Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Pak J Pharm Sci ; 36(5): 1375-1379, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37869912

ABSTRACT

This study was designed to determine the effects of curcumin on some acute phase proteins in rats treated with aflatoxin B1. In this study, healthy 38 male Wistar Albino rats were used. The animals in control group were given food and distilled water. The animals in DMSO group were orally given 1 ml 10% DMSO daily for 60 days, animals in Cur group was orally given 300mg/kg curcumin dissolved in 10% DMSO daily for 60 days. Animals in AFB1 group were orally given 250µg/kg aflatoxin B1 dissolved in 10% DMSO daily for 60 days. Animals in AFB1+Cur group was orally given 250µg/kg aflatoxin B1 dissolved in 10% DMSO and 300mg/kg curcumin dissolved in 10% DMSO daily for 60 days. In blood samples taken from all animals, fibrinogen, prothrombin, albumin and CRP levels were determined. In this study, fibrinogen, prothrombin and CRP levels with AFB1 group were found to be significantly higher than the control group (p<0.05). In the AFB1+Cur group, fibrinogen and CRP levels were lower than in the aflatoxin group (p<0.05). In the study, it was observed that albumin level in rats in AFB1 group significantly decreased compared to the control group (p<0.05) and this difference was found to be eliminated depend on the application of curcumin together with aflatoxin B1 (p<0.05). In conclusion, our findings regarding the ameliorating effects of curcumin on acute phase protein abnormalities caused by aflatoxicosis will contribute to future research.


Subject(s)
Curcumin , Rats , Male , Animals , Curcumin/pharmacology , Rats, Wistar , Aflatoxin B1/toxicity , Acute-Phase Proteins/pharmacology , Prothrombin , Dimethyl Sulfoxide/pharmacology , Fibrinogen , Albumins , Liver
2.
Ocul Immunol Inflamm ; 29(3): 546-554, 2021 Apr 03.
Article in English | MEDLINE | ID: mdl-31738650

ABSTRACT

Purpose: The purpose of this study is to explore the effects of dihydrotestosterone (DHT) on lipopolysaccharide (LPS)-induced proinflammatory cytokine release in human ocular surface epithelial cells exposed to LPS and LPS-binding protein (LBP).Methods: Immortalized human corneal, conjunctival, and meibomian gland epithelial cells were cultured in keratinocyte-free medium. After confluency, they were exposed to a stratification medium Dulbecco's modified Eagle medium (DMEM)/F12 in the presence of fetal bovine serum and were exposed to vehicle, LPS + LBP, or DHT. Culture media were processed for multiplex-bead analysis of specific proinflammatory cytokines including interferon (IFN)-γ, tumor necrosis factor (TNF)-α, interleukin (IL)-2, IL-4, IL-8, IL-6, IL-10, IL-1ß, vascular endothelial growth factor (VEGF)-A. Cytokine concentrations were compared by analysis of variance with Tukey post hoc testing. p < 0.05 was considered statistically significant.Results: The results are LPS + LBP-induced the secretion of IFN-γ, IL-6, IL-10, IL-1ß, VEGF-A cytokines in corneal epithelial cells; TNF-α, IL-2, IL-8, IL-6, IL-1ß, VEGF-A cytokines in conjunctival epithelial cells; and IL-8, IL-6, IL-1ß, VEGF-A cytokines in meibomian gland epithelial cells. When these LPS + LBP-stimulated cells were exposed to DHT for 2 days, it was found that DHT suppressed the secretion of IL-6, IL-10, IL-1ß, VEGF-A cytokines in corneal epithelial cells; TNF-α, IL-6, IL-1ß, VEGF-A cytokines in conjunctival epithelial cells; and IL-6, IL-1ß, VEGF-A cytokines in meibomian gland epithelial cells.Conclusion: LPS + LBP is shown to induce the secretion of certain proinflammatory cytokines from ocular surface and adnexal epithelial cells. DHT showed anti-inflammatory activity by suppressing some of those cytokines in these cell lines.


Subject(s)
Androgens/pharmacology , Conjunctiva/cytology , Cytokines/metabolism , Dihydrotestosterone/pharmacology , Epithelial Cells/drug effects , Epithelium, Corneal/drug effects , Meibomian Glands/cytology , Acute-Phase Proteins/pharmacology , Carrier Proteins/pharmacology , Cell Line , Cell Survival , Cells, Cultured , Epithelial Cells/metabolism , Epithelium, Corneal/metabolism , Humans , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/pharmacology
3.
Probiotics Antimicrob Proteins ; 12(4): 1582-1593, 2020 12.
Article in English | MEDLINE | ID: mdl-32445120

ABSTRACT

Lipopolysaccharide (LPS) is a toxic and immunogenic agent for human. Additionally, LPS is a good target for some antimicrobial compounds, including antimicrobial peptides (AMPs). LPS-binding peptides (LBPs) can recognize and neutralize LPS. Rabbit and human cathelicidins are AMPs with LPS-binding activity. In this study, we designed and synthesized two new truncated LBPs from rabbit and human CAP18 peptides by in silico methods. After synthesis of peptides, the antimicrobial properties and LPS-binding activity of these peptides were evaluated. The parental rabbit and human CAP18 peptides were selected as positive controls. Next, the changes in the secondary structure of these peptides before and after treatment with LPS were measured by circular dichroism (CD). Human cytotoxicity of the peptides was evaluated by MTT and red blood cells (RBCs) hemolysis assays. Finally, field emission scanning electron microscopy (FE-SEM), confocal microscopy, and flow cytometry were performed to study the action mechanism of these peptides. Results indicated that the hCap18 and rCap18 had antibacterial activity (at a MIC of 4-128 µg/mL). The results of the quantitative LAL test demonstrated that LPS-binding activity of hCap18 peptide was better than rCap18, while rCap18 peptide had better antimicrobial properties. Furthermore, rCap18 had less cytotoxicity than hCap18. However, both peptides were nontoxic for normal human skin fibroblast cell in MIC range. In conclusion, rCap18 has good antibacterial properties, while hCap18 can be tested as a diagnostic molecule in our future studies.


Subject(s)
Acute-Phase Proteins/chemical synthesis , Anti-Bacterial Agents/chemical synthesis , Antimicrobial Cationic Peptides/chemical synthesis , Carrier Proteins/chemical synthesis , Drug Design , Lipopolysaccharides/antagonists & inhibitors , Membrane Glycoproteins/chemical synthesis , Acute-Phase Proteins/metabolism , Acute-Phase Proteins/pharmacology , Amino Acid Sequence , Animals , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/pharmacology , Bacillus subtilis/drug effects , Bacillus subtilis/growth & development , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Cell Line , Cell Survival/drug effects , Computer Simulation , Erythrocytes/cytology , Erythrocytes/drug effects , Escherichia coli/drug effects , Escherichia coli/growth & development , Fibroblasts/cytology , Fibroblasts/drug effects , Hemolysis/drug effects , Humans , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/pharmacology , Microbial Sensitivity Tests , Protein Engineering/methods , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Rabbits , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Structure-Activity Relationship
4.
PLoS One ; 13(3): e0192121, 2018.
Article in English | MEDLINE | ID: mdl-29494603

ABSTRACT

The thrombin-induced polymerisation of fibrinogen to form fibrin is well established as a late stage of blood clotting. It is known that Parkinson's Disease (PD) is accompanied by dysregulation in blood clotting, but it is less widely known as a coagulopathy. In recent work, we showed that the presence of tiny amounts of bacterial lipopolysaccharide (LPS) in healthy individuals could cause clots to adopt an amyloid form, and this could be observed via scanning electron microscopy (SEM) or via the fluorescence of thioflavin-T. This could be prevented by the prior addition of lipopolysaccharide-binding protein (LBP). We had also observed by SEM this unusual clotting in the blood of patients with Parkinson's Disease. We hypothesised, and here show, that this too can be prevented by LBP in the context of PD. This adds further evidence implicating inflammatory microbial cell wall products as an accompaniment to the disease, and may be part of its aetiology. This may lead to novel treatment strategies in PD designed to target microbes and their products.


Subject(s)
Acute-Phase Proteins/metabolism , Amyloid/antagonists & inhibitors , Amyloid/metabolism , Carrier Proteins/metabolism , Fibrin/metabolism , Membrane Glycoproteins/metabolism , Parkinson Disease/metabolism , Acute-Phase Proteins/pharmacology , Aged , Amyloid/ultrastructure , Blood Coagulation/drug effects , Carrier Proteins/pharmacology , Drug Discovery , Female , Fibrin/ultrastructure , Humans , Lipopolysaccharides/metabolism , Male , Membrane Glycoproteins/pharmacology , Middle Aged , Parkinson Disease/blood , Parkinson Disease/drug therapy , Parkinson Disease/pathology
5.
Article in English | MEDLINE | ID: mdl-28504094

ABSTRACT

Antimicrobial substances in serum include circulating complement proteins and acute phase proteins (APPs). We identified gene sequences for APPs, haptoglobin (Hp), C-reactive protein (CRP) and serum amyloid A (SAA) in marsupial genomes. Hp and SAA levels were measured in red-tailed phascogale (Phascogale calura) sera using commercially available assays. Hp levels were higher in males than females, while SAA levels suggest the phascogales used in this study were healthy. Serum was co-cultured with four bacterial species. Bacterial growth was inhibited after incubation at 37°C, however effectiveness differed with bacteria and incubation time. The least amount of bacterial growth was noticed after introduction to K. pneumoniae, and most when introduced to P. aeruginosa. Despite marsupials not having mature immune tissues at birth, and unable to mount specific immune responses, this study suggests other immune strategies, such as APPs in serum likely aid marsupials in their defence against pathogens.


Subject(s)
Anti-Bacterial Agents/blood , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Blood Bactericidal Activity , Complement System Proteins/pharmacology , Acute-Phase Proteins/analysis , Acute-Phase Proteins/genetics , Acute-Phase Proteins/pharmacology , Animals , Bacteria/growth & development , C-Reactive Protein/analysis , Haptoglobins/analysis , Haptoglobins/pharmacology , Klebsiella pneumoniae/drug effects , Marsupialia , Pseudomonas aeruginosa/drug effects , Serum/chemistry , Serum Amyloid A Protein/analysis
6.
Drug Dev Res ; 77(2): 94-102, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27009854

ABSTRACT

The aim of this study was to evaluate the anti-cancer effects of lipopolysaccharide binding protein (LBP) analogs derived from the marine resource Paralichthy olivaceus on MKN-28 gastric cancer cells. Five LBP analogs were used: ofLBP1N, ofLBP2A, ofLBP4N, ofLBP5A, and ofLBP6A. ofLBP6A induced cell death of MKN-28 cells at a concentration of 40 µM. While the anti-proliferation effects ofLBP6A showed on MKN-28 cells at concentration of 40 µM, it did not affect non-cancerous HEK-293 cells at the same concentration. The mechanism study showed that ofLBP6A lead to the inhibition of cell proliferation by apoptosis along with morphological changes. The phosphorylation of Fas associated death domain (FADD) as well as the expressions of cleaved caspase-8, -7, and -3 were increased by ofLBP6A treatment. Increased the expression level of cleaved caspase-3 was confirmed by immunofluorescence staining. The expressions of Bid, Bax, and cytochrome C were also increased by the treatment. However, the expressions of cellular FLICE (FADD-like IL-1ß-converting enzyme)-inhibitory protein (FLIP), Bcl-XL, and Bcl-2 were decreased by ofLBP6A treatment. The results of this study were the first to demonstrate the apoptotic anti-cancer effects of ofLBP6A, derived from P. olivavaceus on gastric cancer cells.


Subject(s)
Acute-Phase Proteins/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carrier Proteins/pharmacology , Membrane Glycoproteins/pharmacology , Peptides/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Fishes , Humans
7.
J Gastroenterol Hepatol ; 31(1): 145-54, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26189649

ABSTRACT

BACKGROUND AND AIM: Because neutrophil gelatinase-associated lipocalin (NGAL) is known to provide significant bacteriostatic effects during infectious conditions, we tested the hypothesis that this protein is up-regulated and secreted into the intraluminal cavity of the gut under critically ill conditions and is thus responsible for the regulation of bacterial overgrowth. METHODS: With our institutional approval, male C57BL/6J mouse (6-7 weeks) were enrolled and applied for lipopolysaccharide or peritonitis model compared with naïve control. We assessed NGAL protein concentrations in intestinal lumen and up-regulation of NGAL expression in intestinal tissues in in vivo as well as ex vivo settings. Simultaneously, we examined the effects of NGAL protein administration on the growth of Escherichia coli (E. coli) in in vivo and in vitro experimental settings. The localization of NGAL in intestinal tissues and lumen was also assessed by immunohistological approach using NGAL antibody. RESULTS: Both lipopolysaccharide and peritonitis insults evoked the marked up-regulation of NGAL mRNA and protein levels in gut tissues such as crypt cells. In addition, the administration of NGAL protein significantly inhibited the outgrowth of enteric E. coli under both in vitro and in vivo conditions, accompanied by histological evidence. CONCLUSION: Neutrophil gelatinase-associated lipocalin protein accompanied by apparent bacteriostatic action accumulated in the intestinal wall and streamed into the mucosal layer during critically ill state, thereby possibly shaping microbiota homeostasis in the gut.


Subject(s)
Acute-Phase Proteins/pharmacology , Acute-Phase Proteins/physiology , Intestines/microbiology , Lipocalins/pharmacology , Lipocalins/physiology , Microbiota/drug effects , Oncogene Proteins/pharmacology , Oncogene Proteins/physiology , Acute-Phase Proteins/genetics , Acute-Phase Proteins/metabolism , Animals , Critical Illness , Disease Models, Animal , Escherichia coli/growth & development , Gene Expression , Homeostasis/drug effects , Intestinal Mucosa/metabolism , Lipocalin-2 , Lipocalins/genetics , Lipocalins/metabolism , Lipopolysaccharides , Male , Mice, Inbred C57BL , Microbiota/physiology , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Peritonitis/microbiology , Up-Regulation
8.
Article in Chinese | MEDLINE | ID: mdl-26653647

ABSTRACT

OBJECTIVE: To explore beryllium oxide induced oxidative lung injury and the protective effects of LBP. METHODS: Intoxication of animals were induced by once intratracheal injection and LBP intervention by intragastric administration. The content of HIF-1, VEGF and HO-1 of lung tissues were measured by kits. The pathological changes of lung tissue were showed by pathological section. The changes of lung ultrastructure were observed by electron microscope. RESULTS: Pathological changes of the lung tissue in beryllium oxide exposure group rats were in line with the characteristics of beryllium disease in human. Compared with the control group, HO-1 was increased in beryllium oxide exposure 40 d group and low doses of LBP group, compared with the control group, HO-1 was increased in beryllium oxide exposure 80d group and LBP treatment groups (P < 0.05 or P < 0.01). Compared with the control group, HIF-1 was increased in beryllium oxide exposure 40 d group, LBP treatment groups, beryllium oxide exposure 60 d and 80 d groups (P < 0.05 or P < 0.01). Compared with the control group, VEGF was increased of all phases, especially in beryllium oxide exposure 40d and 80 groups, LBP treatment groups and beryllium oxide exposure 60 d (P < 0.05 or P < 0.01). The content of HO-1 of beryllium oxide exposure group was higher than the LBP treatment for 40d group but below LBP treatment for 80 d group (P < 0.05). The content of HIF1 of beryllium oxide exposure group was higher than high dose of LBP treatment for 60d group and LBP treatment for 80 d group (P < 0.01). The content of VEGF of beryllium oxide exposure group was higher than LBP treatment for 40 d group and high dose of LBP treatment for 60 d (P < 0.05 or P < 0.01). CONCLUSIONS: BeO can cause abnormal expression of related genes of lung tissue in rats, LBP has protective effects on BeO caused lung injury.


Subject(s)
Acute Lung Injury/physiopathology , Acute-Phase Proteins/pharmacology , Beryllium/toxicity , Carrier Proteins/pharmacology , Lung/pathology , Membrane Glycoproteins/pharmacology , Oxidative Stress , Acute Lung Injury/chemically induced , Animals , Heme Oxygenase (Decyclizing)/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/drug effects , Protective Agents/pharmacology , Rats , Vascular Endothelial Growth Factor A/metabolism
9.
Mol Cell Biochem ; 410(1-2): 207-13, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26350566

ABSTRACT

Over-activated PI3K/Akt signaling, a pathway strongly related to cancer survival and proliferation, has been reported recently to be involved in pulmonary artery smooth muscle cell apoptosis and proliferation in pulmonary hypertension (PH). In this study, we observed greatly increased lipocalin-2 (Lcn2) expression accompanied with over-activated PI3K/Akt signaling in a standard rat model of PH induced by monocrotaline. In view of the close relationship between Lcn2 and PI3K/Akt pathway, we hypothesized that the up-regulated Lcn2 might be a trigger of over-activated PI3K/Akt signaling in PH. Our results showed that Lcn2 significantly activated the PI3K/Akt pathway (determined by augmented Akt phosphorylation and up-regulated Mdm2) and significantly promoted proliferation (assessed by Ki67 staining) in cultured human pulmonary artery smooth muscle cells. Furthermore, we demonstrated that inhibition of Akt phosphorylation (LY294002) abrogated the Lcn2-promoted proliferation in cultured human pulmonary artery smooth muscle cells. In conclusion, Lcn2 significantly promoted human pulmonary artery smooth muscle cell proliferation by activating PI3K/Akt pathway. Further study on the role and mechanism of Lcn2 will help explore novel therapeutic strategies based on attenuating over-activated PI3K/Akt signaling in PH.


Subject(s)
Acute-Phase Proteins/pharmacology , Cell Proliferation/drug effects , Lipocalins/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins/pharmacology , Pulmonary Artery/drug effects , Vascular Remodeling/drug effects , Animals , Cells, Cultured , Disease Models, Animal , Enzyme Activation , Humans , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/pathology , Lipocalin-2 , Lipocalins/metabolism , Male , Monocrotaline , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Rats, Sprague-Dawley , Signal Transduction/drug effects , Up-Regulation
10.
Nat Commun ; 6: 8330, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26390966

ABSTRACT

Cyclic diguanylate monophosphate (c-di-GMP) is a well-conserved second messenger in bacteria. During infection, the innate immune system can also sense c-di-GMP; however, whether bacterial pathogens utilize c-di-GMP as a weapon to fight against host defense for survival and possible mechanisms underlying this process remain poorly understood. Siderocalin (LCN2) is a key antibacterial component of the innate immune system and sequesters bacterial siderophores to prevent acquisition of iron. Here we show that c-di-GMP can directly target the human LCN2 protein to inhibit its antibacterial activity. We demonstrate that c-di-GMP specifically binds to LCN2. In addition, c-di-GMP can compete with bacterial ferric siderophores to bind LCN2. Furthermore, c-di-GMP can significantly reduce LCN2-mediated inhibition on the in vitro growth of Escherichia coli. Thus, LCN2 acts as a c-di-GMP receptor. Our findings provide insight into the mechanism by which bacteria utilize c-di-GMP to interfere with the innate immune system for survival.


Subject(s)
Acute-Phase Proteins/metabolism , Anti-Bacterial Agents/metabolism , Carrier Proteins/metabolism , Lipocalins/metabolism , Proto-Oncogene Proteins/metabolism , Acute-Phase Proteins/genetics , Acute-Phase Proteins/pharmacology , Carrier Proteins/genetics , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Databases, Factual , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Humans , Lipocalin-2 , Lipocalins/genetics , Lipocalins/pharmacology , Models, Chemical , Models, Molecular , Molecular Structure , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Protein Binding , Protein Conformation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/pharmacology , Recombinant Proteins
11.
Arthritis Rheumatol ; 67(9): 2447-56, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26016600

ABSTRACT

OBJECTIVE: Giant cell arteritis (GCA) is pathologically characterized by dysfunctional angiogenesis and inflammatory cell infiltration. Acute-phase serum amyloid A (A-SAA) is an acute-phase reactant, but is also produced at sites of inflammation and may contribute to vascular inflammation in atherosclerosis. This study was undertaken to examine the effect of A-SAA on proinflammatory pathways and angiogenesis in GCA, using a novel ex vivo temporal artery tissue explant model. METHODS: Serum A-SAA levels were measured by enzyme-linked immunosorbent assay (ELISA). Temporal artery explants and peripheral blood mononuclear cell (PBMC) cultures were established from patients with GCA. Temporal artery explant morphology, viability, and spontaneous release of proinflammatory mediators following 24-hour culture were assessed by hematoxylin and eosin, calcein viability staining, and ELISA. Temporal artery explants and PBMC cultures were stimulated with A-SAA (10 µg/ml), and interleukin-6 (IL-6), IL-8, vascular endothelial growth factor, Ang2, and matrix metalloproteinase 2 (MMP-2)/MMP-9 were quantified by ELISA and gelatin zymography. The effect of conditioned medium from temporal artery explants on angiogenesis was assessed using endothelial cell Matrigel tube-formation assays. Temporal artery explants were also embedded in Matrigel, and myofibroblast outgrowth was assessed. RESULTS: Serum A-SAA levels were significantly higher in GCA patients versus healthy controls (P < 0.0001). Intact tissue morphology, cell viability, and spontaneous cytokine secretion were demonstrated in temporal artery explants. A-SAA treatment induced a significant increase in the levels of IL-6 and IL-8 from temporal artery explants (P < 0.05) and IL-8 from PBMCs (P < 0.05) compared to basal conditions. Conditioned medium from A-SAA-treated explants significantly induced angiogenic tube formation (P < 0.05 versus basal controls). Finally, A-SAA induced myofibroblast outgrowth and MMP-9 activation. CONCLUSION: Our findings demonstrate a functional role for A-SAA in regulating temporal artery inflammation, angiogenesis, and invasion, all key processes in the pathogenesis of GCA.


Subject(s)
Giant Cell Arteritis/immunology , Myofibroblasts/drug effects , Neovascularization, Pathologic/immunology , Serum Amyloid A Protein/pharmacology , Temporal Arteries/drug effects , Acute-Phase Proteins/immunology , Acute-Phase Proteins/pharmacology , Aged , Aged, 80 and over , Cells, Cultured , Female , Giant Cell Arteritis/metabolism , Humans , In Vitro Techniques , Inflammation/immunology , Interleukin-6/immunology , Interleukin-8/drug effects , Interleukin-8/immunology , Leukocytes, Mononuclear , Male , Matrix Metalloproteinase 2/drug effects , Matrix Metalloproteinase 2/immunology , Matrix Metalloproteinase 9/drug effects , Matrix Metalloproteinase 9/immunology , Middle Aged , Serum Amyloid A Protein/immunology , Temporal Arteries/immunology , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/immunology , Vesicular Transport Proteins/drug effects , Vesicular Transport Proteins/immunology
12.
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
13.
Mol Immunol ; 65(2): 205-14, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25700343

ABSTRACT

Lipopolysaccharide-binding protein (LBP) is an acute-phase protein involved in host response to Gram-negative and Gram-positive pathogens. It has been reported to exert diverse biological activities, such as anti-inflammatory effects. However, what effects it has on bovine mastitis has not been investigated. The aim of this study was to verify the anti-inflammatory properties of LBP on the inflammatory response of primary bovine mammary epithelial cells (BMEC) induced by lipopolysaccharide (LPS), and to determine the underlying mechanism. Bovine mammary epithelial cells were treated with various concentrations of LPS (1, 10, 20, and 100 µg/mL) for 3, 6, 12, and 24h. The results showed that LPS significantly inhibited cell viability in a dose-dependent manner. When cells were treated with LPS (10 µg/mL) for 12 h, the permeability of the cell membrane increased significantly. This promoted apoptosis. Various concentrations (10 and 20 µg/mL) of bovine recombinant lipopolysaccharide binding protein (BRLBP) could weaken the inflammation injury of BMEC induced by LPS without cytotoxicity. Toll-like receptor 4 (TLR4), nuclear factor κB (NF-κB), IL-1ß, and tumor necrosis factor α (TNF-α) from BMEC were decreased. TLR4 and NF-κB P65 protein levels were down-regulated, and nuclear transcription factor κB activity was also weakened. All these results indicated that the protective effects of high concentrations of BRLBP on LPS-induced inflammation injury in BMEC were at least partially achieved by the decreased production of pro-inflammatory cytokines. BRLBP was found to directly inhibit LPS/TLR4-mediated NF-κB activation. One possible anti-inflammatory mechanism can be attributed to the negative role of BRLBP in suppressing TLR4/NF-κB activation mediated by LPS. These findings suggested that BRLBP may be a useful agent to treat LPS-induced mastitis.


Subject(s)
Acute-Phase Proteins/pharmacology , Apoptosis/drug effects , Carrier Proteins/pharmacology , Epithelial Cells/immunology , Lipopolysaccharides/toxicity , Mastitis, Bovine/drug therapy , Membrane Glycoproteins/pharmacology , Animals , Apoptosis/immunology , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Epithelial Cells/pathology , Female , Inflammation/drug therapy , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/immunology , Mammary Glands, Animal , Mastitis, Bovine/immunology , Mastitis, Bovine/pathology , Recombinant Proteins/pharmacology , Time Factors , Toll-Like Receptor 4/immunology , Transcription Factor RelA/immunology , Tumor Necrosis Factor-alpha/immunology
14.
Int Urol Nephrol ; 46(8): 1673-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24973203

ABSTRACT

PURPOSE: The aim of this study was to elucidate the role of neutrophil gelatinase-associated lipocalin (NGAL) in regulating apoptosis of tubular epithelial cells in a hypoxia-reperfusion model. METHODS: A hypoxia-reperfusion model was established with NRK-52E cells to assess apoptosis and cell cycle progression after the addition of NGAL. We investigated the expression of four apoptosis factors, Bcl-2, Bax, Fas and FasL, as well as the expression level of two NGAL receptors, 24p3R and megalin, by both Western blot and real-time PCR. RESULTS: NGAL induced cell proliferation and reduced apoptosis by regulating four apoptosis factors Bcl-2, Bax, Fas and FasL. Western blot demonstrated that the two NGAL receptors, 24p3R and megalin, were increased after hypoxia-reperfusion, which was reduced by exogenous NGAL. Moreover, overexpression of the two receptors induced the expression of the anti-apoptotic factor Bcl-2 and reduced the expression of pro-apoptotic Bax, Fas and FasL. CONCLUSIONS: These findings indicate that NGAL reduces apoptosis by regulating the four apoptosis factors Bcl-2, Bax, Fas and FasL through its two receptors 24p3R and megalin. These results also suggest that ectopic expression of NGAL in renal cells might provide a therapeutic strategy in ischemia-reperfusion by reducing apoptosis and promoting renal cell proliferation.


Subject(s)
Acute-Phase Proteins/pharmacology , Apoptosis/drug effects , Cell Hypoxia/physiology , Epithelial Cells/drug effects , Lipocalins/pharmacology , Proto-Oncogene Proteins/pharmacology , RNA, Messenger/metabolism , Reperfusion Injury/metabolism , Animals , Cell Line , Cell Proliferation , Down-Regulation/drug effects , Epithelial Cells/physiology , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Kidney Tubules , Lipocalin-2 , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Receptors, Cell Surface/metabolism , Up-Regulation/drug effects , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , fas Receptor/genetics , fas Receptor/metabolism
15.
PLoS One ; 9(2): e89497, 2014.
Article in English | MEDLINE | ID: mdl-24586826

ABSTRACT

BACKGROUND: Neutrophil gelatinase-associated lipocalin (NGAL) is emerging as a mediator of various biological and pathological states. However, the specific biological role of this molecule remains unclear, as it serves as a biomarker for many conditions. The high sensitivity of NGAL as a biomarker coupled with relatively low specificity may hide important biological roles. Data point toward an acute compensatory, protective role for NGAL in response to adverse cellular stresses, including inflammatory and oxidative stress. The aim of this study was to understand whether NGAL modulates the T-cell response through regulation of the human leukocyte antigen G (HLA-G) complex, which is a mediator of tolerance. METHODOLOGY/PRINCIPAL FINDINGS: Peripheral blood mononuclear cells (PBMCs) were obtained from eight healthy donors and isolated by centrifugation on a Ficoll gradient. All donors gave informed consent. PBMCs were treated with four different concentrations of NGAL (40-320 ng/ml) in an iron-loaded or iron-free form. Changes in cell phenotype were analyzed by flow cytometry. NGAL stimulated expression of HLA-G on CD4+ T cells in a dose- and iron-dependent manner. Iron deficiency prevented NGAL-mediated effects, such that HLA-G expression was unaltered. Furthermore, NGAL treatment affected stimulation of regulatory T cells and in vitro expansion of CD4(+) CD25(+) FoxP3(+) cells. An NGAL neutralizing antibody limited HLA-G expression and significantly decreased the percentage of CD4(+) CD25(+) FoxP3(+) cells. CONCLUSIONS/SIGNIFICANCE: We provide in vitro evidence that NGAL is involved in cellular immunity. The potential role of NGAL as an immunomodulatory molecule is based on its ability to induce immune tolerance by upregulating HLA-G expression and expansion of T-regulatory cells in healthy donors. Future studies should further evaluate the role of NGAL in immunology and immunomodulation and its possible relationship to immunosuppressive therapy efficacy, tolerance induction in transplant patients, and other immunological disorders.


Subject(s)
Acute-Phase Proteins/metabolism , Lipocalins/metabolism , Proto-Oncogene Proteins/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Acute-Phase Proteins/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Enterobactin/pharmacology , Forkhead Transcription Factors/metabolism , HLA-G Antigens/metabolism , Humans , Immunophenotyping , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lipocalin-2 , Lipocalins/pharmacology , Lymphocyte Activation/drug effects , Proto-Oncogene Proteins/pharmacology , T-Lymphocyte Subsets , T-Lymphocytes, Regulatory/drug effects
16.
Hematology ; 19(8): 487-92, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24580532

ABSTRACT

BACKGROUND: Bacterial contamination of platelet products is the major infectious risk in blood transfusion medicine, which can result in life-threatening sepsis in recipient. Lipocalin 2 (Lcn2) is an iron-sequestering protein in the antibacterial innate immune response, which inhibit bacterial growth. This study was aimed to evaluate the antibacterial property of Lcn2 in preventing bacterial contamination of platelets. METHODS: Recombinant Lcn2 was expressed in a eukaryotic expression system and following purification and characterization of the recombinant Lcn2, its minimum inhibitory concentration was determined. Then, platelet concentrates were inoculated with various concentrations of Staphylococcus epidermidis, Pseudomonas aeruginosa, Klebsiella pneumoniae, Escherichia coli, and Enterococcus faecalis, and the antibacterial effects of Lcn2 was evaluated at 20-24 °C. RESULTS: Results revealed that Lcn2 effectively inhibited the growth of 1.5 × 10(4) CFU/ml S. epidermidis, P. aeruginosa, K. pneumoniae, E. coli, and E. faecalis at 40 ng/ml. At this concentration, Lcn2 also inhibited the growth of 1.5 × 10(3) CFU/ml Staphylococcus aureus and Proteus mirabilis. CONCLUSION: Recombinant Lcn2 inhibited growth of a variety of platelet-contaminating bacteria. Therefore, supplementation of platelet concentrates with Lcn2 may reduce bacterial contamination.


Subject(s)
Acute-Phase Proteins/pharmacology , Anti-Bacterial Agents/pharmacology , Blood Platelets/drug effects , Blood Platelets/microbiology , Lipocalins/pharmacology , Platelet Transfusion/methods , Proto-Oncogene Proteins/pharmacology , Enterococcus faecalis/drug effects , Escherichia coli/drug effects , Humans , Lipocalin-2 , Platelet Transfusion/adverse effects , Pseudomonas aeruginosa/drug effects , Recombinant Proteins/pharmacology , Staphylococcus epidermidis/drug effects
17.
Exp Neurol ; 254: 41-53, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24440229

ABSTRACT

Lipocalin-2 (LCN2) is an acute phase protein induced in response to injury, infection or other inflammatory stimuli. Based on the previously reported involvement of LCN2 in chemokine induction and in the recruitment of neutrophils at the sites of infection or tissue injury, we investigated the role of LCN2 in the pathogenesis of chronic/persistent inflammatory pain hypersensitivity. In the complete Freund's adjuvant (CFA)-induced chronic inflammatory pain model, LCN2 expression was strongly induced in the ipsilateral hindpaws, peaking at 12h after CFA injection and then gradually subsiding. In CFA-injected hindpaw tissues, LCN2 and its receptor 24p3R were mainly expressed in infiltrating neutrophils and macrophages. CFA-induced thermal hyperalgesia and mechanical allodynia were significantly diminished in Lcn2-deficient mice compared to wild-type animals. Furthermore, neutrophil infiltration, myeloperoxidase activity, expression of TNF-α, IL-1ß and MIP-2 in CFA-injected hindpaws, and spinal glial activation were markedly reduced by Lcn2 deficiency. An intraplantar injection of recombinant LCN2 protein induced thermal and mechanical hypersensitivities in naïve mice, and this was accompanied by neutrophil and macrophage infiltration into the hindpaws and glial activation in the dorsal horn of the spinal cord. Taken together, our results show that inflammatory cell-derived LCN2 at the sites of inflammation plays important roles in central sensitization and the subsequent nociceptive behavior in the rodent model of chronic inflammatory pain.


Subject(s)
Acute-Phase Proteins/immunology , Chronic Pain/immunology , Hyperalgesia/immunology , Inflammation/immunology , Lipocalins/immunology , Oncogene Proteins/immunology , Acute-Phase Proteins/genetics , Acute-Phase Proteins/pharmacology , Adjuvants, Immunologic/pharmacology , Animals , Cells, Cultured , Chronic Pain/genetics , Cytokines/immunology , Disease Models, Animal , Female , Freund's Adjuvant/pharmacology , Hyperalgesia/genetics , Inflammation/chemically induced , Inflammation/genetics , Lipocalin-2 , Lipocalins/genetics , Lipocalins/pharmacology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , Neutrophils/immunology , Nociceptors/immunology , Oncogene Proteins/genetics , Oncogene Proteins/pharmacology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Recombinant Proteins/pharmacology , Spinal Cord/immunology
18.
J Oral Pathol Med ; 43(5): 388-94, 2014 May.
Article in English | MEDLINE | ID: mdl-24372705

ABSTRACT

BACKGROUND: Synovial chondromatosis (SC) of temporomandibular joint (TMJ) is a rare proliferative disorder characterized by the formation of cartilaginous or osteocartilaginous nodules in synovium and joint space. Fibroblast growth factor 2 (FGF-2) is frequently applied in chondrogenic differentiation assays. Therefore, we hypothesized that FGF-2 might involved in the pathogenesis of SC. METHODS: SC synovium and loose bodies (LBs) specimens were observed by histological and immunohistochemical methods. Real-time PCR was conducted for comparing genes expressions in SC and normal synovium. SC synoviocytes were stimulated by FGF-2 in the presence or absence of its antagonist long pentraxin-3 (PTX3) for 6 days. Real-time PCR and alkaline phosphatase (ALP) activity were performed to examine the effects exerted by FGF-2 and PTX3. RESULTS: SC synovium, no matter facing the articular cavity or covering LB, was characterized by increased quantity of synoviocytes and blood vessels. FGF-2 was expressed in chondrocytes and fibroblast-like cells of LBs, and the wall of blood vessels. Expressions of chondrogenic genes (Sox9 and Wnt-4), osteogenic genes (Foxc2), FGF-2, and VEGF-A mRNA were significantly higher in SC synovium than that of the control group. The stimulation of FGF-2 on SC synoviocytes increased ALP activity and expressions of chondrogenic genes (Sox9, Col2α1, and Aggrecan), osteogenic genes (Foxc2, osteocalcin, and Col1α1), and VEGF-A, but PTX3 inhibited these effects. CONCLUSION: FGF-2 was responsible for the formation of cartilaginous loose bodies and involved in the pathogenesis of SC.


Subject(s)
Chondromatosis, Synovial/etiology , Fibroblast Growth Factor 2/pharmacology , Temporomandibular Joint Disorders/etiology , Acute-Phase Proteins/pharmacology , Aggrecans/analysis , Alkaline Phosphatase/analysis , Blood Vessels/chemistry , C-Reactive Protein/pharmacology , Cell Culture Techniques , Cells, Cultured , Chondrocytes/chemistry , Chondrogenesis/drug effects , Chondromatosis, Synovial/metabolism , Collagen Type I/analysis , Collagen Type I, alpha 1 Chain , Collagen Type II/analysis , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/antagonists & inhibitors , Fibroblasts/chemistry , Forkhead Transcription Factors/analysis , Humans , Joint Loose Bodies/etiology , Joint Loose Bodies/metabolism , Osteocalcin/analysis , Osteogenesis/drug effects , SOX9 Transcription Factor/analysis , Serum Amyloid P-Component/pharmacology , Synovial Membrane/chemistry , Synovial Membrane/drug effects , Synovial Membrane/pathology , Temporomandibular Joint Disorders/metabolism , Vascular Endothelial Growth Factor A/analysis , Wnt4 Protein/analysis
19.
PLoS Pathog ; 9(12): e1003792, 2013.
Article in English | MEDLINE | ID: mdl-24367257

ABSTRACT

Vertebrate females transfer antibodies via the placenta, colostrum and milk or via the egg yolk to protect their immunologically immature offspring against pathogens. This evolutionarily important transfer of immunity is poorly documented in invertebrates and basic questions remain regarding the nature and extent of parental protection of offspring. In this study, we show that a lipopolysaccharide binding protein/bactericidal permeability increasing protein family member from the invertebrate Biomphalaria glabrata (BgLBP/BPI1) is massively loaded into the eggs of this freshwater snail. Native and recombinant proteins displayed conserved LPS-binding, antibacterial and membrane permeabilizing activities. A broad screening of various pathogens revealed a previously unknown biocidal activity of the protein against pathogenic water molds (oomycetes), which is conserved in human BPI. RNAi-dependent silencing of LBP/BPI in the parent snails resulted in a significant reduction of reproductive success and extensive death of eggs through oomycete infections. This work provides the first functional evidence that a LBP/BPI is involved in the parental immune protection of invertebrate offspring and reveals a novel and conserved biocidal activity for LBP/BPI family members.


Subject(s)
Acute-Phase Proteins/metabolism , Antimicrobial Cationic Peptides/metabolism , Biomphalaria , Blood Proteins/metabolism , Carrier Proteins/metabolism , Immunity, Maternally-Acquired , Infections/immunology , Membrane Glycoproteins/metabolism , Oomycetes , Zygote , Acute-Phase Proteins/genetics , Acute-Phase Proteins/pharmacology , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/pharmacology , Biomphalaria/genetics , Biomphalaria/immunology , Biomphalaria/metabolism , Biomphalaria/parasitology , Blood Proteins/genetics , Blood Proteins/pharmacology , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Cloning, Molecular , Escherichia coli/drug effects , Escherichia coli/growth & development , Escherichia coli/metabolism , Escherichia coli/ultrastructure , Female , Immunity, Maternally-Acquired/genetics , Infections/genetics , Infections/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/pharmacology , Microbial Sensitivity Tests , Oomycetes/drug effects , Oomycetes/immunology , Oomycetes/pathogenicity , Recombinant Proteins/pharmacology , Zygote/immunology , Zygote/metabolism , Zygote/parasitology
20.
J Biol Chem ; 288(33): 24116-27, 2013 Aug 16.
Article in English | MEDLINE | ID: mdl-23836894

ABSTRACT

Lipocalin 2 (LCN2), which is also known as 24p3 and neutrophil gelatinase-associated lipocalin (NGAL), binds small, hydrophobic ligands and interacts with cell surface receptor 24p3R to regulate diverse cellular processes. In the present study, we examined the role of LCN2 in the pathogenesis of neuropathic pain using a mouse model of spared nerve injury (SNI). Lcn2 mRNA levels were significantly increased in the dorsal horn of the spinal cord after SNI, and LCN2 protein was mainly localized in neurons of the dorsal and ventral horns. LCN2 receptor 24p3R was expressed in spinal neurons and microglia after SNI. Lcn2-deficient mice exhibited significantly less mechanical pain hypersensitivity during the early phase after SNI, and an intrathecal injection of recombinant LCN2 protein elicited mechanical pain hypersensitivity in naive animals. Lcn2 deficiency, however, did not affect acute nociceptive pain. Lcn2-deficient mice showed significantly less microglial activation and proalgesic chemokine (CCL2 and CXCL1) production in the spinal cord after SNI than wild-type mice, and recombinant LCN2 protein induced the expression of these chemokines in cultured neurons. Furthermore, the expression of LCN2 and its receptor was detected in neutrophils and macrophages in the sciatic nerve following SNI, suggesting the potential role of peripheral LCN2 in neuropathic pain. Taken together, our results indicate that LCN2 plays a critical role in the development of pain hypersensitivity following peripheral nerve injury and suggest that LCN2 mediates neuropathic pain by inducing chemokine expression and subsequent microglial activation.


Subject(s)
Acute-Phase Proteins/metabolism , Chemokines/metabolism , Lipocalins/metabolism , Neuralgia/etiology , Neuralgia/metabolism , Oncogene Proteins/metabolism , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/metabolism , Signal Transduction , Acute-Phase Proteins/genetics , Acute-Phase Proteins/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/pathology , Chemokines/genetics , Gene Expression Regulation/drug effects , Hyperalgesia/complications , Hyperalgesia/metabolism , Hyperalgesia/pathology , Lipocalin-2 , Lipocalins/genetics , Lipocalins/pharmacology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/enzymology , Microglia/pathology , Models, Biological , Neuralgia/enzymology , Neuralgia/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nociception/drug effects , Oncogene Proteins/genetics , Oncogene Proteins/pharmacology , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/pathology , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/metabolism , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...