Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Arterioscler Thromb Vasc Biol ; 40(6): 1523-1532, 2020 06.
Article in English | MEDLINE | ID: mdl-32321308

ABSTRACT

OBJECTIVE: Although the molecular components of circadian rhythms oscillate in discrete cellular components of the vasculature and many aspects of vascular function display diurnal variation, the cellular connections between the molecular clock and inflammatory cardiovascular diseases remain to be elucidated. Previously we have shown that pre- versus postnatal deletion of Bmal1 (brain and muscle aryl hydrocarbon receptor nuclear translocator-like 1), the nonredundant core clock gene has contrasting effects on atherogenesis. Here we investigated the effect of myeloid cell Bmal1 deletion on atherogenesis and abdominal aortic aneurysm formation in mice. Approach and Results: Mice lacking Bmal1 in myeloid cells were generated by crossing Bmal1 flox/flox mice with lysozyme 2 promoter-driven Cre recombinase mice on a hyperlipidemic low-density lipoprotein receptor-deficient background and were fed on a high-fat diet to induce atherosclerosis. Atherogenesis was restrained, concomitant with a reduction of aortic proinflammatory gene expression in myeloid cell Bmal1 knockout mice. Body weight, blood pressure, blood glucose, triglycerides, and cholesterol were unaltered. Similarly, myeloid cell depletion of Bmal1 also restrained Ang II (angiotensin II) induced formation of abdominal aortic aneurysm in hyperlipidemic mice. In vitro, RNA-Seq analysis demonstrated a proinflammatory response in cultured macrophages in which there was overexpression of Bmal1. CONCLUSIONS: Myeloid cell Bmal1 deletion retards atherogenesis and restrains the formation of abdominal aortic aneurysm and may represent a potential therapeutic target for inflammatory cardiovascular diseases.


Subject(s)
ARNTL Transcription Factors/deficiency , ARNTL Transcription Factors/physiology , Aortic Aneurysm, Abdominal/prevention & control , Atherosclerosis/prevention & control , Hyperlipidemias/complications , Myeloid Cells/chemistry , ARNTL Transcription Factors/genetics , Angiotensin II/pharmacology , Animals , Aortic Aneurysm, Abdominal/chemically induced , Atherosclerosis/etiology , Atherosclerosis/pathology , Cells, Cultured , Crosses, Genetic , Diet, High-Fat , Gene Deletion , Gene Expression , Hyperlipidemias/etiology , Inflammation , Integrases/genetics , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/physiology , Mice , Mice, Knockout , Muramidase/genetics , Promoter Regions, Genetic/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics
2.
J Immunol ; 203(7): 1918-1929, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31484733

ABSTRACT

ESAT-6 is a small secreted protein of Mycobacterium tuberculosis involved in the ESAT-6 secretion system (ESX-1)-mediated virulence and pathogenesis. The protein interacts with ß2M, causing downregulation of MHC class I Ag presentation, which could be one of the mechanisms by which it favors increased survival of the bacilli inside the host. In an earlier study, we have shown that the C-terminal region of ESAT-6 is crucial for its interaction with ß2M. However, the interface of ß2M involved in interaction with ESAT-6 and detailed physicochemical changes associated with ESAT-6:ß2M complexation are not fully defined. In this study, using computational and site-directed mutagenesis studies, we demonstrate the presence of strong noncovalent hydrophobic interactions between ESAT-6 and ß2M in addition to the vital hydrogen bonding between the aspartate residue (Asp53) of ß2M and methionine (Met93) of ESAT-6. Docking-based high-throughput virtual screening followed by 16-point screening on microscale thermophoresis resulted in the identification of two potent inhibitors (SM09 and SM15) that mask the critical Met93 residue of ESAT-6 that is required for ESAT-6:ß2M interaction and could rescue cell surface expression of ß2M and HLA in human macrophages as well as MHC class I Ag presentation suppressed by ESAT-6 in peritoneal macrophages isolated from C57BL/6 mice. Both SM09 and SM15 significantly inhibited intracellular survival of M. tuberculosis in human macrophages. Further, we characterized the physicochemical properties involved in the ESAT-6:ß2M complexation, which may help in understanding host-pathogen interactions.


Subject(s)
Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Molecular Docking Simulation , Mycobacterium tuberculosis/chemistry , beta 2-Microglobulin/chemistry , Amino Acid Substitution , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Aspartic Acid/chemistry , Aspartic Acid/genetics , Aspartic Acid/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Host-Pathogen Interactions/immunology , Humans , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/immunology , Mice , Mutagenesis, Site-Directed , Mutation, Missense , Mycobacterium tuberculosis/physiology , Protein Structure, Quaternary , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
3.
Int J Mol Sci ; 22(23)2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34884899

ABSTRACT

Palmitic acid (C16:0) is the most abundant saturated fatty acid in animals serving as a substrate in synthesis and ß-oxidation of other lipids, and in the modification of proteins called palmitoylation. The influence of dietary palmitic acid on protein S-palmitoylation remains largely unknown. In this study we performed high-throughput proteomic analyses of a membrane-enriched fraction of murine liver to examine the influence of a palm oil-rich diet (HPD) on S-palmitoylation of proteins. HPD feeding for 4 weeks led to an accumulation of C16:0 and C18:1 fatty acids in livers which disappeared after 12-week feeding, in contrast to an accumulation of C16:0 in peritoneal macrophages. Parallel proteomic studies revealed that HPD feeding induced a sequence of changes of the level and/or S-palmitoylation of diverse liver proteins involved in fatty acid, cholesterol and amino acid metabolism, hemostasis, and neutrophil degranulation. The HPD diet did not lead to liver damage, however, it caused progressing obesity, hypercholesterolemia and hyperglycemia. We conclude that the relatively mild negative impact of such diet on liver functioning can be attributed to a lower bioavailability of palm oil-derived C16:0 vs. that of C18:1 and the efficiency of mechanisms preventing liver injury, possibly including dynamic protein S-palmitoylation.


Subject(s)
Liver/metabolism , Palm Oil/administration & dosage , Palmitic Acid/chemistry , Proteomics/methods , Soybean Oil/administration & dosage , Amino Acids/metabolism , Animals , Dietary Supplements , Fatty Acids/analysis , Homeostasis , Liver/drug effects , Macrophages, Peritoneal/chemistry , Male , Mass Spectrometry , Mice , Palm Oil/chemistry , Palm Oil/pharmacology , Soybean Oil/pharmacology
4.
Molecules ; 24(7)2019 Mar 30.
Article in English | MEDLINE | ID: mdl-30934998

ABSTRACT

Leishmaniasis is a neglected tropical disease caused by members of the Leishmania genus of parasitic protozoa that cause different clinical manifestations of the disease. Current treatment options for the cutaneous disease are limited due to severe side effects, poor efficacy, limited availability or accessibility, and developing resistance. Essential oils may provide low cost and readily available treatment options for leishmaniasis. In-vitro screening of a collection of 52 commercially available essential oils has been carried out against promastigotes of Leishmania amazonensis. In addition, cytotoxicity has been determined for the essential oils against mouse peritoneal macrophages in order to determine selectivity. Promising essential oils were further screened against intracellular L. amazonensis amastigotes. Three essential oils showed notable antileishmanial activities: frankincense (Boswellia spp.), coriander (Coriandrum sativum L.), and wintergreen (Gualtheria fragrantissima Wall.) with IC50 values against the amastigotes of 22.1 ± 4.2, 19.1 ± 0.7, and 22.2 ± 3.5 µg/mL and a selectivity of 2, 7, and 6, respectively. These essential oils could be explored as topical treatment options for cutaneous leishmaniasis.


Subject(s)
Antiprotozoal Agents/chemistry , Leishmania/chemistry , Oils, Volatile/chemistry , Animals , Antiprotozoal Agents/pharmacology , Boswellia/chemistry , Cell Line , Cell Survival/drug effects , Coriandrum/chemistry , Drug Evaluation, Preclinical/methods , Inhibitory Concentration 50 , Leishmaniasis, Cutaneous/metabolism , Macrophages, Peritoneal/chemistry , Mice, Inbred BALB C , Oils, Volatile/pharmacology
5.
Anal Chem ; 90(22): 13663-13669, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30359531

ABSTRACT

Quantitative assay of microRNAs (miRNAs) with mass spectrometric detection currently suffers from two major disadvantages, i.e., being insufficient in sensitivity and requiring an extraction or chromatographic separation prior to MS detection. In this work, we developed a facile and sensitive assay of targeted miRNAs based on the combination of cyclic enzymatic amplification (CEA) with microfluidic voltage-assisted liquid desorption electrospray ionization tandem mass spectrometry (VAL-DESI-MS/MS). The single-stranded DNA (ssDNA) probe was designed to have a sequence complementary to the miRNA target with an extension of a two-base nucleotide fragment (i.e., CpC) at the 3'-position as MS signal reporter, thus being easy to prepare and high in stability. In the proposed CEA-VAL-DESI-MS/MS assay, an ssDNA probe was added to a sample solution, forming a DNA-miRNA hybrid. Duplex-specific nuclease (DSN) was then added to cleave specifically the DNA probe in the heteroduplex strands. As the hybridization-cleavage cycle repeated itself for many rounds, a large quantity of CpC molecules was produced that was quantified by VAL-DESI-MS/MS with accuracy and specificity. miRNA-21 was tested as the model target. The assay had a linear calibration equation in the range from 2.5 pM to 1.0 nM with a limit of detection of 0.25 pM. Determination of miRNA-21 in cellular samples was demonstrated. miRNA-21 was found to be 95.3 ± 13.95 amol ( n = 3) in 100 mouse peritoneal macrophages with a recovery of 94.2 ± 2.6% ( n = 3). Interestingly, analysis of exosomes secreted from these cells revealed that exposure of the cells to chemical stimuli caused a 3-fold increase in exosomal level of miRNA-21. The results suggest that the proposed assay may provide an accurate and cost-effective means for quantification of targeted miRNAs in biomedical samples.


Subject(s)
MicroRNAs/analysis , Microfluidics/methods , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Calibration , DNA Probes , DNA, Single-Stranded/chemistry , Limit of Detection , Macrophages, Peritoneal/chemistry , Mice , Reproducibility of Results
6.
Appl Microbiol Biotechnol ; 102(5): 2235-2249, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29353306

ABSTRACT

The cell surface of Toxoplasma gondii is covered by antigens (SAGs) from the SRS family anchored by glycosylphosphatidylinositol (GPI) and includes antigens from the SAG2 family. Among these, the SAG2A surface antigen shows great potential in activating humoral responses and has been used in characterizing the acute phase of infection and in the serological diagnosis of toxoplasmosis. In this study, we aimed to evaluate rSAG2A-induced proteins in BALB/c and C57BL/c mice macrophages and to evaluate the phenotypic polarization induced in the process. We treated the peritoneal macrophages from mouse strains that were resistant or susceptible to T. gondii with rSAG2A to analyze their proteomic profile by mass spectrometry and systems biology. We also examined the gene expression of these cells by RT-qPCR using the phenotypic markers of M1 and M2 macrophages. Differences were observed in the expression of proteins involved in the inflammatory process in both resistant and susceptible cells, and macrophages were preferentially induced to obtain a pro-inflammatory immune response (M1) via the overexpression of IL-1ß in mice susceptible to this parasite. These data suggest that the SAG2A antigen induces phenotypic and classical activation of macrophages in both resistant and susceptible strains of mice during the acute phase of the disease.


Subject(s)
Antigens, Protozoan/immunology , Interleukin-1beta/immunology , Macrophages, Peritoneal/parasitology , Protozoan Proteins/immunology , Toxoplasmosis/immunology , Animals , Antigens, Protozoan/genetics , Cells, Cultured , Female , Humans , Interleukin-1beta/chemistry , Interleukin-1beta/genetics , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proteomics , Protozoan Proteins/genetics , Toxoplasma/genetics , Toxoplasma/immunology , Toxoplasmosis/genetics , Toxoplasmosis/parasitology
7.
Mem Inst Oswaldo Cruz ; 109(6): 767-74, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25317704

ABSTRACT

Lipid bodies [lipid droplets (LBs)] are lipid-rich organelles involved in lipid metabolism, signalling and inflammation. Recent findings suggest a role for LBs in host response to infection; however, the potential functions of this organelle in Toxoplasma gondii infection and how it alters macrophage microbicidal capacity during infection are not well understood. Here, we investigated the role of host LBs in T. gondii infection in mouse peritoneal macrophages in vitro. Macrophages cultured with mouse serum (MS) had higher numbers of LBs than those cultured in foetal bovine serum and can function as a model to study the role of LBs during intracellular pathogen infection. LBs were found in association with the parasitophorous vacuole, suggesting that T. gondii may benefit from this lipid source. Moreover, increased numbers of macrophage LBs correlated with high prostaglandin E2 (PGE2) production and decreased nitric oxide (NO) synthesis. Accordingly, LB-enriched macrophages cultured with MS were less efficient at controlling T. gondii growth. Treatment of macrophages cultured with MS with indomethacin, an inhibitor of PGE2 production, increased the microbicidal capacity against T. gondii. Collectively, these results suggest that culture with MS caused a decrease in microbicidal activity of macrophages against T. gondii by increasing PGE2 while lowering NO production.


Subject(s)
Lipid Droplets/parasitology , Macrophage Activation/physiology , Macrophages, Peritoneal/parasitology , Toxoplasma/physiology , Vacuoles/parasitology , Animals , Cattle , Host-Parasite Interactions , Indomethacin/pharmacology , Lipid Droplets/physiology , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/physiology , Macrophages, Peritoneal/ultrastructure , Male , Mice , Mice, Inbred C3H , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Nitric Oxide/biosynthesis , Primary Cell Culture , Prostaglandins E/antagonists & inhibitors , Prostaglandins E/biosynthesis , Vacuoles/physiology
8.
Biomed Chromatogr ; 27(11): 1580-6, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23760986

ABSTRACT

Cell membrane chromatography (CMC) is a chromatographic biological affinity method that uses specific cell membranes as the stationary phase. In this study, a novel peritoneal macrophage/cell membrane chromatography (PM/CMC)-online-high performance liquid chromatography/mass spectrometry (HPLC/MS) method was established to screen for the anti-inflammatory components from traditional Chinese medicines using hydrocortisone and dexamethasone as standards. The stationary phase of the CMC employed mouse peritoneal macrophage cell membranes. This method was applied to the purification and identification of components in extracts of Chloranthus multistachys Pei. The major component retained by CMC was identified as isofraxidin by HPLC/MS. In vitro experiments revealed that IF was able to inhibit the production of nitric oxide and tumor necrosis factor-α in lipopolysaccharide-stimulated mice and peritoneal macrophages in a dose-dependent manner. The results demonstrated that the PM/CMC-online-HPLC/MS is an effective screening system for the rapid detection, enrichment, and identification of target components from complex samples.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cell Membrane/chemistry , Chromatography, High Pressure Liquid/instrumentation , Coumarins/pharmacology , Drugs, Chinese Herbal/pharmacology , Macrophages, Peritoneal/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/isolation & purification , Cells, Cultured , Coumarins/chemistry , Coumarins/isolation & purification , Drug Evaluation, Preclinical/instrumentation , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/isolation & purification , Equipment Design , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Magnoliopsida/chemistry , Mass Spectrometry/instrumentation , Mice , Nitric Oxide/immunology , Tumor Necrosis Factor-alpha/immunology
9.
Drug Discov Ther ; 16(4): 148-153, 2022 Sep 17.
Article in English | MEDLINE | ID: mdl-36002309

ABSTRACT

Phenochalasin A, a unique phenol-containing cytochalasin produced by the marine-derived fungus Phomopsis sp. FT-0211, was originally discovered in a cell morphological assay of observing the inhibition of lipid droplet formation in mouse peritoneal macrophages. To investigate the mode of action and binding proteins, phenochalasin A was radio-labeled by 125I. Iodinated phenochalasin A exhibited the same biological activity as phenochalasin A. [125I]Phenochalasin A was found to be associated with an approximately 40 kDa protein, which was identified as G-actin. Furthermore, detail analyses of F-actin formation in Chinese hamster ovary cells (CHO-K1 cells) indicated that phenochalasin A (2 µM) caused elimination of F-actin formation on the apical site of the cells, suggesting that actin-oriented specific function(s) in cytoskeletal processes are affected by phenochalasin A.


Subject(s)
Actins , Lipid Droplets , Actins/analysis , Actins/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Cytochalasins/metabolism , Cytochalasins/pharmacology , Indoles , Iodine Radioisotopes , Lactones , Lipid Droplets/chemistry , Lipid Droplets/metabolism , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/metabolism , Mice , Phenols
10.
Biochem Biophys Res Commun ; 407(4): 656-62, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21420383

ABSTRACT

Triglyceride-rich lipoproteins (TGRLs) and low-density-lipoprotein (LDL) cholesterol are independent risk factors for coronary artery disease. We have previously proposed that the very low-density-lipoprotein (VLDL) receptor is one of the receptors required for foam cell formation by TGRLs in human macrophages. However, the VLDL receptor proteins have not been detected in atherosclerotic lesions of several animal models. Here we showed no VLDL receptor protein was detected in mouse macrophage cell lines (Raw264.7 and J774.2) or in mouse peritoneal macrophages in vitro. Furthermore, no VLDL receptor protein was detected in macrophages in atherosclerotic lesions of chow-fed apolipoprotein E-deficient or cholesterol-fed LDL receptor-deficient mice in vivo. In contrast, macrophage VLDL receptor protein was clearly detected in human macrophages in vitro and in atherosclerotic lesions in myocardial infarction-prone Watanabe-heritable hyperlipidemic (WHHLMI) rabbits in vivo. There are species differences in the localization of VLDL receptor protein in vitro and in vivo. Since VLDL receptor is expressed on macrophages in atheromatous plaques of both rabbit and human but not in mouse models, the mechanisms of atherogenesis and/or growth of atherosclerotic lesions in mouse models may be partly different from those of humans and rabbits.


Subject(s)
Macrophages, Peritoneal/metabolism , Receptors, LDL/metabolism , Animals , Humans , Immunohistochemistry , Macrophages, Peritoneal/chemistry , Male , Mice , Mice, Knockout , Middle Aged , Rabbits , Receptors, LDL/analysis , Species Specificity
11.
Crit Care Med ; 39(4): 756-64, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21200322

ABSTRACT

OBJECTIVE: To investigate the role and signaling pathway of peroxiredoxin 6, a newly identified peroxidase, in lipopolysaccharide-induced acute lung injury. DESIGN: Prospective, randomized, controlled study. SETTING: Research laboratory. SUBJECTS: Peroxiredoxin 6 (-/-) and wild-type C57BL/6 mice. INTERVENTIONS: Wild-type or peroxiredoxin 6 (-/-) mice were challenged by intratracheal instillation of lipopolysaccharide (5 mg/kg) for 4 hrs or 24 hrs for lung injury measurement. In other studies, peritoneal macrophages, isolated from wild-type and peroxiredoxin 6 (-/-) mice, were preincubated in presence or absence of mitogen-activated protein kinases inhibitors for 30 mins before being stimulated with lipopolysaccharide (1 µg/mL) for 4 hrs. MEASUREMENTS AND MAIN RESULTS: Bronchoalveolar lavage myeloperoxidase activity and the lung injury score were significantly increased in peroxiredoxin 6 (-/-) mice compared with wild-type mice after lipopolysaccharide instillation at both 4 hrs and 24 hrs. Hydrogen peroxide and malondialdehyde levels, as well as nuclear factor-κB activities, tumor necrosis factor-α, interleukin-1ß, and matrix metalloproteinase-9 messenger RNA, protein concentration, and activities were significantly increased whereas total antioxidative capability was markedly decreased in lungs of peroxiredoxin 6 (-/-) mice compared with wild-type mice. In vitro studies showed intracellular reactive oxygen species levels and release of tumor necrosis factor-α, interleukin-1, and matrix metalloproteinase-9 were significantly increased in macrophages from peroxiredoxin 6 (-/-) mice compared with that from wild-type mice after lipopolysaccharide stimulation. Cytokines release was partially suppressed by extracellular signal-regulated kinase and c-Jun N-terminal kinase inhibitors, but not by the p38 mitogen-activated protein kinase inhibitor. CONCLUSIONS: Deletion of peroxiredoxin 6 exaggerates lipopolysaccharide-induced acute lung injury and inflammation with increased oxidative stress, inflammatory responses, and matrix degradation, all of which were partially dependent on nuclear factor-κB, extracellular signal-regulated kinase, and c-Jun N-terminal kinase pathways.


Subject(s)
Acute Lung Injury/etiology , Peroxiredoxin VI/physiology , Acute Lung Injury/physiopathology , Animals , Hydrogen Peroxide/analysis , Interleukin-1beta/analysis , Lipopolysaccharides/pharmacology , Lung/chemistry , Lung/drug effects , Macrophages, Peritoneal/chemistry , Male , Malondialdehyde/analysis , Matrix Metalloproteinase 9/analysis , Mice , Mice, Inbred C57BL , NF-kappa B/analysis , Oxidative Stress , Peroxiredoxin VI/deficiency , Reactive Oxygen Species/analysis , Tumor Necrosis Factor-alpha/analysis
12.
J Infect Dis ; 202(7): 1136-45, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20731582

ABSTRACT

The fungal pathogen Histoplasma capsulatum evades the innate and adaptive immune responses and thrives within resting macrophages. Cytokines that induce antimicrobial activity, such as granulocyte macrophage colony-stimulating factor (GM-CSF), inhibit H. capsulatum growth in macrophages. Conversely, interleukin 4 inhibits the killing of intracellular pathogens. Using inductively coupled plasma mass spectrometry, we examined alterations in the metal homeostasis of murine H. capsulatum-infected macrophages that were exposed to activating cytokines. Decreases in the levels of iron (Fe(2+) and Fe(3+)) and zinc (Zn(2+)) were observed in infected, GM-CSF-treated macrophages compared with those in infected controls. Interleukin 4 reversed the antifungal activity of GM-CSF-activated macrophages and was associated with increased intracellular Zn(2+) levels. Chelation of Zn(2+) inhibited yeast replication in both the absence of macrophages and the presence of macrophages. Treatment of cells with GM-CSF altered the host Zn(2+) binding species profile. These results establish that Zn(2+) deprivation may be a host defense mechanism utilized by macrophages.


Subject(s)
Cytokines/immunology , Histoplasma/immunology , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/immunology , Zinc/analysis , Zinc/immunology , Animals , Chelating Agents/metabolism , Iron/analysis , Iron/immunology , Macrophages, Peritoneal/microbiology , Mass Spectrometry , Mice , Mice, Inbred C57BL
13.
Eur J Pediatr Surg ; 31(1): 86-94, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32950032

ABSTRACT

INTRODUCTION: Human peritoneal macrophages are resident in the abdominal cavity where they support the specific microenvironmental regulation. We have previously observed a phenotypic switch of murine macrophages during infancy that was associated with a functional development. To investigate the age related changes in human peritoneal macrophages, we analyzed peritoneal macrophages of children undergoing laparoscopic procedures. MATERIALS AND METHODS: Immunologically healthy children who received minimally invasive surgery in our department were included in this study. In all cases, the written consent was obtained. At the beginning of laparoscopy, physiologic NaCl-solution was instilled and manually removed through the umbilical trocar to gain macrophages. Lavage cells were processed for flow cytometry analysis. CD14+ myeloid cells were monitored for specific lineage marker expression. RESULTS: A total of 21 donors (age: 7 days-18 years) were included and divided into three groups. In all age groups, 97% of myeloid cells expressed CD11b. 70% of these expressed CD14. Three subsets of CD14 cells were detected on the basis of CD14/CD16 expression (CD14 + CD16dim, CD14 + CD16inter, and CD14 + CD16high). In neonates, >80% belonged to the CD14 + CD16high subset, reducing to 30% in adolescents. In none of the cases, the M2 markers CD23 and CD25 were expressed. CONCLUSION: This is the first study showing that lineage marker expression of peritoneal macrophages in neonates differs from that in adults. The knowledge about neonatal tissue resident macrophages might help to understand their complex interaction and to use specific macrophage properties for therapeutic approaches.


Subject(s)
Macrophages, Peritoneal/metabolism , Adolescent , Age Factors , Animals , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Macrophages, Peritoneal/chemistry , Male , Peritoneal Lavage/methods
14.
Front Immunol ; 12: 684967, 2021.
Article in English | MEDLINE | ID: mdl-34054877

ABSTRACT

Most multicellular organisms have a major body cavity containing vital organs. This cavity is lined by a mucosa-like serosal surface and filled with serous fluid which suspends many immune cells. Injuries affecting the major body cavity are potentially life-threatening. Here we summarize evidence that unique damage detection and repair mechanisms have evolved to ensure immediate and swift repair of injuries at serosal surfaces. Furthermore, thousands of patients undergo surgery within the abdominal and thoracic cavities each day. While these surgeries are potentially lifesaving, some patients will suffer complications due to inappropriate scar formation when wound healing at serosal surfaces defects. These scars called adhesions cause profound challenges for health care systems and patients. Therefore, reviewing the mechanisms of wound repair at serosal surfaces is of clinical importance. Serosal surfaces will be introduced with a short embryological and microanatomical perspective followed by a discussion of the mechanisms of damage recognition and initiation of sterile inflammation at serosal surfaces. Distinct immune cells populations are free floating within the coelomic (peritoneal) cavity and contribute towards damage recognition and initiation of wound repair. We will highlight the emerging role of resident cavity GATA6+ macrophages in repairing serosal injuries and compare serosal (mesothelial) injuries with injuries to the blood vessel walls. This allows to draw some parallels such as the critical role of the mesothelium in regulating fibrin deposition and how peritoneal macrophages can aggregate in a platelet-like fashion in response to sterile injury. Then, we discuss how serosal wound healing can go wrong, causing adhesions. The current pathogenetic understanding of and potential future therapeutic avenues against adhesions are discussed.


Subject(s)
Macrophages, Peritoneal/immunology , Peritoneum/immunology , Serous Membrane/immunology , Wounds and Injuries/immunology , Animals , Ascitic Fluid/immunology , Blood Platelets/immunology , Cell Aggregation/immunology , GATA6 Transcription Factor/analysis , Humans , Macrophages, Peritoneal/chemistry , Peritoneum/injuries , Tissue Adhesions/immunology
15.
Science ; 371(6533)2021 03 05.
Article in English | MEDLINE | ID: mdl-33674464

ABSTRACT

Most multicellular organisms have a major body cavity that harbors immune cells. In primordial species such as purple sea urchins, these cells perform phagocytic functions but are also crucial in repairing injuries. In mammals, the peritoneal cavity contains large numbers of resident GATA6+ macrophages, which may function similarly. However, it is unclear how cavity macrophages suspended in the fluid phase (peritoneal fluid) identify and migrate toward injuries. In this study, we used intravital microscopy to show that cavity macrophages in fluid rapidly form thrombus-like structures in response to injury by means of primordial scavenger receptor cysteine-rich domains. Aggregates of cavity macrophages physically sealed injuries and promoted rapid repair of focal lesions. In iatrogenic surgical situations, these cavity macrophages formed extensive aggregates that promoted the growth of intra-abdominal scar tissue known as peritoneal adhesions.


Subject(s)
Macrophages, Peritoneal/immunology , Peritoneum/immunology , Peritoneum/injuries , Wounds and Injuries/immunology , Animals , Ascitic Fluid/immunology , Blood Platelets/immunology , Cell Aggregation/immunology , GATA6 Transcription Factor/analysis , Macrophages, Peritoneal/chemistry , Mice , Mice, Inbred C57BL , Scavenger Receptors, Class B/immunology , Thrombosis/immunology , Tissue Adhesions/immunology
16.
Genes Immun ; 11(6): 447-57, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20090772

ABSTRACT

Guanine-nucleotide exchange factors (GEFs) stimulate the intrinsic GDP/GTP exchange activity of Ras and promote the formation of active Ras-GTP, which in turn controls diverse signalling networks important for the regulation of cell proliferation, survival, differentiation, vesicular trafficking, and gene expression. RasGEF1b is a GEF, whose expression is induced in macrophages on stimulation with toll-like receptor (TLR) agonists. Here, we showed that in vitro RasGEF1b expression by macrophages is mostly induced by TLR3 (poly I:C) and TLR4 (lipopolysaccharyde) through the MyD88-independent pathway. In vivo infection with the protozoan parasites Trypanosoma cruzi and Plasmodium chabaudi induced RasGEF1b in an MyD88-, TRIF-, and IFN-gamma-dependent manner. Ectopically expressed RasGEF1b was found, mostly, in the heavy membrane fraction of HEK 293T, and by confocal microscopy, it was found to be located at early endosomes. Computational modelling of the RasGEF1b-Ras interaction revealed that RasGEF1b interacts with the binding domain site of Ras, a critical region for interacting with GEFs involved in the activation of Ras-Raf-MEK-ERK pathway. More important, RasGEF1b was found to be closely associated with Ras in live cells and to trigger Ras activity. Altogether, these results indicate that on TLR activation, RasGEF1b may trigger Ras-like proteins and regulate specific biological activities described for this subtype of GTPases.


Subject(s)
Endosomes/metabolism , Toll-Like Receptors/physiology , ras Guanine Nucleotide Exchange Factors/biosynthesis , Animals , CHO Cells , Cricetinae , Cricetulus , Endosomes/chemistry , Female , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Toll-Like Receptors/metabolism , ras Guanine Nucleotide Exchange Factors/metabolism , ras Guanine Nucleotide Exchange Factors/physiology
17.
J Exp Med ; 183(4): 1899-904, 1996 Apr 01.
Article in English | MEDLINE | ID: mdl-8666946

ABSTRACT

Taxol, a microtubule-binding diterpene, mimics many effects of lipopolysaccharide (LPS) on mouse macrophages. The LPS-mimetic effects of taxol appear to be under the same genetic control as responses to LPS itself. Thus we have postulated a role for microtubule-associated proteins (MAP) in the response of macrophages to LPS. Stimulation of macrophages by LPS quickly induces the activation of mitogen-activated protein kinases (MAPK). MAPK are generally considered cytosolic enzymes. Herein we report that much of the LPS-activatable pool of MAPK in primary mouse peritoneal macrophages is microtubule associated. By immunofluorescence, MAPK were localized to colchicine- and nocodazole-disruptible filaments. From both mouse brain and RAW 264.7 macrophages, MAPK could be coisolated with polymerized tubulin. Fractionation of primary macrophages into cytosol-, microfilament-, microtubule-, and intermediated filament-rich extracts revealed that approximately 10% of MAPK but none of MAPK kinase (MEK1A and MEK2) was microtubule bound. Exposure of macrophages to LPS did not change the proportion of MAPK bound to microtubules, but preferentially activated the microtubule-associated pool. These findings confirm the prediction that LPS activates a kinase bound to microtubules. Together with LPS-mimetic actions of taxol and the shared genetic control of responses to LPS and taxol, these results support the hypothesis that a major LPS-signaling pathway in mouse macrophages may involve activation of one or more microtubule-associated kinases.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Macrophages, Peritoneal/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Animals , Brain Chemistry , Calcium-Calmodulin-Dependent Protein Kinases/isolation & purification , Cell Compartmentation , Cell Fractionation , Cells, Cultured , Enzyme Activation , Female , Fluorescent Antibody Technique , Lipopolysaccharides/pharmacology , Macrophage Activation , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/drug effects , Mice , Microtubule-Associated Proteins/isolation & purification , Microtubules/chemistry , Paclitaxel/pharmacology
18.
J Neurochem ; 109(3): 694-705, 2009 May.
Article in English | MEDLINE | ID: mdl-19187440

ABSTRACT

Microglial activation by blood-borne factors following blood-brain barrier damage may play a significant role in subsequent neuropathogenesis of several neurodegenerative diseases. Exposure of primary cultured rat brain microglia to pure, fatty acid- and lipid-deficient rat serum albumin or fraction V, (fatty acid and lipid-containing rat serum albumin), caused inducible nitric oxide synthase (iNOS) expression, glutamate release, tumour necrosis factor alpha (TNFalpha) and transforming growth factor-beta1 release. iNOS expression was attenuated by the MAPK/extracellular signal-regulated kinase pathway inhibitor U0126 and the phosphorylated forms of extracellular signal-regulated kinase 1 and 2 were detectable in microglia treated with albumin or fraction V. Glutamate release was prevented by l-alpha-aminoadipate and glutathione levels in microglia rose on exposure to albumin. Conditioned medium from microglia exposed to albumin or fraction V was neurotoxic. Peripheral macrophages were resistant to the effects of albumin but both microglia and macrophages responded to lipopolysaccharide, which induced interleukin-1 beta and tumour necrosis factor alpha release, cyclooxygenase-2 and iNOS expression in both cell types, indicating a discrete desensitised pathway in macrophages for albumin which was not desensitised in microglia. Thus, exposure of microglia in the brain to albumin may contribute to neuronal damage following blood-brain barrier breakdown and point to resident microglia rather than infiltrating macrophages as therapeutic targets.


Subject(s)
Cell Differentiation/drug effects , Macrophages, Peritoneal/drug effects , Microglia/drug effects , Serum Albumin/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Animals, Newborn , Caspases/metabolism , Cell Death/drug effects , Cells, Cultured , Cerebellum/cytology , Culture Media, Conditioned/pharmacology , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Glutamic Acid/metabolism , Glutathione/metabolism , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/chemistry , Microglia/chemistry , Neurons/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Polymyxin B/pharmacology , Rats , Rats, Wistar , Time Factors
19.
J Antimicrob Chemother ; 63(2): 317-24, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19036753

ABSTRACT

OBJECTIVES: The aim of this study was to characterize the in vitro antileishmanial activity of quassin, a traditional Chinese herbal medicine. METHODS: The cytotoxic effect of quassin was studied in murine peritoneal macrophages at various concentrations using the 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide method. The role of quassin as an antileishmanial agent was evaluated by microscopic counting of intracellular amastigotes in macrophages stained with Giemsa. To understand the effector mechanism of quassin-treated macrophages against leishmanial parasites, western blot and real-time PCR analysis of inducible nitric oxide (NO) synthase 2 (iNOS2) were done followed by measurement of NO generation by Griess reaction. The effect of quassin on the production of Th1 cytokines such as interleukin (IL)-12 and tumour necrosis factor (TNF)-alpha and Th2 cytokines such as IL-10 and transforming growth factor-beta was measured by ELISA, and the mRNA expression of these cytokines was analysed by real-time PCR. RESULTS: Quassin at a dose of 25 microg/mL (64.36 microM) showed less cytotoxicity to the host murine peritoneal macrophages but at the same dose was effective enough to control the intracellular parasitic load compared with higher doses of quassin. Leishmania donovani is known to exert its pathogenic effects mainly by the suppression of NO generation and subversion of the cellular inflammatory responses in the macrophages. Quassin was found to induce a potent host-protective immune response by enhancing NO generation and iNOS2 expression both at a protein and mRNA level and by up-regulating pro-inflammatory cytokines such as TNF-alpha and IL-12 in L. donovani-infected macrophages with concurrent inhibition of anti-inflammatory responses. CONCLUSIONS: These findings strongly support the effectiveness of quassin as a potent immunomodulatory tool for controlling the establishment of leishmanial parasite within the host macrophages.


Subject(s)
Antiprotozoal Agents/pharmacology , Immunologic Factors/pharmacology , Leishmania donovani/immunology , Macrophages, Peritoneal/immunology , Nitric Oxide/biosynthesis , Quassins/pharmacology , Animals , Antiprotozoal Agents/toxicity , Cell Survival , Cytokines/biosynthesis , Female , Immunologic Factors/toxicity , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/parasitology , Male , Mice , Mice, Inbred BALB C , Nitric Oxide/immunology , Nitric Oxide Synthase Type II/metabolism , Quassins/toxicity
20.
Biol Pharm Bull ; 32(9): 1628-31, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19721246

ABSTRACT

Targeted gene delivery to macrophages is important for the treatments of various immune diseases. Since macrophages express mannose receptors, development of efficient mannosylated non-viral carriers is an effective approach to macrophages-selective in vivo gene transfection. In this study, a pH-sensitive mannosylated cholesterol derivative, Man-His-C4-Chol, which possesses histidine (His) residues, containing lipoplexes (Man-His-lipoplexes) was characterized for transfection both in vitro and in vivo. In primary cultured macrophages, both Man-His-lipoplexes and mannosylated (Man)-lipoplexes showed significantly higher cellular uptake than bare-lipoplexes and there was no significant difference between Man-His-lipoplexes and Man-lipoplexes at 37 degrees C but the cellular uptake of these three lipoplexes was reduced at 4 degrees C. Similarly, the transfection efficacy of Man-His-lipoplexes showed significantly higher gene expression than bare-lipoplexes and Man-lipoplexes. After intraperitoneal administration to mice, Man-His-lipoplexes showed higher gene expression in peritoneally exuded cells (PECs) which contained macrophages than Man-lipoplexes and bare-lipoplexes at 3, 6, and 24 h. In addition, Man-His-lipoplexes showed higher gene expression than Gal-His-lipoplexes in PECs, suggesting that Man-His-lipoplexes are taken up by macrophages via mannose receptor-mediated endocytosis. These results suggest that Man-His-lipoplexes have superior transfection activity to Man-lipoplexes in macrophages.


Subject(s)
Histidine/genetics , Macrophages, Peritoneal/physiology , Mannose/genetics , Transfection/methods , Animals , Cations , Cells, Cultured , Gene Transfer Techniques/trends , Histidine/administration & dosage , Histidine/chemistry , Liposomes , Macrophages, Peritoneal/chemistry , Macrophages, Peritoneal/metabolism , Male , Mannose/administration & dosage , Mannose/chemistry , Mice , Mice, Inbred ICR
SELECTION OF CITATIONS
SEARCH DETAIL