we explore how targeted interventions for cellular senescence and/or the stabilisation of the primary cilia improve biliary regeneration following ischemic injury. CONCLUSIONS: Primary cilia play an essential role in biliary regeneration and we demonstrate that senolytics and cilia-stabilising treatments provide a potential therapeutic opportunity to reduce the rate of biliary complications and improve clinical outcomes in liver transplantation. IMPACT AND IMPLICATIONS: Up to 25% of liver transplants result in biliary complications, leading to additional surgery, retransplants, or death. We found that the incidence of biliary complications is increased by damage to the primary cilium, an antenna that protrudes from the cell and is key to regeneration. Here, we show that treatments that preserve the primary cilia during the transplant process provide a potential solution to reduce the rates of biliary complications.

6.
Blood ; 139(2): 281-286, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34411229

ABSTRACT

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.


Subject(s)
Glycerophosphates/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitochondria/metabolism , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Cell Hypoxia , Cells, Cultured , Humans , Protein Stability
7.
Am J Respir Crit Care Med ; 207(8): 998-1011, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36724365

ABSTRACT

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.


Subject(s)
NF-E2-Related Factor 2 , Pulmonary Disease, Chronic Obstructive , Humans , Macrophages/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/physiopathology , Malate Dehydrogenase/metabolism
8.
J Manipulative Physiol Ther ; 45(2): 163-169, 2022 02.
Article in English | MEDLINE | ID: mdl-35753872

ABSTRACT

OBJECTIVE: The purpose of this study was to determine the feasibility of using detuned laser as a placebo intervention in manual therapy research. METHODS: We performed a secondary data analysis of a randomized controlled trial. In our analysis, 30 participants with chronic ankle instability (manual therapy group: n = 13, age = 33.1 ± 8.1 years, female participants = 50%; detuned laser group: n = 17, age = 31.9 ± 11.8 years, female participants = 72%) were asked to indicate which intervention (manual therapy [active] or detuned laser [placebo]), they thought they had received and to give a confidence rating on their response regarding the received intervention at the conclusion of the course of intervention. Independent t tests were used to compare the groups. Participants in both groups were asked the following open-ended question: "What did you think of the intervention?". RESULTS: There were 52.9% participants in the detuned laser group and 53.8% participants in the manual therapy group who perceived that they had received the active intervention. The confidence ratings about their perceptions (6.7 ± 2.0, detuned laser group; 6.3 ± 2.4, manual therapy group) (P = .66) and the self-reported recovery ratings (1.9 ± 1.5 and 1.8 ± 1.2, respectively) (P = .77) were similar. CONCLUSIONS: Participants in this study confidently perceived that detuned laser was an active intervention. They positively rated their recovery following the course of the placebo intervention and perceived that detuned laser was effective in treating their condition. Therefore, it is feasible for detuned laser to be used as a placebo for manual therapy trials.


Subject(s)
Joint Instability/therapy , Laser Therapy , Musculoskeletal Manipulations , Adult , Feasibility Studies , Female , Humans , Lasers/classification , Male , Young Adult
9.
EMBO Rep ; 20(5)2019 05.
Article in English | MEDLINE | ID: mdl-30872317

ABSTRACT

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.


Subject(s)
Inflammation/metabolism , Myeloid Cells/metabolism , Adaptive Immunity/immunology , Animals , Cell Hypoxia/immunology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunity, Innate/immunology , Inflammation/immunology , Myeloid Cells/immunology , Oxygen/metabolism , Signal Transduction/immunology
10.
Nature ; 522(7556): 349-53, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-25985180

ABSTRACT

Mutations or amplification of the MET proto-oncogene are involved in the pathogenesis of several tumours, which rely on the constitutive engagement of this pathway for their growth and survival. However, MET is expressed not only by cancer cells but also by tumour-associated stromal cells, although its precise role in this compartment is not well characterized. Here we show that MET is required for neutrophil chemoattraction and cytotoxicity in response to its ligand hepatocyte growth factor (HGF). Met deletion in mouse neutrophils enhances tumour growth and metastasis. This phenotype correlates with reduced neutrophil infiltration to both the primary tumour and metastatic sites. Similarly, Met is necessary for neutrophil transudation during colitis, skin rash or peritonitis. Mechanistically, Met is induced by tumour-derived tumour necrosis factor (TNF)-α or other inflammatory stimuli in both mouse and human neutrophils. This induction is instrumental for neutrophil transmigration across an activated endothelium and for inducible nitric oxide synthase production upon HGF stimulation. Consequently, HGF/MET-dependent nitric oxide release by neutrophils promotes cancer cell killing, which abates tumour growth and metastasis. After systemic administration of a MET kinase inhibitor, we prove that the therapeutic benefit of MET targeting in cancer cells is partly countered by the pro-tumoural effect arising from MET blockade in neutrophils. Our work identifies an unprecedented role of MET in neutrophils, suggests a potential 'Achilles' heel' of MET-targeted therapies in cancer, and supports the rationale for evaluating anti-MET drugs in certain inflammatory diseases.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Neoplasms/immunology , Neoplasms/metabolism , Neutrophils/immunology , Proto-Oncogene Proteins c-met/metabolism , Aged , Animals , Disease Models, Animal , Disease Progression , Female , Gene Deletion , Hepatocyte Growth Factor , Humans , Inflammation/immunology , Inflammation/pathology , Male , Mice , Middle Aged , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/pathology , Neutrophils/drug effects , Neutrophils/metabolism , Nitric Oxide/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/deficiency , Proto-Oncogene Proteins c-met/genetics , Solubility , Transendothelial and Transepithelial Migration , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays
11.
Nature ; 573(7772): 41-42, 2019 09.
Article in English | MEDLINE | ID: mdl-31481766
12.
Am J Respir Crit Care Med ; 200(2): 235-246, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30849228

ABSTRACT

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.


Subject(s)
Acute Lung Injury/immunology , Interleukin-4 Receptor alpha Subunit/immunology , Interleukin-4/immunology , Neutrophils/immunology , Receptors, Cell Surface/immunology , Respiratory Distress Syndrome/immunology , Acute Lung Injury/metabolism , Animals , Apoptosis/drug effects , Cell Hypoxia/immunology , Cell Survival/drug effects , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-4/metabolism , Interleukin-4/pharmacology , Interleukin-4 Receptor alpha Subunit/genetics , Interleukin-4 Receptor alpha Subunit/metabolism , Mice , Mice, Knockout , Neutrophils/drug effects , Neutrophils/metabolism , Receptors, Cell Surface/metabolism , Respiratory Distress Syndrome/metabolism , STAT Transcription Factors/metabolism , Signal Transduction
13.
Am J Respir Crit Care Med ; 198(6): 739-750, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29547002

ABSTRACT

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.


Subject(s)
NF-E2-Related Factor 2/antagonists & inhibitors , Phagocytosis/drug effects , Pulmonary Disease, Chronic Obstructive/physiopathology , Adult , Aged , Case-Control Studies , Female , Humans , Isothiocyanates/pharmacology , Macrophages/drug effects , Macrophages/physiology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/physiology , Male , Middle Aged , Phagocytosis/physiology , Streptococcus pneumoniae , Sulfoxides
14.
Br Med Bull ; 128(1): 5-14, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30137312

ABSTRACT

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.


Subject(s)
Neoplasms/immunology , Neutrophils/pathology , Tumor Microenvironment/immunology , Humans , Immunity, Cellular/physiology , Neoplasms/pathology , Neutrophils/immunology , Signal Transduction
15.
Blood ; 123(18): 2753-4, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24786455

ABSTRACT

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.


Subject(s)
Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/metabolism , Neutrophils/metabolism , Humans
16.
Blood ; 123(3): 366-76, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24196071

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation , Inflammation , Neutrophils/metabolism , Animals , Apoptosis , Cell Hypoxia , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Muramidase , Neutrophils/cytology , Phagocytosis , Phenotype , RNA/metabolism , Respiratory Burst , Zebrafish
17.
PLoS Pathog ; 9(12): e1003789, 2013.
Article in English | MEDLINE | ID: mdl-24367256

ABSTRACT

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.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium marinum , Nitric Oxide/metabolism , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/physiology , Cells, Cultured , Disease Models, Animal , Genetic Predisposition to Disease , Mycobacterium Infections, Nontuberculous/genetics , Mycobacterium Infections, Nontuberculous/microbiology , Neutrophils/metabolism , Nitric Oxide Synthase Type II/physiology , Nitrosation , Signal Transduction/genetics , Zebrafish
18.
Circ Res ; 112(12): 1583-91, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23564640

ABSTRACT

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.


Subject(s)
Endopeptidases/metabolism , Endothelial Cells/enzymology , Inflammation/prevention & control , Kidney/blood supply , Reperfusion Injury/enzymology , TNF Receptor-Associated Factor 6/metabolism , Animals , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , Endopeptidases/deficiency , Endopeptidases/genetics , Endothelial Cells/immunology , Humans , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Oxygen/metabolism , RNA Interference , Rats , Rats, Inbred F344 , Reperfusion Injury/genetics , Reperfusion Injury/immunology , Signal Transduction , TNF Receptor-Associated Factor 6/genetics , Time Factors , Transcription, Genetic , Transfection , Ubiquitination , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
19.
J Manipulative Physiol Ther ; 38(3): 218-24, 2015.
Article in English | MEDLINE | ID: mdl-25704219

ABSTRACT

OBJECTIVE: The purpose of this study was to determine the intrarater reliability and reproducibility of a standardized procedure for measuring passive shoulder movement in asymptomatic individuals. METHODS: A single assessor used a digital inclinometer and standardized protocol to measure the passive range of motion of 7 shoulder movements in 168 asymptomatic shoulders. Following a warm-up maneuver, 3 measurements were taken for each movement on 2 occasions. Both shoulders were measured using a standardized order of movement. Selection of measurement beginning with left or right shoulder was randomly determined. The entire process was repeated 7 days later to assess reproducibility. Intraclass correlation coefficients (ICCs) with 95% confidence intervals and standard errors of measurement (SEMs) were calculated to assess the intrarater reliability of the methods. RESULTS: The intrarater reliability of our methods was substantial for total shoulder flexion (ICC = 0.82, SEM = 12.3°), whereas all other movements demonstrated moderate reliability (ICC range = 0.64-0.75) except external rotation in neutral abduction, for which reliability was classed as slight (ICC = 0.28, SEM = 31°). Moderate reliability was evident for all movements on follow-up at 7 days (ICC range = 0.60-0.77). CONCLUSIONS: These methods of measurement have moderate to substantial reliability for the majority of tested passive shoulder movements, with moderate reliability sustained after 1 week, in a large sample of asymptomatic individuals.


Subject(s)
Range of Motion, Articular , Shoulder Joint/physiology , Female , Humans , Male , Observer Variation , Reproducibility of Results , Young Adult
20.
J Immunol ; 189(4): 1955-65, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22786772

ABSTRACT

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.


Subject(s)
Immunity, Innate/immunology , Macrophages/immunology , Procollagen-Proline Dioxygenase/immunology , Sepsis/immunology , Signal Transduction/immunology , Animals , Blotting, Western , Chemotaxis, Leukocyte/immunology , Cytokines/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Macrophages/metabolism , Mice , Mice, Knockout , NF-kappa B/immunology , NF-kappa B/metabolism , Procollagen-Proline Dioxygenase/metabolism , Real-Time Polymerase Chain Reaction , Sepsis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL