Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Mol Pharmacol ; 99(4): 277-285, 2021 04.
Article in English | MEDLINE | ID: mdl-33436521

ABSTRACT

Acetaminophen (APAP) is a commonly used pain and fever reliever but is also the most frequent cause of drug-induced liver injury. The mechanism pertaining acetaminophen toxicity has been well documented, whereas mechanisms of hepatotoxicity are not well established. Serine (or cysteine) peptidase inhibitor, clade A, member 3N (SerpinA3N), a serine protease inhibitor, is synthesized in the liver but the role of SerpinA3N in relation to APAP-induced liver injury is not known. Wild-type and hepatocyte-specific SerpinA3N knockout (HKO) mice were injected intraperitoneally with a single dose of PBS or APAP (400 mg/kg) for 12 hours, and markers of liver injury, cell death, and inflammation were assessed. SerpinA3N expression was highly induced in mice with APAP overdose. SerpinA3N HKO mice had diminished liver injury and necrosis as shown by lower alanine aminotransferase and interleukin-6 levels, accompanied by suppressed inflammatory cytokines and reduced neutrophil infiltration. The reduced oxidative stress was associated with enhanced antioxidant enzyme capabilities. Taken together, hepatocyte SerpinA3N deficiency reduced APAP-induced liver injury by ameliorating inflammation and modulating the 5' AMP-activated protein kinase-unc-51-like autophagy activating kinase 1 signaling pathway. Our study provides novel insights into a potential role for SerpinA3N in APAP-induced liver injury. SIGNIFICANCE STATEMENT: Our studies indicate that serine (or cysteine) peptidase inhibitor, clade A, member 3N (SerpinA3N) may have a pathophysiological role in modulating acetaminophen (APAP)-induced liver injury. More specifically, mice with hepatic deletion of SerpinA3N suppressed inflammation and liver injury to reduce APAP-induced hepatotoxicity. Controlling the inflammatory response offers possible approaches for novel therapeutics; therefore, understanding the pathophysiological role of SerpinA3N in inducing liver injury may add to the development of more efficacious treatments.


Subject(s)
Acetaminophen/toxicity , Acute-Phase Proteins/deficiency , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Serpins/deficiency , Acute-Phase Proteins/genetics , Animals , Chemical and Drug Induced Liver Injury/genetics , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Serpins/genetics
2.
J Gastroenterol Hepatol ; 32(3): 708-715, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27404046

ABSTRACT

BACKGROUND AND AIM: It has been suggested in several studies that an increased translocation of bacterial lipopolysaccharide (LPS) and, subsequently, an activation of toll-like receptor (TLR)-dependent signaling pathways in the liver may contribute to the development of non-alcoholic fatty liver disease. METHODS: Eight-week-old lipopolysaccharide-binding protein (LBP)-/- and wild-type (WT) mice were pair fed either a liquid diet rich in fat, fructose, and cholesterol (Western-style diet [WSD]) or a control liquid diet for 8 weeks. Parameters of liver injury, markers of TLR-4-dependent signaling pathway, and glucose/lipid metabolism were determined. RESULTS: Despite similar total caloric intake, weight gain, fasting blood glucose levels, and liver-to-bodyweight ratio, indices of liver damage determined by liver histology and transaminases were markedly lower in WSD-fed LBP-/- mice than in WSD-fed WT animals. In line with these findings, number of neutrophils, F4/80 positive cells, and plasminogen activator inhibitor 1 were only found to be significantly increased in livers of WSD-fed WT mice. While mRNA expressions of TLR-4 and myeloid differentiation primary response 88 were similar between WSD-fed groups, concentrations of inducible nitric oxide synthase protein and 4-hydroxynonenal protein adducts were significantly higher in livers of WSD-fed WT mice than in WSD-fed LBP-/- animals. Markers of lipid metabolism, for example, sterol regulatory element-binding protein 1c and fatty acid synthase per se, were significantly lower in livers of LBP-/- mice; however, mRNA expressions did not differ between controls and WSD-fed mice within the respective mouse strain. CONCLUSION: Taken together, our results suggest that LBP is a critical factor in the development of non-alcoholic fatty liver disease in mice.


Subject(s)
Acute-Phase Proteins/deficiency , Acute-Phase Proteins/physiology , Carrier Proteins/physiology , Lipopolysaccharides/metabolism , Liver/metabolism , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/physiology , Non-alcoholic Fatty Liver Disease/etiology , Animals , Disease Models, Animal , Glucose/metabolism , Lipid Metabolism , Mice, Inbred BALB C , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Signal Transduction/physiology , Toll-Like Receptors/metabolism
3.
Hypertension ; 66(1): 158-66, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25987661

ABSTRACT

Activation of the mineralocorticoid receptor has been shown to be deleterious in cardiovascular diseases (CVDs). We have recently shown that lipocalin 2 (Lcn2), or neutrophil gelatinase-associated lipocalin (NGAL), is a primary target of aldosterone/mineralocorticoid receptor in the cardiovascular system. Lcn2 is a circulating protein, which binds matrix metalloproteinase 9 and modulates its stability. We hypothesized that Lcn2 could be a mediator of aldosterone/mineralocorticoid receptor profibrotic effects in the cardiovascular system. Correlations between aldosterone and profibrotic markers, such as procollagen type I N-terminal peptide, were investigated in healthy subjects and subjects with abdominal obesity. The implication of Lcn2 in the mineralocorticoid pathway was studied using Lcn2 knockout mice subjected to a nephrectomy/aldosterone/salt (NAS) challenge for 4 weeks. In human subjects, NGAL/matrix metalloproteinase 9 was positively correlated with plasma aldosterone and fibrosis biomarkers. In mice, loss of Lcn2 prevented the NAS-induced increase of plasma procollagen type I N-terminal peptide, as well as the increase of collagen fibers deposition and collagen I expression in the coronary vessels and the aorta. The lack of Lcn2 also blunted the NAS-induced increase in systolic blood pressure. Ex vivo, treatment of human fibroblasts with recombinant Lcn2 induced the expression of collagen I and the profibrotic galectin-3 and cardiotrophin-1 molecules. Our results showed that Lcn2 plays a key role in aldosterone/mineralocorticoid receptor-mediated vascular fibrosis. The clinical data indicate that this may translate in human patients. Lcn2 is, therefore, a new biotarget in cardiovascular fibrosis induced by mineralocorticoid activation.


Subject(s)
Acute-Phase Proteins/physiology , Aldosterone/toxicity , Lipocalins/physiology , Obesity, Abdominal/physiopathology , Oncogene Proteins/physiology , Proto-Oncogene Proteins/physiology , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Acute-Phase Proteins/pharmacology , Aldosterone/blood , Animals , Aorta/drug effects , Aorta/pathology , Cardiomyopathy, Hypertrophic/chemically induced , Cardiomyopathy, Hypertrophic/physiopathology , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Female , Fibroblasts , Fibrosis , Galectin 3/biosynthesis , Galectin 3/blood , Galectin 3/genetics , Humans , Hypertension/physiopathology , Hypertrophy , Kidney/pathology , Lipocalin-2 , Lipocalins/blood , Lipocalins/genetics , Lipocalins/pharmacology , Male , Mice , Myocardium/cytology , Myocardium/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/physiology , Nephrectomy/adverse effects , Obesity, Abdominal/blood , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Peptide Fragments/blood , Procollagen/blood , Proto-Oncogene Proteins/blood , Proto-Oncogene Proteins/pharmacology , Rats , Recombinant Proteins/pharmacology
4.
Liver Int ; 35(4): 1195-202, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25040147

ABSTRACT

BACKGROUND & AIMS: Various immune mediators such as interleukin-6 (IL-6) have been implicated in the process of liver regeneration. Lipocalin-2 (LCN2) has been recently characterized as a prototypic immune mediator produced by various cell types being involved mainly in host defence. In addition, numerous studies have demonstrated its clinical value as a biomarker. This study aimed at defining the role of LCN2 in liver regeneration. METHODS: We studied LCN2 expression in wild-type mice in a model of partial hepatectomy (PH). Furthermore, we evaluated liver regeneration after PH in LCN-deficient mice compared to littermate controls. Serum levels of LCN2 were assessed in a small group of patients undergoing hepatic resection. RESULTS: LCN2 is dramatically induced in livers and sera of wild-type mice after PH, whereas liver LCN2-receptor expression was decreased. Sham operations did not affect hepatic and serum LCN2 expression. Although LCN2-deficient mice exhibited increased baseline liver expression indices, LCN2-deficient mice did not differ from wild-type mice with respect to hepatic proliferation suggesting that this molecule is not involved in hepatic repair. Only serum IL-1ß levels were slightly lower in LCN(-/-) mice, whereas IL-6 serum levels did not differ between various tested animal groups. In humans undergoing hepatic resection, LCN2 levels increased significantly within 24 h following surgery. CONCLUSIONS: LCN2, although massively induced in mice after PH, is not relevant in murine hepatic regeneration. Further, human studies have to define whether LCN2 could evolve as biomarker after liver surgery.


Subject(s)
Acute-Phase Proteins/metabolism , Lipocalins/blood , Lipocalins/metabolism , Liver Regeneration , Liver/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/blood , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Adult , Aged , Animals , Biomarkers/blood , Female , Hepatectomy/methods , Heterozygote , Homozygote , Humans , Interleukin-6/blood , Lipocalin-2 , Lipocalins/genetics , Liver/physiopathology , Liver/surgery , Male , Mice, Knockout , Middle Aged , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Phenotype , Signal Transduction , Time Factors , Up-Regulation
5.
PLoS One ; 9(7): e101881, 2014.
Article in English | MEDLINE | ID: mdl-25010215

ABSTRACT

Lipocalin 2 (LCN2), which is highly expressed by dendritic cells (DCs) when treated with dexamethasone (Dex) and lipopolysaccharide (LPS), plays a key role in the defence against bacteria and is also involved in the autocrine apoptosis of T-cells. However, the function of LCN2 when secreted by DCs is unknown: this is a critical gap in our understanding of the regulation of innate and adaptive immune systems. Tolerance, stimulation and suppression are functions of DCs that facilitate the fine-tuning of the immune responses and which are possibly influenced by LCN2 secretion. We therefore examined the role of LCN2 in DC/T-cell interaction. WT or Lcn2-/- bone marrow-derived DCs were stimulated with LPS or LPS+IFN-γ with and without Dex and subsequently co-cultured with T-cells from ovalbumin-specific TCR transgenic (OT-I and OT-II) mice. We found that CD8+ T-cell apoptosis was highly reduced when Lcn2-/- DCs were compared with WT. An in vivo CTL assay, using LPS-treated DCs, showed diminished killing ability in mice that had received Lcn2-/- DCs compared with WT DCs. As a consequence, we analysed T-cell proliferation and found that LCN2 participates in T-cell-priming in a dose-dependent manner and promotes a TH1 microenvironment. DC-secreted LCN2, whose function has previously been unknown, may in fact have an important role in regulating the balance between TH1 and TH2. Our results yield insights into DC-secreted LCN2 activity, which could play a pivotal role in cellular immune therapy and in regulating immune responses.


Subject(s)
Acute-Phase Proteins/metabolism , Apoptosis/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/metabolism , Lipocalins/metabolism , Oncogene Proteins/metabolism , Phenotype , Th1 Cells/cytology , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Animals , Apoptosis/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cellular Microenvironment/drug effects , Cellular Microenvironment/immunology , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Immunization , Interferon-gamma/pharmacology , Lipocalin-2 , Lipocalins/genetics , Lipopolysaccharides/pharmacology , Mice , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Th1 Cells/drug effects , Th1 Cells/immunology
6.
J Clin Invest ; 124(7): 2963-76, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24937428

ABSTRACT

α-Intercalated cells (A-ICs) within the collecting duct of the kidney are critical for acid-base homeostasis. Here, we have shown that A-ICs also serve as both sentinels and effectors in the defense against urinary infections. In a murine urinary tract infection model, A-ICs bound uropathogenic E. coli and responded by acidifying the urine and secreting the bacteriostatic protein lipocalin 2 (LCN2; also known as NGAL). A-IC-dependent LCN2 secretion required TLR4, as mice expressing an LPS-insensitive form of TLR4 expressed reduced levels of LCN2. The presence of LCN2 in urine was both necessary and sufficient to control the urinary tract infection through iron sequestration, even in the harsh condition of urine acidification. In mice lacking A-ICs, both urinary LCN2 and urinary acidification were reduced, and consequently bacterial clearance was limited. Together these results indicate that A-ICs, which are known to regulate acid-base metabolism, are also critical for urinary defense against pathogenic bacteria. They respond to both cystitis and pyelonephritis by delivering bacteriostatic chemical agents to the lower urinary system.


Subject(s)
Acute-Phase Proteins/urine , Escherichia coli Infections/prevention & control , Kidney Tubules, Collecting/metabolism , Lipocalins/urine , Oncogene Proteins/urine , Proto-Oncogene Proteins/urine , Urinary Tract Infections/prevention & control , Uropathogenic Escherichia coli , Acid-Base Equilibrium , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Animals , Disease Models, Animal , Escherichia coli Infections/microbiology , Escherichia coli Infections/urine , Female , Humans , Hydrogen-Ion Concentration , Iron/metabolism , Kidney Tubules, Collecting/pathology , Lipocalin-2 , Lipocalins/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Toll-Like Receptor 4/metabolism , Urinary Tract Infections/microbiology , Urinary Tract Infections/urine
7.
Stroke ; 45(7): 2141-3, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24893611

ABSTRACT

BACKGROUND AND PURPOSE: White matter injury occurs after subarachnoid hemorrhage (SAH) and has not been well studied. In this study, we investigated acute white matter injury in a mouse SAH model and the role of lipocalin 2 (LCN2) in that injury. METHODS: SAH was induced by endovascular perforation in wild-type (WT) or LCN2 knockout (LCN2-/-) mice. Sham WT mice underwent the same procedure without perforation. MRI was performed 24 hours after SAH and the volumes of the T2-hyperintensity in white matter were measured. Immunohistochemistry was performed to determine white matter injury. RESULTS: Mortality rates and SAH severity were not significantly different between WT and LCN2-/- animals. T2-hyperintensity in the white matter was observed in all WT animals at 24 hours after SAH (6.1±2.7 versus 0.06±0.07 mm3 in sham; P<0.001), and the volume of T2-hyperintensity tended to correlate with SAH severity (r=0.30; P=0.055). In WT animals with SAH, numerous LCN2-positive cells were observed in white matter. In contrast, LCN2-/- animals scarcely developed white matter T2-hyperintensity after SAH (0.5±0.5 mm3; P<0.001, versus WT). Markers of axonal damage and myelin degradation were increased in white matter after SAH in WT compared with those in LCN2-/- animals (P<0.05). CONCLUSIONS: SAH results in an acute white matter injury at 24 hours in mice, and LCN2 plays an important role in SAH-induced white matter injury.


Subject(s)
Acute-Phase Proteins/physiology , Leukoencephalopathies/physiopathology , Lipocalins/physiology , Oncogene Proteins/physiology , Subarachnoid Hemorrhage/genetics , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Animals , Disease Models, Animal , Female , Leukoencephalopathies/genetics , Leukoencephalopathies/pathology , Lipocalin-2 , Lipocalins/genetics , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Subarachnoid Hemorrhage/complications , Subarachnoid Hemorrhage/physiopathology , Time Factors
8.
J Biol Chem ; 289(32): 22063-77, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24917675

ABSTRACT

In this study, we report that lipocalin 2 (Lcn2), a recently characterized adipokine/cytokine, is a novel regulator of brown adipose tissue (BAT) activation by modulating the adrenergic independent p38 MAPK-PGC-1α-UCP1 pathway. Global Lcn2 knock-out (Lcn2(-/-)) mice have defective BAT thermogenic activation caused by cold stimulation and decreased BAT activity under high fat diet-induced obesity. Nevertheless, Lcn2(-/-) mice maintain normal sympathetic nervous system activation as evidenced by normal catecholamine release and lipolytic activity in response to cold stimulation. Further studies showed that Lcn2 deficiency impairs peroxisomal and mitochondrial oxidation of lipids and attenuates cold-induced Pgc1a and Ucp1 expression and p38 MAPK phosphorylation in BAT. Moreover, in vitro studies showed that Lcn2 deficiency reduces the thermogenic activity of brown adipocytes. Lcn2(-/-) differentiated brown adipocytes have significantly decreased expression levels of brown fat markers, decreased p38 MAPK phosphorylation, and decreased mitochondrial oxidation capacity. However, Lcn2(-/-) brown adipocytes have normal norepinephrine-stimulated p38 MAPK and hormone-sensitive lipase phosphorylation and Pgc1a and Ucp1 expression, suggesting an intact ß-adrenergic signaling activation. More intriguingly, recombinant Lcn2 was able to significantly stimulate p38 MAPK phosphorylation in brown adipocytes. Activating peroxisome proliferator-activated receptor γ, a downstream effector of PGC-1α, by thiazolidinedione administration fully reverses the BAT function of Lcn2(-/-) mice. Our findings provide evidence for the novel role Lcn2 plays in oxidative metabolism and BAT activation via an adrenergic independent mechanism.


Subject(s)
Acute-Phase Proteins/metabolism , Adipose Tissue, Brown/metabolism , Lipocalins/metabolism , Oncogene Proteins/metabolism , Thermogenesis/physiology , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Adipocytes, Brown/metabolism , Animals , Catecholamines/metabolism , Gene Expression , Ion Channels/genetics , Ion Channels/metabolism , Lipid Metabolism , Lipocalin-2 , Lipocalins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Oxidation-Reduction , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Peroxisomes/metabolism , Phosphorylation , Thermogenesis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Uncoupling Protein 1 , p38 Mitogen-Activated Protein Kinases/metabolism
9.
J Cereb Blood Flow Metab ; 34(8): 1306-14, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24780901

ABSTRACT

Lipocalin-2 (LCN2) is a secreted protein of the lipocalin family, but little is known about the expression or the role of LCN2 in the central nervous system. Here, we investigated the role of LCN2 in ischemic stroke using a rodent model of transient cerebral ischemia. Lipocalin-2 expression was highly induced in the ischemic brain and peaked at 24 hours after reperfusion. After transient middle cerebral artery occlusion, LCN2 was predominantly expressed in astrocytes and endothelial cells, whereas its receptor (24p3R) was mainly detected in neurons, astrocytes, and endothelial cells. Brain infarct volumes, neurologic scores, blood-brain barrier (BBB) permeabilities, glial activation, and inflammatory mediator expression were significantly lower in LCN2-deficient mice than in wild-type animals. Lipocalin-2 deficiency also attenuated glial neurotoxicity in astrocyte/neuron cocultures after oxygen-glucose deprivation. Our results indicate LCN2 has a critical role in brain injury after ischemia/reperfusion, and that LCN2 may contribute to neuronal cell death in the ischemic brain by promoting neurotoxic glial activation, neuroinflammation, and BBB disruption.


Subject(s)
Acute-Phase Proteins/deficiency , Infarction, Middle Cerebral Artery/metabolism , Ischemic Attack, Transient/metabolism , Neutrophil Infiltration/immunology , Oncogene Proteins/deficiency , Reperfusion Injury/metabolism , Acute-Phase Proteins/genetics , Animals , Astrocytes/immunology , Astrocytes/metabolism , Astrocytes/pathology , Cell Proliferation , Cell Survival , Coculture Techniques , Culture Media , Disease Models, Animal , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/immunology , Ischemic Attack, Transient/pathology , Lipocalin-2 , Lipocalins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroglia/immunology , Neuroglia/pathology , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Oncogene Proteins/genetics , Reperfusion Injury/etiology , Reperfusion Injury/immunology , Reperfusion Injury/pathology
10.
J Am Heart Assoc ; 3(2): e000837, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24721803

ABSTRACT

BACKGROUND: Lipocalin-2 is a proinflammatory adipokine upregulated in obese humans and animals. A pathogenic role of lipocalin-2 in hypertension has been suggested. Mice lacking lipocalin-2 are protected from dietary obesity-induced cardiovascular dysfunctions. Administration of lipocalin-2 causes abnormal vasodilator responses in mice on a high-fat diet (HFD). METHODS AND RESULTS: Wild-type and lipocalin-2 knockout mice were fed with standard chow or HFD. Immunoassays were performed for evaluating the circulating and tissue contents of lipocalin-2. The relaxation and contraction of arteries were studied using a wire myograph. Blood pressure was monitored with implantable radio telemetry. Dietary obesity promoted the accumulation of lipocalin-2 protein in blood and arteries. Deficiency of this adipokine protected mice from dietary obesity-induced elevation of blood pressure. Mass spectrometry analysis revealed that human and murine lipocalin-2 were modified by polyamination. Polyaminated lipocalin-2 was rapidly cleared from the circulation. Adipose tissue was a major site for lipocalin-2 deamidation. The circulating levels and the arterial accumulation of deamidated lipocalin-2 were significantly enhanced by treatment with linoleic acid (18:2n-6), which bound to lipocalin-2 with high affinity and prevented its interactions with matrix metalloproteinase 9 (MMP9). Combined administration of linoleic acid with lipocalin-2 caused vascular inflammation and endothelial dysfunction and raised the blood pressure of mice receiving standard chow. A human lipocalin-2 mutant with cysteine 87 replaced by alanine (C87A) contained less polyamines and exhibited a reduced capacity to form heterodimeric complexes with MMP9. After treatment, C87A remained in the circulation for a prolonged period of time and evoked endothelial dysfunction in the absence of linoleic acid. CONCLUSIONS: Polyamination facilitates the clearance of lipocalin-2, whereas the accumulation of deamidated lipocalin-2 in arteries causes vascular inflammation, endothelial dysfunction, and hypertension.


Subject(s)
Acute-Phase Proteins/metabolism , Aorta/metabolism , Diet, High-Fat , Endothelium, Vascular/physiopathology , Hypertension/etiology , Lipocalins/metabolism , Obesity/complications , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Vasodilation , Acute-Phase Proteins/administration & dosage , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Adipose Tissue/metabolism , Animals , Aorta/physiopathology , Blood Pressure , Deamination , Disease Models, Animal , Endothelium, Vascular/metabolism , Humans , Hypertension/genetics , Hypertension/metabolism , Hypertension/physiopathology , Hypertension/prevention & control , Linoleic Acid/administration & dosage , Linoleic Acid/metabolism , Lipocalin-2 , Lipocalins/administration & dosage , Lipocalins/genetics , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Obesity/physiopathology , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Proto-Oncogene Proteins/administration & dosage , Proto-Oncogene Proteins/genetics , Time Factors
11.
J Immunol ; 191(10): 5204-19, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24089194

ABSTRACT

Astrocytes provide structural and functional support for neurons, as well as display neurotoxic or neuroprotective phenotypes depending upon the presence of an immune or inflammatory microenvironment. This study was undertaken to characterize multiple phenotypes of activated astrocytes and to investigate the regulatory mechanisms involved. We report that activated astrocytes in culture exhibit two functional phenotypes with respect to pro- or anti-inflammatory gene expression, glial fibrillary acidic protein expression, and neurotoxic or neuroprotective activities. The two distinct functional phenotypes of astrocytes were also demonstrated in a mouse neuroinflammation model, which showed pro- or anti-inflammatory gene expression in astrocytes following challenge with classical or alternative activation stimuli; similar results were obtained in the absence of microglia. Subsequent studies involving recombinant lipocalin-2 (LCN2) protein treatment or Lcn2-deficient mice indicated that the pro- or anti-inflammatory functionally polarized phenotypes of astrocytes and their intracellular signaling pathway were critically regulated by LCN2 under in vitro and in vivo conditions. Astrocyte-derived LCN2 promoted classical proinflammatory activation of astrocytes but inhibited IL-4-STAT6 signaling, a canonical pathway involved in alternative anti-inflammatory activation. Our results suggest that the secreted protein LCN2 is an autocrine modulator of the functional polarization of astrocytes in the presence of immune or inflammatory stimuli and that LCN2 could be targeted therapeutically to dampen proinflammatory astrocytic activation and related pathologies in the CNS.


Subject(s)
Acute-Phase Proteins/metabolism , Astrocytes/metabolism , Brain/immunology , Lipocalins/metabolism , Oncogene Proteins/metabolism , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Animals , Astrocytes/cytology , Astrocytes/immunology , Cell Polarity , Cells, Cultured , Inflammation/immunology , Interleukin-4/metabolism , Lipocalin-2 , Lipocalins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , STAT6 Transcription Factor/metabolism , Signal Transduction/immunology
12.
PLoS One ; 8(7): e67693, 2013.
Article in English | MEDLINE | ID: mdl-23861783

ABSTRACT

Lipocalin-2 (Lcn-2) is involved in divergent processes such as acute kidney injury or bacterial host defence. Our study was designed to evaluate the functional role of Lcn-2 in nephrotoxic serum nephritis (NTS). Since Lcn-2 is expressed in tubular epithelial cells as well as in cells of innate immunity such as macrophages and polymorphonuclear neutrophils (PMN), we induced NTS in wild-type (WT), Lcn-2 knock-out (KO) mice and WT/Lcn-2 KO chimeras. Mice lacking Lcn-2 exhibited more glomerular damage with increased proteinuria and interstitial leukocyte accumulation compared to WT mice. Chimeras able to express Lcn-2 in macrophages and PMN but not in epithelial cells were found to develop NTS comparable to wild-type controls. In contrast, chimeras expressing Lcn-2 in tubular epithelial cells with no expression in innate immune cells developed increased NTS due to decreased concerted apoptosis but increased necrosis and formation of damage-associated molecular patterns (DAMPs) such as high-mobility group box 1 (HMGB-1) in the kidney. In vivo blockade of HMGB-1, a toll-like receptor (TLR)-2 agonist, significantly reduced inflammation and NTS in Lcn-2 knock-out mice. In parallel, TLR-2 signalling was found to drive Lcn-2 transcription in vitro. Taken together, Lcn-2 expressed in innate immune cells is protective in NTS by inducing concerted apoptosis and inhibiting the formation of HMGB-1 thereby limiting cytokine production via TLR-2 signalling. In parallel, TLR-2 dependent transcription of Lcn-2 is an endogenous inhibitor of inflammation in NTS.


Subject(s)
Acute-Phase Proteins/genetics , Kidney Glomerulus/metabolism , Lipocalins/genetics , Macrophages/metabolism , Nephritis/genetics , Neutrophils/metabolism , Oncogene Proteins/genetics , Proteinuria/genetics , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/immunology , Animals , Apoptosis , Cytokines/biosynthesis , Cytokines/immunology , Female , Gene Expression Regulation , HMGB1 Protein/genetics , HMGB1 Protein/immunology , Immunity, Innate , Inflammation , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Lipocalin-2 , Lipocalins/immunology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Knockout , Nephritis/immunology , Nephritis/metabolism , Nephritis/pathology , Neutrophils/immunology , Neutrophils/pathology , Oncogene Proteins/deficiency , Oncogene Proteins/immunology , Proteinuria/immunology , Proteinuria/metabolism , Proteinuria/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Signal Transduction , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology
13.
J Clin Invest ; 123(8): 3363-72, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23863624

ABSTRACT

Macrophages play a key role in responding to pathogens and initiate an inflammatory response to combat microbe multiplication. Deactivation of macrophages facilitates resolution of the inflammatory response. Deactivated macrophages are characterized by an immunosuppressive phenotype, but the lack of unique markers that can reliably identify these cells explains the poorly defined biological role of this macrophage subset. We identified lipocalin 2 (LCN2) as both a marker of deactivated macrophages and a macrophage deactivator. We show that LCN2 attenuated the early inflammatory response and impaired bacterial clearance, leading to impaired survival of mice suffering from pneumococcal pneumonia. LCN2 induced IL-10 formation by macrophages, skewing macrophage polarization in a STAT3-dependent manner. Pulmonary LCN2 levels were tremendously elevated during bacterial pneumonia in humans, and high LCN2 levels were indicative of a detrimental outcome from pneumonia with Gram-positive bacteria. Our data emphasize the importance of macrophage deactivation for the outcome of pneumococcal infections and highlight the role of LCN2 and IL-10 as determinants of macrophage performance in the respiratory tract.


Subject(s)
Acute-Phase Proteins/immunology , Lipocalins/immunology , Macrophages, Alveolar/immunology , Oncogene Proteins/immunology , Pneumonia, Pneumococcal/immunology , Proto-Oncogene Proteins/immunology , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Adult , Aged , Animals , Female , Humans , Immune Tolerance , Interleukin-10/biosynthesis , Lipocalin-2 , Lipocalins/genetics , Lung/immunology , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Pneumonia, Pneumococcal/etiology , Transplantation Chimera/immunology
14.
Prostate ; 73(12): 1281-90, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23775308

ABSTRACT

BACKGROUND: Lipocalin-2 (LCN2) is a member of the lipocalin superfamily, and it has an important role in the regulation of cellular oncogenesis and apoptosis. However, the role for LCN2 in prostate cancer remains unclear. METHOD: LCN2 expression has been determined by Western blotting, qRT-PCR, and immunohistochemistry in the human prostate cell lines PC3, DU145, LNCaP, and 22Rv, and in human prostate tissue array. In this study, we identified shRNA-LCN2 to determine the role of LCN2 in prostate-cancer cell proliferation, migration, and invasion. Cell proliferative ability was measured by MTT, colony-formation, and cell-cycle analysis. The role of LCN2 in prostate-cancer cell migration and invasion was analyzed by cell-migration assay and Matrigel invasion assay. The effect of LCN2 knockdown on prostate tumor growth was assessed in a subcutaneous xenograft model. RESULTS: LCN2 protein and mRNA expression are higher in PC3 and DU145 cells than in LNCaP and 22Rv cells, and prostate cancer tissue correlated significantly with tumor differentiation (P < 0.017) and Gleason's grade (P < 0.02). LCN2 knockdown in PC3 and DU145 cells decreased cell proliferation, colony formation, cell cycle arrest, migration, and invasion. Conversely, LCN2 overexpression in 22Rv cells produced the opposite effect. Subcutaneous xenografts in mice models showed decreased tumor growth in the LCN2-knockdown mice. CONCLUSIONS: Our results suggest that LCN2 might play an important role in regulation of proliferation and invasion of human prostate cancer, and that it can be a valuable marker of prostate cancer progression.


Subject(s)
Acute-Phase Proteins/deficiency , Cell Proliferation , Down-Regulation , Gene Knockdown Techniques , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins/deficiency , Acute-Phase Proteins/genetics , Aged , Animals , Cell Line, Tumor , Down-Regulation/genetics , Gene Knockdown Techniques/methods , Humans , Lipocalin-2 , Lipocalins/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins/genetics
15.
Immunobiology ; 218(7): 969-78, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23317919

ABSTRACT

Neutrophil gelatinase-associated lipocalin (NGAL/Lipocalin-2/Lcn-2) is a 25kDa protein which is involved in host defence against certain Gram negative bacteria upon binding of iron loaded bacterial siderophores thereby limiting the availability of this essential nutrient to bacteria resulting in inhibition of their growth and pathogenicity. As iron is important for the growth of the intracellular bacterium Chlamydia pneumoniae we questioned whether Lcn-2 affects the course of this infection. We employed primary peritoneal macrophages obtained from wildtype and Lcn-2 -/- mice and RAW 264.7 cells which were infected with C. pneumoniae. In addition, we studied C. pneumoniae multiplication in vivo in mice receiving diets with varying iron contents. We analyzed C. pneumoniae numbers by immunohistochemistry and RT-PCR and studied the expression of iron metabolism and cytokine genes by RT-PCR, Western blot or ELISA. Infection with Chlamydiae ex vivo and in vivo revealed a significantly higher bacterial growth in peritoneal macrophages of Lcn-2 -/- than of wildtype mice. These differences were significantly more pronounced upon iron challenge, which stimulated bacterial growth. Accordingly, treatment with an anti-Lnc-2 antibody increased whereas addition of recombinant Lcn-2 reduced bacterial growth in infected macrophages. When investigating the underlying mechanisms we observed partly different expression of several iron metabolism genes between Lcn-2 +/+ and Lcn-2 -/- macrophages and most strikingly an increased formation of the anti-inflammatory cytokine IL-10 by Lcn-2 -/- macrophages. Upon treatment with an anti-IL10 antibody we experienced a significant increase of Chlamydial growth within Lcn-2 -/- macrophages along with a reduction of the major iron storage protein ferritin. Herein we provide first time evidence that Lcn-2 is involved in host defence against Chlamydia presumably by limiting the availability of iron to the pathogen. In the absence of Lcn-2, increased formation of IL-10 exerts protective effects by increasing the intracellular formation of ferritin, thereby reducing the access of iron for bacteria.


Subject(s)
Acute-Phase Proteins/immunology , Chlamydophila pneumoniae/metabolism , Ferritins/immunology , Interleukin-10/immunology , Iron/metabolism , Lipocalins/immunology , Macrophages, Peritoneal/immunology , Oncogene Proteins/immunology , Acute-Phase Proteins/antagonists & inhibitors , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Animals , Antibodies/pharmacology , Bacterial Load , Cell Line , Chlamydophila pneumoniae/growth & development , Chlamydophila pneumoniae/immunology , Female , Ferritins/genetics , Gene Expression Regulation/drug effects , Homeostasis , Host-Pathogen Interactions , Interleukin-10/genetics , Iron/pharmacology , Iron, Dietary/administration & dosage , Lipocalin-2 , Lipocalins/antagonists & inhibitors , Lipocalins/genetics , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/microbiology , Mice , Mice, Inbred C57BL , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Primary Cell Culture
16.
Oncogene ; 32(10): 1233-9, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-22614012

ABSTRACT

Evidence that lipocalin 2 (LCN2) is oncogenic has grown in recent years and comes from both animal models and expression analysis from a variety of human cancers. In the intestine, LCN2 is overexpressed in colitis patients and its overexpression is a negative prognostic indicator in colorectal cancer. Functionally, LCN2 has a number of different activities that may contribute to its oncogenic potential, including increasing matrix metalloproteinase activity, control of iron availability and stimulating inflammation. In this report, we examined APCmin intestinal tumorigenesis in an LCN2-deficient background. We found that the loss of LCN2 increased tumor multiplicity specifically in the duodenum, suggesting a potential tumor-suppressive activity. Concurrently, however, LCN2 increased the average small intestinal tumor size particularly in the distal small intestine. We found that this increase was correlated to tumor iron(II) content, suggesting that an iron-scavenging role is important for LCN2 oncogenic activity in the intestine.


Subject(s)
Acute-Phase Proteins/biosynthesis , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Lipocalins/biosynthesis , Oncogene Proteins/biosynthesis , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Animals , Apoptosis/physiology , Disease Progression , Female , Genes, APC , Intestinal Neoplasms/genetics , Lipocalin-2 , Lipocalins/genetics , Male , Mice , Mice, Inbred C57BL , Oncogene Proteins/deficiency , Oncogene Proteins/genetics
17.
mBio ; 3(6)2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23169997

ABSTRACT

UNLABELLED: Pathogenic bacteria require iron for replication within their host. Klebsiella pneumoniae and other Gram-negative pathogens produce the prototypical siderophore enterobactin (Ent) to scavenge iron in vivo. In response, mucosal surfaces secrete lipocalin 2 (Lcn2), an innate immune protein that binds Ent to disrupt bacterial iron acquisition and promote acute inflammation during colonization. A subset of K. pneumoniae isolates attempt to evade Lcn2 by producing glycosylated Ent (Gly-Ent, salmochelin) or the alternative siderophore yersiniabactin (Ybt). However, these siderophores are not functionally equivalent and differ in their abilities to promote growth in the upper respiratory tract, lungs, and serum. To understand how Lcn2 exploits functional differences between siderophores, isogenic mutants of an Ent(+) Gly-Ent(+) Ybt(+) K. pneumoniae strain were inoculated into Lcn2(+/+) and Lcn2(-/-) mice, and the pattern of pneumonia was examined. Lcn2 effectively protected against the iroA ybtS mutant (Ent(+) Gly-Ent(-) Ybt(-)). Lcn2(+/+) mice had small foci of pneumonia, whereas Lcn2(-/-) mice had many bacteria in the perivascular space. The entB mutant (Ent(-) Ybt(+) Gly-Ent(-)) caused moderate bronchopneumonia but did not invade the transferrin-containing perivascular space. Accordingly, transferrin blocked Ybt-dependent growth in vitro. The wild type and the iroA mutant, which both produce Ent and Ybt, had a mixed phenotype, causing a moderate bronchopneumonia in Lcn2(+/+) mice and perivascular overgrowth in Lcn2(-/-) mice. Together, these data indicate that Lcn2, in combination with transferrin, confines K. pneumoniae to the airways and prevents invasion into tissue containing the pulmonary vasculature. IMPORTANCE: Gram-negative bacteria are a common cause of severe hospital-acquired infections. To cause disease, they must obtain iron and secrete the small molecule enterobactin to do so. Animal models of pneumonia using Klebsiella pneumoniae indicate that enterobactin promotes severe disease. Accordingly, the host defense protein lipocalin 2 exploits this common target by binding enterobactin and disrupting its function. However, pathogenic bacteria often make additional siderophores that lipocalin 2 cannot bind, such as yersiniabactin, which could make this host defense ineffective. This work compares the pattern and severity of pneumonia caused by K. pneumoniae based on which siderophores it produces. The results indicate that enterobactin promotes growth around blood vessels that are rich in the iron-binding protein transferrin, but yersiniabactin does not. Together, transferrin and lipocalin 2 protect this space against all types of K. pneumoniae tested. Therefore, the ability to acquire iron determines where bacteria can grow in the lung.


Subject(s)
Acute-Phase Proteins/metabolism , Enterobactin/metabolism , Host-Pathogen Interactions , Klebsiella Infections/pathology , Klebsiella pneumoniae/pathogenicity , Lipocalins/metabolism , Oncogene Proteins/metabolism , Pneumonia, Bacterial/pathology , Transferrin/metabolism , Acute-Phase Proteins/deficiency , Animals , Enterobactin/antagonists & inhibitors , Enterobactin/genetics , Lipocalin-2 , Mice , Mice, Knockout , Oncogene Proteins/deficiency , Phenols/antagonists & inhibitors , Phenols/metabolism , Protein Binding , Thiazoles/antagonists & inhibitors , Thiazoles/metabolism , Virulence Factors/antagonists & inhibitors , Virulence Factors/genetics , Virulence Factors/metabolism
18.
PLoS One ; 7(11): e50052, 2012.
Article in English | MEDLINE | ID: mdl-23185529

ABSTRACT

Pulmonary tuberculosis (TB), caused by the intracellular bacteria Mycobacterium tuberculosis, is a worldwide disease that continues to kill more than 1.5 million people every year worldwide. The accumulation of lymphocytes mediates the formation of the tubercle granuloma in the lung and is crucial for host protection against M.tuberculosis infection. However, paradoxically the tubercle granuloma is also the basis for the immunopathology associated with the disease and very little is known about the regulatory mechanisms that constrain the inflammation associated with the granulomas. Lipocalin 2 (Lcn2) is a member of the lipocalin family of proteins and binds to bacterial siderophores thereby sequestering iron required for bacterial growth. Thus far, it is not known whether Lcn2 plays a role in the inflammatory response to mycobacterial pulmonary infections. In the present study, using models of acute and chronic mycobacterial pulmonary infections, we reveal a novel role for Lcn2 in constraining T cell lymphocytic accumulation and inflammation by inhibiting inflammatory chemokines, such as CXCL9. In contrast, Lcn2 promotes neutrophil recruitment during mycobacterial pulmonary infection, by inducing G-CSF and KC in alveolar macrophages. Importantly, despite a common role for Lcn2 in regulating chemokines during mycobacterial pulmonary infections, Lcn2 deficient mice are more susceptible to acute M.bovis BCG, but not low dose M.tuberculosis pulmonary infection.


Subject(s)
Acute-Phase Proteins/immunology , Granuloma, Respiratory Tract/veterinary , Lipocalins/immunology , Mycobacterium Infections/veterinary , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , Oncogene Proteins/immunology , Tuberculosis/veterinary , Acute Disease , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Animals , Cell Movement , Chemokine CXCL9/antagonists & inhibitors , Chemokine CXCL9/immunology , Chronic Disease , Gene Expression , Granuloma, Respiratory Tract/immunology , Granuloma, Respiratory Tract/metabolism , Granuloma, Respiratory Tract/microbiology , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Inflammation/veterinary , Iron/metabolism , Lipocalin-2 , Lipocalins/genetics , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium Infections/immunology , Mycobacterium Infections/metabolism , Mycobacterium Infections/microbiology , Neutrophils/immunology , Neutrophils/microbiology , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Siderophores/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , Tuberculosis/immunology , Tuberculosis/metabolism , Tuberculosis/microbiology
19.
J Immunol ; 189(4): 1911-9, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22786765

ABSTRACT

Various states of inflammation, including sepsis, are associated with hypoferremia, which limits iron availability to pathogens and reduces iron-mediated oxidative stress. Lipocalin 2 (Lcn2; siderocalin, 24p3) plays a central role in iron transport. Accordingly, Lcn2-deficient (Lcn2KO) mice exhibit elevated intracellular labile iron. In this study, we report that LPS induced systemic Lcn2 by 150-fold in wild-type mice at 24 h. Relative to wild-type littermates, Lcn2KO mice were markedly more sensitive to endotoxemia, exhibiting elevated indices of organ damage (transaminasemia, lactate dehydrogenase) and increased mortality. Such exacerbated endotoxemia was associated with substantially increased caspase-3 cleavage and concomitantly elevated immune cell apoptosis. Furthermore, cells from Lcn2KO mice were hyperresponsive to LPS ex vivo, exhibiting elevated cytokine secretion. Additionally, Lcn2KO mice exhibited delayed LPS-induced hypoferremia despite normal hepatic hepcidin expression and displayed decreased levels of the tissue redox state indicators cysteine and glutathione in liver and plasma. Desferroxamine, an iron chelator, significantly protects Lcn2KO mice from LPS-induced toxicity, including mortality, suggesting that Lcn2 may act as an antioxidant in vivo by regulating iron homeostasis. Thus, Lcn2-mediated regulation of labile iron protects the host against sepsis. Its small size and simple structure may make Lcn2 a deployable treatment for sepsis.


Subject(s)
Acute-Phase Proteins/metabolism , Homeostasis/physiology , Iron/metabolism , Lipocalins/metabolism , Oncogene Proteins/metabolism , Sepsis/metabolism , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/immunology , Animals , Apoptosis/physiology , Endotoxins/toxicity , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunoblotting , Lipocalin-2 , Lipocalins/immunology , Male , Mice , Mice, Knockout , Oncogene Proteins/deficiency , Oncogene Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction , Sepsis/chemically induced , Sepsis/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...